Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Leukemia ; 33(2): 390-402, 2019 02.
Article in English | MEDLINE | ID: mdl-30038380

ABSTRACT

Genomic analyses of chronic lymphocytic leukemia (CLL) identified somatic mutations and associations of clonal diversity with adverse outcomes. Clonal evolution likely has therapeutic implications but its dynamic is less well studied. We studied clonal composition and prognostic value of seven recurrently mutated driver genes using targeted next-generation sequencing in 643 CLL patients and found higher frequencies of mutations in TP53 (35 vs. 12%, p < 0.001) and SF3B1 (20 vs. 11%, p < 0.05) and increased number of (sub)clonal (p < 0.0001) mutations in treated patients. We next performed an in-depth evaluation of clonal evolution on untreated CLL patients (50 "progressors" and 17 matched "non-progressors") using a 404 gene-sequencing panel and identified novel mutated genes such as AXIN1, SDHA, SUZ12, and FOXO3. Progressors carried more mutations at initial presentation (2.5 vs. 1, p < 0.0001). Mutations in specific genes were associated with increased (SF3B1, ATM, and FBXW7) or decreased progression risk (AXIN1 and MYD88). Mutations affecting specific signaling pathways, such as Notch and MAP kinase pathway were enriched in progressive relative to non-progressive patients. These data extend earlier findings that specific genomic alterations and diversity of subclones are associated with disease progression and persistence of disease in CLL and identify novel recurrently mutated genes and associated outcomes.


Subject(s)
Biomarkers, Tumor/genetics , Clonal Evolution , Leukemia, Lymphocytic, Chronic, B-Cell/mortality , Mutation , Clinical Trials as Topic , Disease Progression , High-Throughput Nucleotide Sequencing , Humans , Leukemia, Lymphocytic, Chronic, B-Cell/genetics , Leukemia, Lymphocytic, Chronic, B-Cell/pathology , Prognosis , Survival Rate
2.
Bioorg Med Chem ; 22(21): 5917-23, 2014 Nov 01.
Article in English | MEDLINE | ID: mdl-25284254

ABSTRACT

Immunoadjuvants are used to potentiate the activity of modern subunit vaccines that are based on molecular antigens. An emerging approach involves the combination of multiple adjuvants in a single formulation to achieve optimal vaccine efficacy. Herein, to investigate such potential synergies, we synthesized novel adjuvant conjugates based on the saponin natural product QS-21 and the aldehyde tucaresol via chemoselective acylation of an amine at the terminus of the acyl chain domain in QS saponin variants. In a preclinical mouse vaccination model, these QS saponin-tucaresol conjugates induced antibody responses similar to or slightly higher than those generated with related QS saponin variants lacking the tucaresol motif. The conjugates retained potent adjuvant activity, low toxicity, and improved activity-toxicity profiles relative to QS-21 itself and induced IgG subclass profiles similar to those of QS-21, indicative of both Th1 cellular and Th2 humoral immune responses. This study opens the door to installation of other substituents at the terminus of the acyl chain domain to develop additional QS saponin conjugates with desirable immunologic properties.


Subject(s)
Adjuvants, Immunologic/chemistry , Adjuvants, Immunologic/pharmacology , Benzaldehydes/chemistry , Benzaldehydes/pharmacology , Benzoates/chemistry , Benzoates/pharmacology , Saponins/chemistry , Saponins/pharmacology , Adjuvants, Immunologic/administration & dosage , Adjuvants, Immunologic/chemical synthesis , Animals , Benzaldehydes/administration & dosage , Benzaldehydes/chemical synthesis , Benzoates/administration & dosage , Benzoates/chemical synthesis , Immunity, Cellular/drug effects , Immunity, Humoral/drug effects , Immunoglobulin G/analysis , Immunoglobulin G/immunology , Mice , Mice, Inbred C57BL , Saponins/administration & dosage , Saponins/chemical synthesis
3.
Nat Chem ; 6(7): 635-43, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24950335

ABSTRACT

Adjuvants are materials added to vaccines to enhance the immunological response to an antigen. QS-21 is a natural product adjuvant under investigation in numerous vaccine clinical trials, but its use is constrained by scarcity, toxicity, instability and an enigmatic molecular mechanism of action. Herein we describe the development of a minimal QS-21 analogue that decouples adjuvant activity from toxicity and provides a powerful platform for mechanistic investigations. We found that the entire branched trisaccharide domain of QS-21 is dispensable for adjuvant activity and that the C4-aldehyde substituent, previously proposed to bind covalently to an unknown cellular target, is also not required. Biodistribution studies revealed that active adjuvants were retained preferentially at the injection site and the nearest draining lymph nodes compared with the attenuated variants. Overall, these studies have yielded critical insights into saponin structure-function relationships, provided practical synthetic access to non-toxic adjuvants, and established a platform for detailed mechanistic studies.


Subject(s)
Adjuvants, Immunologic/chemistry , Saponins/chemistry , Vaccines/chemical synthesis , Adjuvants, Immunologic/pharmacology , Humans , Saponins/immunology , Saponins/pharmacology , Vaccines/chemistry
4.
J Am Chem Soc ; 134(32): 13448-57, 2012 Aug 15.
Article in English | MEDLINE | ID: mdl-22866694

ABSTRACT

QS-21 is a potent immunostimulatory saponin that is currently under clinical investigation as an adjuvant in various vaccines to treat infectious diseases, cancers, and cognitive disorders. Herein, we report the design, synthesis, and preclinical evaluation of simplified QS-21 congeners to define key structural features that are critical for adjuvant activity. Truncation of the linear tetrasaccharide domain revealed that a trisaccharide variant is equipotent to QS-21, while the corresponding disaccharide and monosaccharide congeners are more toxic and less potent, respectively. Modification of the acyl chain domain in the trisaccharide series revealed that a terminal carboxylic acid is well-tolerated while a terminal amine results in reduced adjuvant activity. Acylation of the terminal amine can, in some cases, restore adjuvant activity and enables the synthesis of fluorescently labeled QS-21 variants. Cellular studies with these probes revealed that, contrary to conventional wisdom, the most highly adjuvant active of these fluorescently labeled saponins does not simply associate with the plasma membrane, but rather is internalized by dendritic cells.


Subject(s)
Adjuvants, Immunologic/pharmacology , Dendritic Cells/drug effects , Saponins/chemistry , Acylation , Adjuvants, Immunologic/chemistry , Animals , Female , Mice , Mice, Inbred C57BL , Molecular Structure
5.
Expert Rev Vaccines ; 10(4): 463-70, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21506644

ABSTRACT

One of the most widely used and potent immunological adjuvants is a mixture of soluble triterpene glycosides purified from the soap bark tree (Quillaja saponaria). Despite challenges in production, quality control, stability and toxicity, the QS-21 fraction from this extract has exhibited exceptional adjuvant properties for a range of antigens. It possesses an ability to augment clinically significant antibody and T-cell responses to vaccine antigens against a variety of infectious diseases, degenerative disorders and cancers. The recent synthesis of active molecules of QS-21 has provided a robust method to produce this leading vaccine adjuvant in high purity as well as to produce novel synthetic QS-21 congeners designed to induce increased immune responsiveness and decreased toxicity.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Cancer Vaccines/immunology , Neoplasms/therapy , Saponins/administration & dosage , Saponins/chemical synthesis , Adjuvants, Immunologic/chemical synthesis , Adjuvants, Immunologic/isolation & purification , Humans , Quillaja/chemistry , Saponins/chemistry , Saponins/isolation & purification
6.
Nucleic Acids Res ; 39(6): 2458-69, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21030439

ABSTRACT

Long-term survival still eludes most patients with leukemia and non-Hodgkin's lymphoma. No approved therapies target the hallmark of the B cell, its mIgM, also known as the B-cell receptor (BCR). Aptamers are small oligonucleotides that can specifically bind to a wide range of target molecules and offer some advantages over antibodies as therapeutic agents. Here, we report the rational engineering of aptamer TD05 into multimeric forms reactive with the BCR that may be useful in biomedical applications. Systematic truncation of TD05 coupled with modification with locked nucleic acids (LNA) increased conformational stability and nuclease resistance. Trimeric and tetrameric versions with optimized polyethyleneglycol (PEG) linker lengths exhibited high avidity at physiological temperatures both in vitro and in vivo. Competition and protease studies showed that the multimeric, optimized aptamer bound to membrane-associated human mIgM, but not with soluble IgM in plasma, allowing the possibility of targeting leukemias and lymphomas in vivo. The B-cell specificity of the multivalent aptamer was confirmed on lymphoma cell lines and fresh clinical leukemia samples. The chemically engineered aptamers, with significantly improved kinetic and biochemical features, unique specificity and desirable pharmacological properties, may be useful in biomedical applications.


Subject(s)
Aptamers, Nucleotide/chemistry , Leukemia, B-Cell/metabolism , Lymphoma, B-Cell/metabolism , Receptors, Antigen, B-Cell/metabolism , Animals , Aptamers, Nucleotide/metabolism , Cell Line, Tumor , Female , Humans , Immunoglobulin M/metabolism , Mice , Mice, Nude , Oligonucleotides/chemistry
7.
Blood ; 115(14): 2919-27, 2010 Apr 08.
Article in English | MEDLINE | ID: mdl-20154217

ABSTRACT

The discovery of JAK2 and MPL mutations in patients with myeloproliferative neoplasms (MPNs) provided important insight into the genetic basis of these disorders and led to the development of JAK2 kinase inhibitors for MPN therapy. Although recent studies have shown that JAK2 kinase inhibitors demonstrate efficacy in a JAK2V617F murine bone marrow transplantation model, the effects of JAK2 inhibitors on MPLW515L-mediated myeloproliferation have not been investigated. In this report, we describe the in vitro and in vivo effects of INCB16562, a small-molecule JAK2 inhibitor. INCB16562 inhibited proliferation and signaling in cell lines transformed by JAK2 and MPL mutations. Compared with vehicle treatment, INCB16562 treatment improved survival, normalized white blood cell counts and platelet counts, and markedly reduced extramedullary hematopoeisis and bone marrow fibrosis. We observed inhibition of STAT3 and STAT5 phosphorylation in vivo consistent with potent inhibition of JAK-STAT signaling. These data suggest JAK2 inhibitor therapy may be of value in the treatment of JAK2V617F-negative MPNs. However, we did not observe a decrease in the size of the malignant clone in the bone marrow of treated mice at the end of therapy, which suggests that JAK2 inhibitor therapy, by itself, was not curative in this MPN model.


Subject(s)
Hematologic Neoplasms/drug therapy , Janus Kinase 2/antagonists & inhibitors , Mutation, Missense , Primary Myelofibrosis/drug therapy , Protein Kinase Inhibitors/pharmacology , Receptors, Thrombopoietin/metabolism , Thrombocytosis/drug therapy , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Disease Models, Animal , Drug Screening Assays, Antitumor/methods , Female , Hematologic Neoplasms/genetics , Hematologic Neoplasms/metabolism , Humans , Janus Kinase 2/genetics , Janus Kinase 2/metabolism , Mice , Mice, Inbred BALB C , Phosphorylation/drug effects , Phosphorylation/genetics , Platelet Count , Primary Myelofibrosis/blood , Primary Myelofibrosis/genetics , Receptors, Thrombopoietin/genetics , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism , STAT5 Transcription Factor/genetics , STAT5 Transcription Factor/metabolism , Signal Transduction/drug effects , Thrombocytosis/blood , Thrombocytosis/genetics
8.
Cancer Res ; 68(19): 8031-8, 2008 Oct 01.
Article in English | MEDLINE | ID: mdl-18829561

ABSTRACT

Vitamin C is an antioxidant vitamin that has been hypothesized to antagonize the effects of reactive oxygen species-generating antineoplastic drugs. The therapeutic efficacy of the widely used antineoplastic drugs doxorubicin, cisplatin, vincristine, methotrexate, and imatinib were compared in leukemia (K562) and lymphoma (RL) cell lines with and without pretreatment with dehydroascorbic acid, the commonly transported form of vitamin C. The effect of vitamin C on viability, clonogenicity, apoptosis, P-glycoprotein, reactive oxygen species (ROS), and mitochondrial membrane potential was determined. Pretreatment with vitamin C caused a dose-dependent attenuation of cytotoxicity, as measured by trypan blue exclusion and colony formation after treatment with all antineoplastic agents tested. Vitamin C given before doxorubicin treatment led to a substantial reduction of therapeutic efficacy in mice with RL cell-derived xenogeneic tumors. Vitamin C treatment led to a dose-dependent decrease in apoptosis in cells treated with the antineoplastic agents that was not due to up-regulation of P-glycoprotein or vitamin C retention modulated by antineoplastics. Vitamin C had only modest effects on intracellular ROS and a more general cytoprotective profile than N-acetylcysteine, suggesting a mechanism of action that is not mediated by ROS. All antineoplastic agents tested caused mitochondrial membrane depolarization that was inhibited by vitamin C. These findings indicate that vitamin C given before mechanistically dissimilar antineoplastic agents antagonizes therapeutic efficacy in a model of human hematopoietic cancers by preserving mitochondrial membrane potential. These results support the hypothesis that vitamin C supplementation during cancer treatment may detrimentally affect therapeutic response.


Subject(s)
Antineoplastic Agents/antagonists & inhibitors , Ascorbic Acid/pharmacology , Neoplasms/pathology , Animals , Antineoplastic Agents/therapeutic use , Ascorbic Acid/metabolism , Cytoprotection/drug effects , Dehydroascorbic Acid/pharmacokinetics , Dose-Response Relationship, Drug , Drug Resistance, Neoplasm/drug effects , Humans , K562 Cells , Mice , Mice, Inbred ICR , Mice, SCID , Neoplasms/drug therapy , Neoplasms/metabolism , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
9.
Blood ; 112(7): 2906-16, 2008 Oct 01.
Article in English | MEDLINE | ID: mdl-18591385

ABSTRACT

Overexpression of antiapoptotic members of the Bcl-2 family is observed in approximately 80% of B-cell lymphomas, contributing to intrinsic and acquired drug resistance. Nullifying the antiapoptotic influence of these proteins can potentially overcome this resistance, and may complement conventional chemotherapy. ABT-737 is a BH3-only mimetic and potent inhibitor of the antiapoptotic Bcl-2 family members Bcl-2, Bcl-X(L), and Bcl-w. In vitro, ABT-737 exhibited concentration-dependent cytotoxicity against a broad panel of lymphoma cell lines including mantle cell lymphoma (MCL) and diffuse large B-cell lymphoma (DLBCL). ABT-737 showed synergism when combined with the proteasome inhibitors bortezomib or carfilzomib in select lymphoma cell lines and induced potent mitochondrial membrane depolarization and apoptosis when combined with either. ABT-737 plus bortezomib also induced significant apoptosis in primary samples of MCL, DLBCL, and chronic lymphocytic leukemia (CLL) but no significant cytotoxic effect was observed in peripheral blood mononuclear cells from healthy donors. In severe combined immunodeficient beige mouse models of MCL, the addition of ABT-737 to bortezomib enhanced efficacy compared with either drug alone and with the control. Collectively, these data suggest that ABT-737 alone or in combination with a proteasome inhibitor represents a novel and potentially important platform for the treatment of B-cell malignancies.


Subject(s)
Antineoplastic Agents/pharmacology , Biphenyl Compounds/pharmacology , Enzyme Inhibitors/pharmacology , Lymphoma/enzymology , Lymphoma/pathology , Molecular Mimicry/drug effects , Nitrophenols/pharmacology , Proteasome Inhibitors , Sulfonamides/pharmacology , Animals , Boronic Acids/pharmacology , Bortezomib , Cell Death/drug effects , Cell Line, Tumor , Dose-Response Relationship, Drug , Drug Resistance, Neoplasm/drug effects , Drug Synergism , Health , Humans , Leukemia, Lymphocytic, Chronic, B-Cell/pathology , Leukocytes, Mononuclear/drug effects , Lymphoma, Large B-Cell, Diffuse/pathology , Lymphoma, Mantle-Cell/pathology , Membrane Potential, Mitochondrial/drug effects , Mice , Microscopy, Confocal , Piperazines/pharmacology , Proto-Oncogene Proteins c-bcl-2/metabolism , Pyrazines/pharmacology , Tissue Donors , Xenograft Model Antitumor Assays
10.
Blood ; 111(11): 5350-8, 2008 Jun 01.
Article in English | MEDLINE | ID: mdl-18292288

ABSTRACT

Overexpression of antiapoptotic members of the Bcl-2 family are observed in approximately 80% of B-cell lymphomas, contributing to intrinsic and acquired drug resistance. Nullifying antiapoptotic function can potentially overcome this in-trinsic and acquired drug resistance. AT-101 is a BH3 mimetic known to be a potent inhibitor of antiapoptotic Bcl-2 family members including Bcl-2, Bcl-X(L), and Mcl-1. In vitro, AT-101 exhibits concentration- and time-dependent cytotoxicity against lymphoma and multiple myeloma cell lines, enhancing the activity of cytotoxic agents. The IC(50) for AT-101 is between 1 and 10 microM for a diverse panel of B-cell lymphomas. AT-101 was synergistic with carfilzomib (C), etoposide (E), doxorubicin (D), and 4-hydroxycyclophosphamide (4-HC) in mantle cell lymphoma (MCL) lines. In a transformed large B-cell lymphoma line (RL), AT-101 was synergistic when sequentially combined with 4-HC, but not when both drugs were added simultaneously. AT-101 also induced potent mitochondrial membrane depolarization (Delta Psi m) and apoptosis when combined with carfilzomib, but not with bortezomib in MCL. In severe combined immunodeficient (SCID) beige mouse models of drug-resistant B-cell lymphoma, 35 mg/kg per day of AT-101 was safe and efficacious. The addition of AT-101 to cyclophosphamide (Cy) and rituximab (R) in a schedule-dependent manner enhanced the efficacy of the conventional therapy.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Gossypol/analogs & derivatives , Lymphoma, B-Cell/drug therapy , Proto-Oncogene Proteins c-bcl-2/drug effects , Animals , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal, Murine-Derived , Apoptosis/drug effects , BH3 Interacting Domain Death Agonist Protein , Biomimetics , Blotting, Western , Cell Line, Tumor , Cyclophosphamide/administration & dosage , Disease Models, Animal , Flow Cytometry , Gossypol/pharmacology , Humans , In Vitro Techniques , Membrane Potential, Mitochondrial/drug effects , Mice , Microscopy, Confocal , Rituximab , Xenograft Model Antitumor Assays
11.
J Clin Invest ; 114(8): 1107-16, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15489958

ABSTRACT

Peptide deformylase activity was thought to be limited to ribosomal protein synthesis in prokaryotes, where new peptides are initiated with an N-formylated methionine. We describe here a new human peptide deformylase (Homo sapiens PDF, or HsPDF) that is localized to the mitochondria. HsPDF is capable of removing formyl groups from N-terminal methionines of newly synthesized mitochondrial proteins, an activity previously not thought to be necessary in mammalian cells. We show that actinonin, a peptidomimetic antibiotic that inhibits HsPDF, also inhibits the proliferation of 16 human cancer cell lines. We designed and synthesized 33 chemical analogs of actinonin; all of the molecules with potent activity against HsPDF also inhibited tumor cell growth, and vice versa, confirming target specificity. Small interfering RNA inhibition of HsPDF protein expression was also antiproliferative. Actinonin treatment of cells led to a tumor-specific mitochondrial membrane depolarization and ATP depletion in a time- and dose-dependent manner; removal of actinonin led to a recovery of the membrane potential consistent with indirect effects on the electron transport chain. In animal models, oral or parenteral actinonin was well tolerated and inhibited human prostate cancer and lung cancer growth. We conclude that HsPDF is a new human mitochondrial enzyme that may provide a novel selective target for anticancer therapy by use of actinonin-based antibiotics.


Subject(s)
Amidohydrolases/metabolism , Anti-Bacterial Agents/metabolism , Antineoplastic Agents/metabolism , Hydroxamic Acids/metabolism , Mitochondria/enzymology , Amidohydrolases/genetics , Amino Acid Sequence , Animals , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Cell Division/physiology , Cell Line, Tumor/drug effects , Enzyme Inhibitors/metabolism , Humans , Hydroxamic Acids/chemistry , Hydroxamic Acids/pharmacology , Membrane Potentials/physiology , Mice , Mitochondria/drug effects , Molecular Sequence Data , Molecular Structure , Neoplasm Transplantation , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Sequence Alignment , Transplantation, Heterologous
SELECTION OF CITATIONS
SEARCH DETAIL
...