Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Mol Ther Methods Clin Dev ; 12: 175-183, 2019 Mar 15.
Article in English | MEDLINE | ID: mdl-30705922

ABSTRACT

Elements within the γ-hemoglobin promoters (HBG1 and HBG2) function to bind transcription complexes that mediate repression of fetal hemoglobin expression. Sickle cell disease (SCD) subjects with a 13-bp deletion in the HBG1 promoter exhibit a clinically favorable hereditary persistence of fetal hemoglobin (HPFH) phenotype. We developed TALENs targeting the homologous HBG promoters to de-repress fetal hemoglobin. Transfection of human CD34+ cells with TALEN mRNA resulted in indel generation in HBG1 (43%) and HBG2 (74%) including the 13-bp HPFH deletion (∼6%). Erythroid differentiation of edited cells revealed a 4.6-fold increase in γ-hemoglobin expression as detected by HPLC. Assessment of TALEN-edited CD34+ cells in vivo in a humanized mouse model demonstrated sustained presence of indels in hematopoietic cells up to 24 weeks. Indel rates remained unchanged following secondary transplantation consistent with editing of long-term repopulating stem cells (LT-HSCs). Human γ-hemoglobin expressing F cells were detected by flow cytometry approximately 50% more frequently in edited animals compared to mock. Together, these findings demonstrate that TALEN-mediated indel generation in the γ-hemoglobin promoter leads to high levels of fetal hemoglobin expression in vitro and in vivo, suggesting that this approach can provide therapeutic benefit in patients with SCD or ß-thalassemia.

4.
Blood ; 109(10): 4168-70, 2007 May 15.
Article in English | MEDLINE | ID: mdl-17227830

ABSTRACT

Gemtuzumab ozogamicin (GO) contains an anti-CD33 antibody to facilitate uptake of a toxic calicheamicin-gamma(1) derivative. While recent in vitro data demonstrated a quantitative relationship between CD33 expression and GO cytotoxicity, previous correlative studies failed to identify a significant association between CD33 expression and clinical outcome. Studying patients undergoing GO monotherapy for relapsed acute myeloid leukemia (AML), we now find that AML blasts of responders have a significantly higher mean CD33 level and lower P-glycoprotein (Pgp) activity compared with nonresponders. CD33 expression and Pgp activity are inversely correlated. While both variables are associated with outcome, Pgp remains significantly associated with outcome even after adjusting for CD33, whereas CD33 does not show such an association after adjusting for Pgp. The inverse relationship between CD33 and Pgp suggests a maturation-stage-dependent expression of both proteins, and offers the rationale for using cell differentiation-promoting agents to enhance GO-induced cytotoxicity.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Aminoglycosides/pharmacokinetics , Aminoglycosides/therapeutic use , Antibodies, Monoclonal/pharmacokinetics , Antibodies, Monoclonal/therapeutic use , Antigens, CD/metabolism , Antigens, Differentiation, Myelomonocytic/metabolism , Leukemia, Myeloid/drug therapy , Leukemia, Myeloid/metabolism , Acute Disease , Adult , Aged , Aged, 80 and over , Antibodies, Monoclonal, Humanized , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/therapeutic use , Clinical Trials, Phase II as Topic , Gemtuzumab , Humans , Middle Aged , Multicenter Studies as Topic , Remission Induction , Sialic Acid Binding Ig-like Lectin 3 , Treatment Outcome
5.
Blood ; 102(4): 1466-73, 2003 Aug 15.
Article in English | MEDLINE | ID: mdl-12689934

ABSTRACT

Gemtuzumab ozogamicin (GO) is a novel immunoconjugate therapy for acute myeloid leukemia (AML). P-glycoprotein (Pgp) confers resistance to GO and is associated with a worse clinical response. To address whether multidrug resistance protein (MRP) affects GO susceptibility, we characterized Pgp, MRP1, and MRP2 expression in CD33+ cell lines and CD33+ AML samples and analyzed the effect of the Pgp inhibitor cyclosporine (CSA) and the MRP inhibitor MK-571 on GO-induced cytotoxicity. MRP1, but not MRP2, expression correlated with MRP activity. MK-571 enhanced GO-induced cytotoxicity in Pgp-negative/MRP-positive NB4 and HL-60 cells. CSA, but not MK-571 alone, restored GO susceptibility in Pgp-positive/MRP-positive TF1 cells; however, MK-571 enhanced cytotoxicity in the presence of CSA. All patient samples exhibited MRP activity, and 17 of 23 exhibited Pgp activity. CSA increased GO-induced cytotoxicity in 12 Pgp-positive samples, whereas MK-571 alone was effective in only one sample with minimal Pgp activity. In 3 Pgp-positive/MRP-positive samples, MK-571 enhanced GO-induced cytotoxicity in the presence of CSA. Thus, MRP1 may attenuate susceptibility to GO. This effect was comparatively less than that for Pgp and required the inhibition of Pgp for detection in cells that coexpressed both transporters. Because MK-571 and CSA failed to affect cytotoxicity in a portion of Pgp-positive/MRP-positive AML samples, additional resistance mechanisms are likely important.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B/metabolism , Aminoglycosides , Anti-Bacterial Agents/pharmacology , Antibodies, Monoclonal/pharmacology , Immunotoxins/pharmacology , Leukemia, Promyelocytic, Acute/drug therapy , Leukemia, Promyelocytic, Acute/metabolism , ATP Binding Cassette Transporter, Subfamily B/antagonists & inhibitors , ATP Binding Cassette Transporter, Subfamily B/biosynthesis , ATP Binding Cassette Transporter, Subfamily B/genetics , Antibodies, Monoclonal, Humanized , Antigens, CD/immunology , Antigens, Differentiation, Myelomonocytic/immunology , Cell Survival/drug effects , Cyclosporins/pharmacology , Drug Resistance, Multiple , Drug Resistance, Neoplasm , Gemtuzumab , HL-60 Cells , Humans , Propionates/pharmacology , Quinolines/pharmacology , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Sialic Acid Binding Ig-like Lectin 3 , Tumor Cells, Cultured
6.
Bioconjug Chem ; 13(1): 40-6, 2002.
Article in English | MEDLINE | ID: mdl-11792177

ABSTRACT

The anti-CD33 antibody, P67.6, has been chosen to target the potently cytotoxic calicheamicin antitumor antibiotics to acute myeloid leukemia (AML) due to the presence of CD33 on >80% of patient samples and its lack of expression outside the myeloid cell lineages, especially its lack of expression on pluripotent stem cells. Previous calicheamicin conjugates relied on the attachment of a hydrazide derivative to the oxidized carbohydrates that occur naturally on antibodies. This results in a "carbohydrate conjugate" capable of releasing active drug by hydrolysis of a hydrazone bond in the lysozomes where the pH is low. Conjugates have now been made that are formed by reacting a calicheamicin derivative containing an activated ester with the lysines of antibodies. This results in an "amide conjugate" that is stable to hydrolysis, leaving the disulfide that is present in all calicheamicin conjugates as the likely site of drug release from the conjugate. In this article, these two classes of calicheamicin-antibody conjugates are compared for potential use in AML with the anti-CD33 antibody P67.6. Conjugates of P67.6 are shown to require the site of hydrolytic release afforded by the carbohydrate conjugates in order to retain good potency and selectivity in vitro, in vivo, and ex vivo. The P67.6 carbohydrate conjugate of calicheamicin is selectively cytotoxic at <0.006 ng/mL of calicheamicin equivalents (cal equiv) toward HL-60 promyelocytic leukemia cells in tissue culture. Long-term, tumor-free survivors are seen in xenograft models when mice bearing HL-60 subcutaneous tumors are treated with the P67.6 carbohydrate conjugate at a dose of 300 microg/kg cal equiv given three times. This conjugate also selectively inhibits the formation of colonies from AML marrow samples at 2 ng/mL cal equiv. The P67.6 carbohydrate conjugate of calicheamicin therefore appears to have promise as an antibody-targeted chemotherapeutic agent for CD33-positive diseases such as AML.


Subject(s)
Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/therapeutic use , Antibiotics, Antineoplastic/chemistry , Antibiotics, Antineoplastic/therapeutic use , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/therapeutic use , Antigens, CD/immunology , Antigens, Differentiation, Myelomonocytic/immunology , Immunotoxins/chemistry , Immunotoxins/therapeutic use , Leukemia, Myeloid/drug therapy , Acute Disease , Aminoglycosides , Animals , HL-60 Cells , Humans , Immunochemistry , Magnetic Resonance Spectroscopy , Mice , Mice, Nude , Sialic Acid Binding Ig-like Lectin 3 , Spectrophotometry, Infrared , Spectrophotometry, Ultraviolet
7.
Bioconjug Chem ; 13(1): 47-58, 2002.
Article in English | MEDLINE | ID: mdl-11792178

ABSTRACT

CD33 is expressed by acute myeloid leukemia (AML) cells in >80% of patients but not by normal hematopoietic stem cells, suggesting that elimination of CD33(+) cells may be therapeutically beneficial. A conjugate of a calicheamicin hydrazide derivative attached via hydrazone formation to the oxidized carbohydrates of the anti-CD33 murine antibody P67.6 had been chosen for use in AML prior to humanization of this antibody. However, the CDR-grafted humanized P67.6 could not be used to make the carbohydrate conjugate because of the unexpected sensitivity of this antibody to periodate oxidation. Exploration of a series of bifunctional linkers resulted in a new class of calicheamicin conjugates, termed the hybrid conjugates, that allows for the attachment of the calicheamicin to lysines but incorporates the site of hydrolytic release, a hydrazone, previously shown to be required for activity. The optimized conjugate chosen for clinical trials, gemtuzumab ozogamicin ("gem-ozo", Mylotarg, formerly designated CMA-676), was significantly more potent and selective than the carbohydrate conjugate it replaced. It was selectively cytotoxic to HL-60 leukemia cells in tissue culture with an IC(50) in the low to sub-pg cal/mL range (cal = calicheamicin equivalents). Doses of gem-ozo as low as 50 microg cal/kg given three times to mice bearing HL-60 xenografts routinely resulted in long-term, tumor-free survivors, while a nonbinding control conjugate was relatively inactive. Gem-ozo at a concentration of 2 to 10 ng cal/mL selectively inhibited leukemia colony formation by marrow cells from a significant proportion of AML patients. Gem-ozo has also shown significant activity against AML in Phase II trials and is the first antibody-targeted chemotherapeutic agent approved by the FDA.


Subject(s)
Aminoglycosides , Anti-Bacterial Agents/therapeutic use , Antibodies, Monoclonal/therapeutic use , Antigens, CD/immunology , Antigens, Differentiation, Myelomonocytic/immunology , Immunotoxins/therapeutic use , Leukemia, Myeloid/drug therapy , Acute Disease , Animals , Antibodies, Monoclonal, Humanized , Cross-Linking Reagents , Gemtuzumab , HL-60 Cells , Humans , Indicators and Reagents , Mice , Mice, Nude , Neoplasm Transplantation , Sialic Acid Binding Ig-like Lectin 3 , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...