Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 40
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Hum Mol Genet ; 29(17): 2920-2935, 2020 10 10.
Article in English | MEDLINE | ID: mdl-32803224

ABSTRACT

Neuroinflammation is a key contributor to the pathology of Alzheimer's disease (AD). CD33 (Siglec-3) is a transmembrane sialic acid-binding receptor on the surface of microglial cells. CD33 is upregulated on microglial cells from post-mortem AD patient brains, and high levels of CD33 inhibit uptake and clearance of amyloid beta (Aß) in microglial cell cultures. Furthermore, knockout of CD33 reduces amyloid plaque burden in mouse models of AD. Here, we tested whether a gene therapy strategy to reduce CD33 on microglia in AD could decrease Aß plaque load. Intracerebroventricular injection of an adeno-associated virus (AAV) vector-based system encoding an artificial microRNA targeting CD33 (miRCD33) into APP/PS1 mice reduced CD33 mRNA and TBS-soluble Aß40 and Aß42 levels in brain extracts. Treatment of APP/PS1 mice with miRCD33 vector at an early age (2 months) was more effective at reducing Aß plaque burden than intervening at later times (8 months). Furthermore, early intervention downregulated several microglial receptor transcripts (e.g. CD11c, CD47 and CD36) and pro-inflammatory activation genes (e.g. Tlr4 and Il1b). Marked reductions in the chemokine Ccl2 and the pro-inflammatory cytokine Tnfα were observed at the protein level in the brain of APP/PS1 mice treated with miRCD33 vector. Overall, our data indicate that CD33 is a viable target for AAV-based knockdown strategies to reduce AD pathology. One Sentence Summary: A gene therapy approach for Alzheimer's disease using adeno-associated virus vector-based knockdown of CD33 reduced amyloid beta accumulation and neuroinflammation.


Subject(s)
Alzheimer Disease/therapy , Genetic Therapy , Presenilin-1/genetics , Sialic Acid Binding Ig-like Lectin 3/genetics , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Amyloid beta-Peptides/genetics , Amyloid beta-Protein Precursor/genetics , Animals , Brain/metabolism , Brain/pathology , Dependovirus/genetics , Disease Models, Animal , Humans , Mice , Mice, Transgenic/genetics , Microglia/metabolism , Microglia/pathology , Plaque, Amyloid/genetics , Plaque, Amyloid/pathology , Sialic Acid Binding Ig-like Lectin 3/antagonists & inhibitors
2.
Blood ; 135(2): 85-96, 2020 01 09.
Article in English | MEDLINE | ID: mdl-31765470

ABSTRACT

The acute myeloid leukemia (AML) treatment landscape has changed substantially since 2017. New targeted drugs have emerged, including venetoclax to target B-cell lymphoma 2, midostaurin and gilteritinib to target FLT3, and ivosidenib and enasidenib to target mutant isocitrate dehydrogenase 1 and 2, respectively. Other additions include reapproval of gemtuzumab ozogomycin to target CD33, glasdegib to target the hedgehog pathway, and a liposomal formulation of daunorubicin and cytarabine (CPX-351). Genomically heterogeneous AML has a tendency to evolve, particularly under selective treatment pressure. For decades, treatment decisions have largely centered around chemotherapy drug intensity. Physicians now have access to an increasing number of drugs with novel mechanisms of action and distinctive side-effect profiles. Key issues faced by hematologists in this era of new drugs include (1) the timely identification of actionable mutations at diagnosis and at relapse; (2) deciding which drug to use among several therapeutic options; and (3) increasing awareness of how to anticipate, mitigate, and manage common complications associated with these new agents. This article will use 3 case presentations to discuss some of the new treatment challenges encountered in AML management, with the goal of providing practical guidance to aid the practicing physician.


Subject(s)
Antineoplastic Agents/therapeutic use , Biomarkers, Tumor/genetics , Leukemia, Myeloid, Acute/drug therapy , Molecular Targeted Therapy , Mutation , Adult , Aged , Aminopyridines/therapeutic use , Aniline Compounds/therapeutic use , Bridged Bicyclo Compounds, Heterocyclic/therapeutic use , Cytarabine/therapeutic use , Daunorubicin/therapeutic use , Female , Glycine/analogs & derivatives , Glycine/therapeutic use , Humans , Isocitrate Dehydrogenase/antagonists & inhibitors , Isocitrate Dehydrogenase/genetics , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/pathology , Male , Prognosis , Pyrazines/therapeutic use , Pyridines/therapeutic use , Sialic Acid Binding Ig-like Lectin 3/antagonists & inhibitors , Sialic Acid Binding Ig-like Lectin 3/genetics , Staurosporine/analogs & derivatives , Staurosporine/therapeutic use , Sulfonamides/therapeutic use , Triazines/therapeutic use , fms-Like Tyrosine Kinase 3/antagonists & inhibitors , fms-Like Tyrosine Kinase 3/genetics
3.
Br J Haematol ; 186(4): 538-548, 2019 08.
Article in English | MEDLINE | ID: mdl-31115909

ABSTRACT

The differential immunophenotypic characteristics of early T precursor (ETP) acute lymphoblastic leukaemia/lymphoma (ALL) remain incompletely characterized. The study group (n = 142) included 106 (74·7%) men and 36 (25·3%) women with a median age of 34·9 years (range, 2-79) at diagnosis. Patients were subtyped by flow cytometry immunophenotyping as follows: 33 (23·2%) ETP; 32 (22·5%) early non-ETP; 60 (42·2%) thymic; and 17 (12·1%) mature. Excepting definitional markers, there was a significant differential expression of the markers CD2, CD10, CD33 and TdT between ETP-ALL and non-ETP-ALL. Positive CD33 expression (≥20% of leukaemic blasts) was detected in 21/33 (63%) ETP-ALL compared with 17/95 (17·9%) non-ETP-ALL (P < 0·001). Notably, targeted anti-CD33 therapy with IMGN779 resulted in significant growth inhibition and increased apoptosis in ETP-ALL cells in vitro. An 11-marker T-ALL immunophenotype score discriminated reliably between ETP and non-ETP ALL. Longitudinal analysis of ETP-ALL cases in this study demonstrated that the immunophenotype may be occasionally dynamic but is largely stable over the disease course. In summary, identification of ETP-ALL might be enhanced by using an 11-marker T-ALL immunophenotype score. CD33 expression is frequent in ETP-ALL, and in vitro data suggest that exploring anti-CD33 therapy in ETP-ALL is warranted.


Subject(s)
Immunophenotyping , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Sialic Acid Binding Ig-like Lectin 3/metabolism , Adult , Aged , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Apoptosis , Biomarkers , Cell Line , Female , Flow Cytometry , Gene Expression , Humans , Male , Middle Aged , Molecular Targeted Therapy , Neoplasm, Residual/diagnosis , Neoplasm, Residual/metabolism , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/diagnosis , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/etiology , Prognosis , Proportional Hazards Models , Sialic Acid Binding Ig-like Lectin 3/antagonists & inhibitors , Sialic Acid Binding Ig-like Lectin 3/genetics , Young Adult
4.
Oncologist ; 24(5): e171-e179, 2019 05.
Article in English | MEDLINE | ID: mdl-30898889

ABSTRACT

On February 22, 2018, the Committee for Medicinal Products for Human Use (CHMP) adopted a positive opinion, recommending the granting of a marketing authorization for the medicinal product gemtuzumab ozogamicin (Mylotarg; Pfizer, New York City, NY), intended for the treatment of acute myeloid leukemia. Mylotarg was designated as an orphan medicinal product on October 18, 2000. The applicant for this medicinal product was Pfizer Limited (marketing authorization now held by Pfizer Europe MA EEIG).The demonstrated benefit with Mylotarg is improvement in event-free survival. This has been shown in the pivotal ALFA-0701 (MF-3) study. In addition, an individual patient data meta-analysis from five randomized controlled trials (3,325 patients) showed that the addition of Mylotarg significantly reduced the risk of relapse (odds ratio [OR] 0.81; 95% CI: 0.73-0.90; p = .0001), and improved overall survival at 5 years (OR 0.90; 95% CI: 0.82-0.98; p = .01) [Lancet Oncol 2014;15:986-996]. The most common (>30%) side effects of Mylotarg when used together with daunorubicin and cytarabine are hemorrhage and infection.The full indication is as follows: "Mylotarg is indicated for combination therapy with daunorubicin (DNR) and cytarabine (AraC) for the treatment of patients age 15 years and above with previously untreated, de novo CD33-positive acute myeloid leukemia (AML), except acute promyelocytic leukemia (APL)."The objective of this article is to summarize the scientific review done by the CHMP of the application leading to regulatory approval in the European Union. The full scientific assessment report and product information, including the Summary of Product Characteristics, are available on the European Medicines Agency website (www.ema.europa.eu). IMPLICATIONS FOR PRACTICE: This article reflects the scientific assessment of Mylotarg (gemtuzumab ozogamicin; Pfizer, New York City, NY) use for the treatment of acute myeloid leukemia based on important contributions from the rapporteur and co-rapporteur assessment teams, Committee for Medicinal Products for Human Use members, and additional experts following the application for a marketing authorization from the company. It's a unique opportunity to look at the data from a regulatory point of view and the importance of assessing the benefit-risk.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Drug Approval , Gemtuzumab/administration & dosage , Leukemia, Myeloid, Acute/drug therapy , Age Factors , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Cytarabine/administration & dosage , Cytarabine/adverse effects , Daunorubicin/administration & dosage , Daunorubicin/adverse effects , Disease-Free Survival , Dose-Response Relationship, Drug , Drug Administration Schedule , Europe , Gemtuzumab/adverse effects , Humans , Kaplan-Meier Estimate , Leukemia, Myeloid, Acute/mortality , Leukemia, Myeloid, Acute/pathology , Meta-Analysis as Topic , Randomized Controlled Trials as Topic , Risk Assessment , Sialic Acid Binding Ig-like Lectin 3/antagonists & inhibitors , Sialic Acid Binding Ig-like Lectin 3/metabolism
5.
Blood ; 130(23): 2469-2474, 2017 12 07.
Article in English | MEDLINE | ID: mdl-29051180

ABSTRACT

In 2017, 4 drugs received US Food and Drug Administration marketing approval for acute myeloid leukemia (AML) treatment: targeted therapies for mutant FLT3 and IDH2, a liposomal cytarabine-daunorubicin formulation for therapy-related AML and AML with myelodysplasia-related changes, and resurgence of an antibody-drug conjugate designed to target CD33. Promising results also emerged for the BCL-2 inhibitor venetoclax combined with low-intensity therapy in older patients unfit for intensive chemotherapy. This quintet of new drugs is likely to reshape the therapeutic landscape of AML.


Subject(s)
Aminoglycosides/therapeutic use , Aminopyridines/therapeutic use , Antibodies, Monoclonal, Humanized/therapeutic use , Bridged Bicyclo Compounds, Heterocyclic/therapeutic use , Cytarabine/administration & dosage , Daunorubicin/administration & dosage , Leukemia, Myeloid, Acute/drug therapy , Molecular Targeted Therapy , Staurosporine/analogs & derivatives , Sulfonamides/therapeutic use , Triazines/therapeutic use , Aminoglycosides/pharmacology , Aminopyridines/pharmacology , Antibodies, Monoclonal, Humanized/pharmacology , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Clinical Trials as Topic , Gemtuzumab , Humans , Isocitrate Dehydrogenase/antagonists & inhibitors , Isocitrate Dehydrogenase/genetics , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/mortality , Liposomes , Mutation , Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors , Sialic Acid Binding Ig-like Lectin 3/antagonists & inhibitors , Staurosporine/pharmacology , Staurosporine/therapeutic use , Sulfonamides/pharmacology , Treatment Outcome , Triazines/pharmacology , fms-Like Tyrosine Kinase 3/antagonists & inhibitors , fms-Like Tyrosine Kinase 3/genetics
6.
Bioorg Med Chem Lett ; 29(18): 2609-2612, 2019 09 15.
Article in English | MEDLINE | ID: mdl-31387789

ABSTRACT

To identify sialic acid binding proteins from complex proteomes, three photocrosslinking affinity-based probes were constructed using Neu5Ac (5 and 6) and Neu5Ac2en (7) scaffolds. Kinetic inhibition assays and Western blotting revealed the Neu5Ac2en-based 7 to be an effective probe for the labeling of a purified gut microbial sialidase (BDI_2946) and a purified human sialic acid binding protein (hCD33). Additionally, LC-MS/MS affinity-based protein profiling verified the ability of 7 to enrich a low-abundance sialic acid binding protein (complement factor H) from human serum thus validating the utility of this probe in a complex context.


Subject(s)
Enzyme Inhibitors/pharmacology , Molecular Probes/pharmacology , N-Acetylneuraminic Acid/pharmacology , Sialic Acid Binding Ig-like Lectin 3/antagonists & inhibitors , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Humans , Molecular Probes/chemical synthesis , Molecular Probes/chemistry , Molecular Structure , N-Acetylneuraminic Acid/chemical synthesis , N-Acetylneuraminic Acid/chemistry , Neuraminidase/antagonists & inhibitors , Neuraminidase/metabolism , Sialic Acid Binding Ig-like Lectin 3/isolation & purification , Sialic Acid Binding Ig-like Lectin 3/metabolism , Structure-Activity Relationship
7.
Future Oncol ; 14(30): 3199-3213, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30039981

ABSTRACT

In September 2017, the US FDA announced re-approval of gemtuzumab ozogamicin (GO), a CD33-targeting immunoconjugate, for treatment of newly diagnosed and relapsed/refractory acute myeloid leukemia (AML). This is a very significant step toward defining new treatment regimens in AML, as the treatment has essentially stayed unchanged with the '7 + 3 induction regimen' (7 days cytarabine and 3 days of anthracycline) since 1973. GO is the first antibody-drug conjugate to receive FDA approval for treating cancer. This review article discusses the challenges faced and lessons learned during the journey of GO for AML treatment. Selected trials that have made significant contribution in our understanding of the most efficacious and safe use of GO for treating AML patients as well as factors influencing GO response are highlighted in this article.


Subject(s)
Aminoglycosides/therapeutic use , Antibodies, Monoclonal, Humanized/therapeutic use , Antineoplastic Agents, Immunological/therapeutic use , Biomarkers, Tumor , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/metabolism , Sialic Acid Binding Ig-like Lectin 3/antagonists & inhibitors , Sialic Acid Binding Ig-like Lectin 3/metabolism , Age Factors , Aminoglycosides/administration & dosage , Aminoglycosides/adverse effects , Antibodies, Monoclonal, Humanized/administration & dosage , Antibodies, Monoclonal, Humanized/adverse effects , Antineoplastic Agents, Immunological/administration & dosage , Antineoplastic Agents, Immunological/adverse effects , Drug Discovery , France , Gemtuzumab , Humans , Leukemia, Myeloid, Acute/diagnosis , Leukemia, Myeloid, Acute/genetics , Molecular Targeted Therapy , Randomized Controlled Trials as Topic , Sialic Acid Binding Ig-like Lectin 3/genetics , Treatment Outcome , United States
8.
Hum Mol Genet ; 24(12): 3557-70, 2015 Jun 15.
Article in English | MEDLINE | ID: mdl-25762156

ABSTRACT

The CD33 single-nucleotide polymorphism (SNP) rs3865444 has been associated with the risk of Alzheimer's disease (AD). Rs3865444 is in linkage disequilibrium with rs12459419 which has been associated with efficacy of an acute myeloid leukemia (AML) chemotherapeutic agent based on a CD33 antibody. We seek to evaluate the extent to which CD33 genetics in AD and AML can inform one another and advance human disease therapy. We have previously shown that these SNPs are associated with skipping of CD33 exon 2 in brain mRNA. Here, we report that these CD33 SNPs are associated with exon 2 skipping in leukocytes from AML patients and with a novel CD33 splice variant that retains CD33 intron 1. Each copy of the minor rs12459419T allele decreases prototypic full-length CD33 expression by ∼ 25% and decreases the AD odds ratio by ∼ 0.10. These results suggest that CD33 antagonists may be useful in reducing AD risk. CD33 inhibitors may include humanized CD33 antibodies such as lintuzumab which was safe but ineffective in AML clinical trials. Here, we report that lintuzumab downregulates cell-surface CD33 by 80% in phorbol-ester differentiated U937 cells, at concentrations as low as 10 ng/ml. Overall, we propose a model wherein a modest effect on RNA splicing is sufficient to mediate the CD33 association with AD risk and suggest the potential for an anti-CD33 antibody as an AD-relevant pharmacologic agent.


Subject(s)
Alzheimer Disease/genetics , Genetic Association Studies , Leukemia, Myeloid, Acute/genetics , Sialic Acid Binding Ig-like Lectin 3/genetics , Aged , Aged, 80 and over , Alleles , Alternative Splicing , Alzheimer Disease/metabolism , Antibodies, Monoclonal, Humanized/pharmacology , Cell Line , Exons , Female , Gene Expression , Genetic Predisposition to Disease , Genotype , Humans , Introns , Leukemia, Myeloid, Acute/metabolism , Male , Polymorphism, Single Nucleotide , RNA Stability , RNA, Messenger/genetics , Sialic Acid Binding Ig-like Lectin 3/antagonists & inhibitors , Sialic Acid Binding Ig-like Lectin 3/metabolism
9.
Cancer ; 121(16): 2689-704, 2015 Aug 15.
Article in English | MEDLINE | ID: mdl-26095886

ABSTRACT

Despite the remarkable progress made in some leukemias such as CML and CLL, cytotoxic treatment for AML remains essentially unchanged over the last 4 decades. Several lines of evidence, including the graft versus leukemia effect associated with allogeneic hematopoietic stem cell transplantation (HSCT), suggest that immunotherapy is an active modality in AML. Given the lack of progress for chemotherapy in this disease, many novel immunologic treatment approaches have been explored. The goals of non-transplant-based immune approaches have largely consisted of the stimulation or restoration of endogenous immune responses or the targeting of specific tumor antigens by immune cells. These strategies have been associated with less toxicity than allogeneic HSCT but typically have inferior efficacy. Allogeneic HSCT exploits major and minor histocompatibility differences between the donor and recipient in order to recognize and eradicate malignancy. With the recognition that the immune system itself provides a basis for treating AML, immunotherapy continues to be an attractive modality to exploit in the treatment of this disease.


Subject(s)
Immunotherapy , Leukemia, Myeloid, Acute/therapy , Antibodies, Bispecific/therapeutic use , Antibodies, Monoclonal/therapeutic use , Antigens, CD , Cancer Vaccines/therapeutic use , Cell Adhesion Molecules/antagonists & inhibitors , Cytokines/therapeutic use , Hematopoietic Stem Cell Transplantation , Humans , Killer Cells, Natural/immunology , Radioimmunotherapy , Sialic Acid Binding Ig-like Lectin 3/antagonists & inhibitors
10.
Haematologica ; 100(3): 336-44, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25480499

ABSTRACT

Current therapies for acute myeloid leukemia are associated with high failure and relapse rates. Adoptive immunotherapies, which have shown promise in the treatment of hematologic malignancies, have the potential to target acute myeloid leukemia through pathways that are distinct and complementary to current approaches. Here, we describe the development of a novel adoptive immunotherapy specific for this disease. We generated a second generation CD33-specific chimeric antigen receptor capable of redirecting cytolytic effector T cells against leukemic cells. CD33 is expressed in approximately 90% of acute myeloid leukemia cases and has demonstrated utility as a target of therapeutic antibodies. Chimeric antigen receptor-modified T cells efficiently killed leukemia cell lines and primary tumor cells in vitro. The anti-leukemia effect was CD33-specific, mediated through T-cell effector functions, and displayed tumor lysis at effector:target ratios as low as 1:20. Furthermore, the CD33-redirected T cells were effective in vivo, preventing the development of leukemia after prophylactic administration and delaying the progression of established disease in mice. These data provide pre-clinical validation of the effectiveness of a second-generation anti-CD33 chimeric antigen receptor therapy for acute myeloid leukemia, and support its continued development as a clinical therapeutic.


Subject(s)
Immunotherapy, Adoptive/methods , Leukemia, Myeloid, Acute/therapy , Mutant Chimeric Proteins/immunology , Receptors, Antigen, T-Cell/immunology , Sialic Acid Binding Ig-like Lectin 3/antagonists & inhibitors , T-Lymphocytes, Cytotoxic/immunology , Animals , Bone Marrow/immunology , Bone Marrow/pathology , Cell Line, Tumor , Coculture Techniques , Cytotoxicity, Immunologic , Gene Expression , Humans , Interleukin Receptor Common gamma Subunit/deficiency , Interleukin Receptor Common gamma Subunit/genetics , Interleukin Receptor Common gamma Subunit/immunology , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/immunology , Leukemia, Myeloid, Acute/pathology , Liver/immunology , Liver/pathology , Mice , Mice, Inbred NOD , Mice, SCID , Mutant Chimeric Proteins/genetics , Plasmids/chemistry , Plasmids/genetics , Receptors, Antigen, T-Cell/genetics , Sialic Acid Binding Ig-like Lectin 3/genetics , Sialic Acid Binding Ig-like Lectin 3/immunology , Spleen/immunology , Spleen/pathology , T-Lymphocytes, Cytotoxic/cytology , T-Lymphocytes, Cytotoxic/transplantation , Transfection
11.
Ann Hematol ; 94(3): 361-73, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25284166

ABSTRACT

Conventional chemotherapy is ineffective in the majority of patients with acute myeloid leukaemia (AML), and monoclonal antibodies recognising CD33 expressed on myeloid progenitors (e.g. gemtuzumab ozogamicin (GO)) have been reported to improve outcome in patients with AML. Reports of excess toxicity have resulted in GO's licence being withdrawn. As a result, the role of these agents remains unclear. A systematic review and meta-analysis included studies of patients with AML who had entered a randomised control trial (RCT), where one arm included anti-CD33 antibody therapy. Fixed effect meta-analysis was used, involving calculation of observed minus expected number of events, and variance for each endpoint in each trial, with the overall treatment effect expressed as Peto's odds ratio with 95 % confidence interval. Meta-analysis of 11 RCTs with 13 randomisations involving GO was undertaken. Although GO increased induction deaths (p = 0.02), it led to a reduction in resistant disease (p = 0.0009); hence, there was no improvement in complete remission. Whilst GO improved relapse-free survival (hazard ratio (HR) = 0.90, 95 % confidence interval (CI) = 0.84-0.98, p = 0.01), there was no overall benefit of GO in overall survival (OS) (HR = 0.96, 95 % CI = 0.90-1.02, p = 0.2). GO improved OS in patients with favourable cytogenetics, with no evidence of benefit in patients with intermediate or adverse cytogenetics (test for heterogeneity between subtotals p = 0.01). GO has a potent clinically detectable anti-leukaemic effect. Further trials to investigate its optimum delivery and identification of patient populations who may benefit are needed.


Subject(s)
Aminoglycosides/therapeutic use , Antibodies, Monoclonal, Humanized/therapeutic use , Antineoplastic Agents/therapeutic use , Leukemia, Myeloid, Acute/drug therapy , Gemtuzumab , Humans , Randomized Controlled Trials as Topic , Sialic Acid Binding Ig-like Lectin 3/antagonists & inhibitors , Sialic Acid Binding Ig-like Lectin 3/immunology
14.
Blood Adv ; 8(9): 2059-2073, 2024 May 14.
Article in English | MEDLINE | ID: mdl-38266153

ABSTRACT

ABSTRACT: Novel therapies are needed for effective treatment of acute myeloid leukemia (AML). Relapse is common and salvage treatment with cytotoxic chemotherapy is rarely curative. CD123 and CD33, 2 clinically validated targets in AML, are jointly expressed on blasts and leukemic stem cells in >95% of patients with AML. However, their expression is heterogenous between subclones and between patients, which may affect the efficacy of single-targeting agents in certain patient populations. We present here a dual-targeting CD33/CD123 NANOBODY T-cell engager (CD33/CD123-TCE) that was designed to decrease the risk of relapse from possible single antigen-negative clones and to increase coverage within and across patients. CD33/CD123-TCE killed AML tumor cells expressing 1 or both antigens in vitro. Compared with single-targeting control compounds, CD33/CD123-TCE conferred equal or better ex vivo killing of AML blasts in most primary AML samples tested, suggesting a broader effectiveness across patients. In a disseminated cell-line-derived xenograft mouse model of AML, CD33/CD123-TCE cleared cancer cells in long bones and in soft tissues. As cytokine release syndrome is a well-documented adverse effect of TCE, the compound was tested in a cytokine release assay and shown to induce less cytokines compared to a CD123 single-targeting control. In an exploratory single-dose nonhuman primate study, CD33/CD123-TCE revealed a favorable PK profile. Depletion of CD123 and CD33 expressing cells was observed, but there were neither signs of cytokine release syndrome nor clinical signs of toxicity. Taken together, the CD33/CD123 dual-targeting NANOBODY TCE exhibits potent and safe anti-AML activity and promises a broad patient coverage.


Subject(s)
Interleukin-3 Receptor alpha Subunit , Leukemia, Myeloid, Acute , Sialic Acid Binding Ig-like Lectin 3 , Single-Domain Antibodies , Humans , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/immunology , Sialic Acid Binding Ig-like Lectin 3/antagonists & inhibitors , Interleukin-3 Receptor alpha Subunit/antagonists & inhibitors , Interleukin-3 Receptor alpha Subunit/immunology , Animals , Mice , Single-Domain Antibodies/therapeutic use , Single-Domain Antibodies/pharmacology , Xenograft Model Antitumor Assays , T-Lymphocytes/immunology , T-Lymphocytes/drug effects , T-Lymphocytes/metabolism , Cell Line, Tumor , Female
15.
Mol Cancer Ther ; 23(8): 1073-1083, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-38561023

ABSTRACT

CD33 (Siglec-3) is a cell surface receptor expressed in approximately 90% of acute myeloid leukemia (AML) blasts, making it an attractive target for therapy of AML. Although previous CD33-targeting antibody-drug conjugates (ADC) like gemtuzumab ozogamicin (GO, Mylotarg) have shown efficacy in AML treatment, they have suffered from toxicity and narrow therapeutic window. This study aimed to develop a novelADCwith improved tolerability and a wider therapeutic window. GLK-33 consists of the anti-CD33 antibody lintuzumab and eight mavg-MMAU auristatin linkerpayloads per antibody. The experimental methods included testing in cell cultures, patient-derived samples, mouse xenograft models, and rat toxicology studies. GLK-33 exhibited remarkable efficacy in reducing cell viability within CD33-positive leukemia cell lines and primary AML samples. Notably, GLK-33 demonstrated antitumor activity at single dose as low as 300 mg/kg in mice, while maintaining tolerability at single dose of 20 to 30 mg/kg in rats. In contrast with both GO and lintuzumab vedotin, GLK-33 exhibited a wide therapeutic window and activity against multidrug-resistant cells. The development of GLK-33 addresses the limitations of previous ADCs, offering a wider therapeutic window, improved tolerability, and activity against drug-resistant leukemia cells. These findings encourage further exploration of GLK-33 in AML through clinical trials.


Subject(s)
Antibodies, Monoclonal, Humanized , Immunoconjugates , Leukemia, Myeloid, Acute , Oligopeptides , Sialic Acid Binding Ig-like Lectin 3 , Humans , Animals , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/pathology , Mice , Sialic Acid Binding Ig-like Lectin 3/antagonists & inhibitors , Sialic Acid Binding Ig-like Lectin 3/metabolism , Rats , Antibodies, Monoclonal, Humanized/pharmacology , Oligopeptides/pharmacology , Immunoconjugates/pharmacology , Immunoconjugates/therapeutic use , Aminobenzoates/pharmacology , Xenograft Model Antitumor Assays , Cell Line, Tumor , Female
16.
Mol Neurodegener ; 16(1): 19, 2021 03 25.
Article in English | MEDLINE | ID: mdl-33766097

ABSTRACT

BACKGROUND: CD33 is genetically linked to Alzheimer's disease (AD) susceptibility through differential expression of isoforms in microglia. The role of the human CD33 short isoform (hCD33m), preferentially encoded by an AD-protective CD33 allele (rs12459419T), is unknown. Here, we test whether hCD33m represents a loss-of-function or gain-of-function variant. METHODS: We have developed two models to test the role of hCD33m. The first is a new strain of transgenic mice expressing hCD33m in the microglial cell lineage. The second is U937 cells where the CD33 gene was disrupted by CRISPR/Cas9 and complemented with different variants of hCD33. Primary microglia and U937 cells were tested in phagocytosis assays and single cell RNA sequencing (scRNAseq) was carried out on the primary microglia. Furthermore, a new monoclonal antibody was developed to detect hCD33m more efficiently. RESULTS: In both primary microglia and U937 cells, we find that hCD33m enhances phagocytosis. This contrasts with the human CD33 long isoform (hCD33M) that represses phagocytosis, as previously demonstrated. As revealed by scRNAseq, hCD33m+ microglia are enriched in a cluster of cells defined by an upregulated expression and gene regulatory network of immediate early genes, which was further validated within microglia in situ. Using a new hCD33m-specific antibody enabled hCD33m expression to be examined, demonstrating a preference for an intracellular location. Moreover, this newly discovered gain-of-function role for hCD33m is dependent on its cytoplasmic signaling motifs, dominant over hCD33M, and not due to loss of glycan ligand binding. CONCLUSIONS: These results provide strong support that hCD33m represents a gain-of-function isoform and offers insight into what it may take to therapeutically capture the AD-protective CD33 allele.


Subject(s)
Amyloid beta-Peptides/metabolism , Microglia/physiology , Peptide Fragments/metabolism , Phagocytosis/genetics , Sialic Acid Binding Ig-like Lectin 3/genetics , Alleles , Animals , CRISPR-Cas Systems , Crosses, Genetic , Female , Gain of Function Mutation , Gene Editing , Gene Regulatory Networks , Genes, Immediate-Early , Humans , Male , Mice, Inbred C57BL , Mice, Transgenic , Polysaccharides/metabolism , Protein Isoforms/genetics , Protein Isoforms/physiology , RNA-Seq , Sialic Acid Binding Ig-like Lectin 3/antagonists & inhibitors , Sialic Acid Binding Ig-like Lectin 3/physiology , Single-Cell Analysis , U937 Cells
17.
Leukemia ; 35(9): 2496-2507, 2021 09.
Article in English | MEDLINE | ID: mdl-33589747

ABSTRACT

There is increasing interest in targeting CD33 in malignant and non-malignant disorders. In acute myeloid leukemia, longer survival with the CD33 antibody-drug conjugate gemtuzumab ozogamicin (GO) validates this strategy. Still, GO benefits only some patients, prompting efforts to develop more potent CD33-directed therapeutics. As one limitation, CD33 antibodies typically recognize the membrane-distal V-set domain. Using various artificial CD33 proteins, in which this domain was differentially positioned within the extracellular portion of the molecule, we tested whether targeting membrane-proximal epitopes enhances the effector functions of CD33 antibody-based therapeutics. Consistent with this idea, a CD33V-set/CD3 bispecific antibody (BsAb) and CD33V-set-directed chimeric antigen receptor (CAR)-modified T cells elicited substantially greater cytotoxicity against cells expressing a CD33 variant lacking the entire C2-set domain than cells expressing full-length CD33, whereas cytotoxic effects induced by GO were independent of the position of the V-set domain. We therefore raised murine and human antibodies against the C2-set domain of human CD33 and identified antibodies that bound CD33 regardless of the presence/absence of the V-set domain ("CD33PAN antibodies"). These antibodies internalized when bound to CD33 and, as CD33PAN/CD3 BsAb, had potent cytolytic effects against CD33+ cells. Together, our data provide the rationale for further development of CD33PAN antibody-based therapeutics.


Subject(s)
Antibodies, Monoclonal, Humanized/chemistry , Gemtuzumab/chemistry , Immunoconjugates/pharmacology , Immunotherapy/methods , Leukemia, Myeloid, Acute/therapy , Sialic Acid Binding Ig-like Lectin 3/antagonists & inhibitors , Antibodies, Monoclonal, Humanized/biosynthesis , Antineoplastic Agents, Immunological/chemistry , Humans , Immunoconjugates/chemistry , Leukemia, Myeloid, Acute/immunology , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/pathology , Sialic Acid Binding Ig-like Lectin 3/immunology , Tumor Cells, Cultured
18.
Front Immunol ; 12: 683595, 2021.
Article in English | MEDLINE | ID: mdl-34484181

ABSTRACT

Background: Previous trials demonstrated evidence involving the total effects of gemtuzumab ozogamicin (GO), an anti-CD33 humanized antibody, on treating acute myeloid leukemia (AML). In this updated systematic review, meta-analysis, and network meta-analysis (NMA), we aimed to comprehensively explore the clinical benefits and safety of GO in various subtypes of AML. Methods: PubMed, Embase, Cochrane, and Chinese databases were filtered to search randomized controlled trials (RCTs) and retrospective cohort studies that compared clinical efficiency and toxicity of GO with non-GO groups in AML. Random-effects models were used to calculate pooled effect sizes and 95% confidence intervals (CIs). Relative risk (RR) was used for estimating complete remission (CR), early death, and toxicity. Hazard risk (HR) was accomplished to evaluate survival. Results: Fifteen RCTs and 15 retrospective cohort studies were identified (GO: 4,768; Control: 6,466). GO tended to improve CR (RR 0.95, p = 0.084), followed by significantly improved survival (overall survival: HR 0.86, p = 0.003; event-free survival: HR 0.86, p = 0.015; relapse-free survival: HR 0.83, p = 0.001; cumulative incidence of relapse: HR 0.82, p < 0.001). GO benefits of CR and survival were evident in favorable- and intermediate-risk karyotypes (p ≤ 0.023). GO advantages were also associated with nucleophosmin 1 mutations (p ≤ 0.04), wild-type FMS-like tyrosine kinase 3 internal tandem duplication gene (p ≤ 0.03), age of <70 years (p < 0.05), de novo AML (p ≤ 0.017), and CD33(+) (p ≤ 0.021). Both adding GO into induction therapy (p ≤ 0.011) and a lower (<6 mg/m2) dose of GO (p ≤ 0.03) enhanced survival. Prognosis of combined regimens with GO was heterogeneous in both meta-analysis and NMA, with several binding strategies showing improved prognosis. Additionally, GO was related to increased risk of early death at a higher dose (≥6 mg/m2) (RR 2.01, p = 0.005), hepatic-related adverse effects (RR 1.29, p = 0.02), and a tendency of higher risk for hepatic veno-occlusive disease or sinusoidal obstruction syndrome (RR 1.56, p = 0.072). Conclusions: These data indicated therapeutic benefits and safety of GO in AML, especially in some subtypes, for which further head-to-head RCTs are warranted. Systematic Review Registration: [PROSPERO: https://www.crd.york.ac.uk/prospero/], identifier [CRD42020158540].


Subject(s)
Antineoplastic Agents, Immunological/therapeutic use , Gemtuzumab/therapeutic use , Leukemia, Myeloid, Acute/drug therapy , Antineoplastic Agents, Immunological/administration & dosage , Antineoplastic Agents, Immunological/adverse effects , Biomarkers, Tumor , Gemtuzumab/administration & dosage , Gemtuzumab/adverse effects , Humans , Karyotype , Leukemia, Myeloid, Acute/diagnosis , Leukemia, Myeloid, Acute/etiology , Leukemia, Myeloid, Acute/mortality , Molecular Targeted Therapy , Mutation , Prognosis , Sialic Acid Binding Ig-like Lectin 3/antagonists & inhibitors , Treatment Outcome
19.
Neurochem Int ; 150: 105186, 2021 11.
Article in English | MEDLINE | ID: mdl-34530055

ABSTRACT

Recent genetic and molecular studies have indicated that the innate immune system, especially microglia, have a crucial role in the accumulation of ß-amyloid plaques in Alzheimer's disease (AD). In particular, the CD33 receptor, also called Siglec-3, inhibits the TREM2 receptor-induced phagocytic activity of microglia. CD33 receptors recognize the α2,3 and α2,6-linked sialic groups in tissue glycocalyx, especially sialylated gangliosides in human brain. The CD33 receptor triggers cell-type specific responses, e.g., in microglia, CD33 inhibits phagocytosis, whereas in natural killer cells, it inhibits the cytotoxic activity of the NKG2D receptor. Nonetheless, the regulation of the activity of CD33 receptor needs to be clarified. For example, it seems that hypoxia/ischemia, a potential cause of AD pathology, increases the expression of CD33 and its downstream target SHP-1, a tyrosine phosphatase which suppresses the phagocytosis driven by TREM2. Moreover, hypoxia/ischemia increases the deposition of sialylated gangliosides, e.g., GM1, GM2, GM3, and GD1, which are ligands for inhibitory CD33/Siglec-3 receptors. In addition, ß-amyloid peptides bind to the sialylated gangliosides in raft-like clusters and subsequently these gangliosides act as seeds for the formation of ß-amyloid plaques in AD pathology. It is known that senile plaques contain sialylated GM1, GM2, and GM3 gangliosides, i.e., the same species induced by hypoxia/ischemia treatment. Sialylated gangliosides in plaques might stimulate the CD33/Siglec-3 receptors of microglia and thus impede TREM2-driven phagocytosis. We propose that hypoxia/ischemia, e.g., via the accumulation of sialylated gangliosides, prevents the phagocytosis of ß-amyloid deposits by inhibiting CD33/TREM2 signaling.


Subject(s)
Alzheimer Disease/metabolism , Brain/metabolism , Hypoxia-Ischemia, Brain/metabolism , Membrane Glycoproteins/metabolism , Receptors, Immunologic/metabolism , Sialic Acid Binding Ig-like Lectin 3/metabolism , Signal Transduction/physiology , Alzheimer Disease/pathology , Animals , Brain/pathology , Humans , Hypoxia-Ischemia, Brain/pathology , Membrane Glycoproteins/antagonists & inhibitors , Receptors, Immunologic/antagonists & inhibitors , Sialic Acid Binding Ig-like Lectin 3/antagonists & inhibitors
20.
Clin Cancer Res ; 27(2): 622-631, 2021 01 15.
Article in English | MEDLINE | ID: mdl-33148666

ABSTRACT

PURPOSE: Mortality due to acute myeloid leukemia (AML) remains high, and the management of relapsed or refractory AML continues to be therapeutically challenging. The reapproval of Mylotarg, an anti-CD33-calicheamicin antibody-drug conjugate (ADC), has provided a proof of concept for an ADC-based therapeutic for AML. Several other ADCs have since entered clinical development of AML, but have met with limited success. We sought to develop a next-generation ADC for AML with a wide therapeutic index (TI) that overcomes the shortcomings of previous generations of ADCs. EXPERIMENTAL DESIGN: We compared the TI of our novel CD33-targeted ADC platform with other currently available CD33-targeted ADCs in preclinical models of AML. Next, using this next-generation ADC platform, we performed a head-to-head comparison of two attractive AML antigens, CD33 and CD123. RESULTS: Our novel ADC platform offered improved safety and TI when compared with certain currently available ADC platforms in preclinical models of AML. Differentiation between the CD33- and CD123-targeted ADCs was observed in safety studies conducted in cynomolgus monkeys. The CD33-targeted ADC produced severe hematologic toxicity, whereas minimal hematologic toxicity was observed with the CD123-targeted ADC at the same doses and exposures. The improved toxicity profile of an ADC targeting CD123 over CD33 was consistent with the more restricted expression of CD123 in normal tissues. CONCLUSIONS: We optimized all components of ADC design (i.e., leukemia antigen, antibody, and linker-payload) to develop an ADC that has the potential to translate into an effective new therapy against AML.


Subject(s)
Gemtuzumab/therapeutic use , Immunoconjugates/therapeutic use , Interleukin-3 Receptor alpha Subunit/antagonists & inhibitors , Leukemia, Myeloid, Acute/drug therapy , Sialic Acid Binding Ig-like Lectin 3/antagonists & inhibitors , Animals , Antineoplastic Agents, Immunological/immunology , Antineoplastic Agents, Immunological/pharmacokinetics , Antineoplastic Agents, Immunological/therapeutic use , Area Under Curve , Cell Line, Tumor , Cell Survival/drug effects , Disease Models, Animal , Gemtuzumab/immunology , Gemtuzumab/pharmacokinetics , HL-60 Cells , Humans , Immunoconjugates/immunology , Immunoconjugates/pharmacokinetics , Interleukin-3 Receptor alpha Subunit/immunology , Leukemia, Myeloid, Acute/immunology , Leukemia, Myeloid, Acute/metabolism , Macaca fascicularis , Mice , Sialic Acid Binding Ig-like Lectin 3/immunology , Tumor Burden/drug effects , Xenograft Model Antitumor Assays/methods
SELECTION OF CITATIONS
SEARCH DETAIL