Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Pept Sci (Hoboken) ; 113(3): e24199, 2021 May.
Article in English | MEDLINE | ID: mdl-35859761

ABSTRACT

HYD1 is an all D-amino acid linear 10-mer peptide that was discovered by one-bead-one-compound screening. HYD1 has five hydrophobic amino acids flanked by polar amino acids. Alanine scanning studies showed that alternating hydrophobic amino acid residues and N- and C-terminal lysine side chains were contributors to the biological activity of the linear 10-mer analogs. This observation led us to hypothesize that display of the hydrophobic pentapeptide sequence of HYD1 in a cyclic beta-hairpin-like scaffold could lead to better bioavailability and biological activity. An amphipathic pentapeptide sequence was used to form an antiparallel strand and those strands were linked via dipeptide-like sequences selected to promote ß-turns. Early cyclic analogs were more active but otherwise mimicked the biological activity of the linear HYD1 peptide. The cyclic peptidomimetics were synthesized using standard Fmoc solid phase synthesis to form linear peptides, followed by solution phase or on-resin cyclization. SAR studies were carried out with an aim to increase the potency of these drug candidates for the killing of multiple myeloma cells in vitro. The solution structures of 1, 5, and 10 were elucidated using NMR spectroscopy. 1H NMR and 2D TOCSY studies of these peptides revealed a downfield Hα proton chemical shift and 2D NOE spectral analysis consistent with a ß-hairpin-like structure.

3.
Neoplasia ; 19(9): 716-733, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28830009

ABSTRACT

JAA-F11 is a highly specific mouse monoclonal to the Thomsen-Friedenreich Antigen (TF-Ag) which is an alpha-O-linked disaccharide antigen on the surface of ~80% of human carcinomas, including breast, lung, colon, bladder, ovarian, and prostate cancers, and is cryptic on normal cells. JAA-F11 has potential, when humanized, for cancer immunotherapy for multiple cancer types. Humanization of JAA-F11, was performed utilizing complementarity determining regions grafting on a homology framework. The objective herein is to test the specificity, affinity and biology efficacy of the humanized JAA-F11 (hJAA-F11). Using a 609 target glycan array, 2 hJAA-F11 constructs were shown to have excellent chemical specificity, binding only to TF-Ag alpha-linked structures and not to TF-Ag beta-linked structures. The relative affinity of these hJAA-F11 constructs for TF-Ag was improved over the mouse antibody, while T20 scoring predicted low clinical immunogenicity. The hJAA-F11 constructs produced antibody-dependent cellular cytotoxicity in breast and lung tumor lines shown to express TF-Ag by flow cytometry. Internalization of hJAA-F11 into cancer cells was also shown using a surface binding ELISA and confirmed by immunofluorescence microscopy. Both the naked hJAA-F11 and a maytansine-conjugated antibody (hJAA-F11-DM1) suppressed in vivo tumor progression in a human breast cancer xenograft model in SCID mice. Together, our results support the conclusion that the humanized antibody to the TF-Ag has potential as an adjunct therapy, either directly or as part of an antibody drug conjugate, to treat breast cancer, including triple negative breast cancer which currently has no targeted therapy, as well as lung cancer.

4.
Mol Cancer Ther ; 12(11): 2446-58, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24048737

ABSTRACT

Our laboratory recently reported that treatment with the d-amino acid containing peptide HYD1 induces necrotic cell death in multiple myeloma cell lines. Because of the intriguing biological activity and promising in vivo activity of HYD1, we pursued strategies for increasing the therapeutic efficacy of the linear peptide. These efforts led to a cyclized peptidomimetic, MTI-101, with increased in vitro activity and robust in vivo activity as a single agent using two myeloma models that consider the bone marrow microenvironment. MTI-101 treatment similar to HYD1 induced reactive oxygen species, depleted ATP levels, and failed to activate caspase-3. Moreover, MTI-101 is cross-resistant in H929 cells selected for acquired resistance to HYD1. Here, we pursued an unbiased chemical biology approach using biotinylated peptide affinity purification and liquid chromatography/tandem mass spectrometry analysis to identify binding partners of MTI-101. Using this approach, CD44 was identified as a predominant binding partner. Reducing the expression of CD44 was sufficient to induce cell death in multiple myeloma cell lines, indicating that multiple myeloma cells require CD44 expression for survival. Ectopic expression of CD44s correlated with increased binding of the FAM-conjugated peptide. However, ectopic expression of CD44s was not sufficient to increase the sensitivity to MTI-101-induced cell death. Mechanistically, we show that MTI-101-induced cell death occurs via a Rip1-, Rip3-, or Drp1-dependent and -independent pathway. Finally, we show that MTI-101 has robust activity as a single agent in the SCID-Hu bone implant and 5TGM1 in vivo model of multiple myeloma.


Subject(s)
Antineoplastic Agents/metabolism , Antineoplastic Agents/pharmacology , Hyaluronan Receptors/metabolism , Multiple Myeloma/metabolism , Multiple Myeloma/pathology , Oligopeptides/pharmacology , Peptides, Cyclic/pharmacology , Animals , Antineoplastic Agents/therapeutic use , Caspase 3/metabolism , Cell Line, Tumor , Chromatography, Liquid , Cyclization , Humans , Mice , Mice, Inbred C57BL , Necrosis/chemically induced , Neoplasms, Experimental , Oligopeptides/chemistry , Oligopeptides/therapeutic use , Peptides, Cyclic/metabolism , Peptides, Cyclic/therapeutic use , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , Tandem Mass Spectrometry
5.
J Med Chem ; 56(10): 3768-82, 2013 May 23.
Article in English | MEDLINE | ID: mdl-23600925

ABSTRACT

Cyclin-dependent kinases (CDKs) are serine/threonine protein kinases that act as key regulatory elements in cell cycle progression. We describe the development of highly potent diaminothiazole inhibitors of CDK2 (IC50 = 0.0009-0.0015 µM) from a single hit compound with weak inhibitory activity (IC50 = 15 µM), discovered by high-throughput screening. Structure-based design was performed using 35 cocrystal structures of CDK2 liganded with distinct analogues of the parent compound. The profiling of compound 51 against a panel of 339 kinases revealed high selectivity for CDKs, with preference for CDK2 and CDK5 over CDK9, CDK1, CDK4, and CDK6. Compound 51 inhibited the proliferation of 13 out of 15 cancer cell lines with IC50 values between 0.27 and 6.9 µM, which correlated with the complete suppression of retinoblastoma phosphorylation and the onset of apoptosis. Combined, the results demonstrate the potential of this new inhibitors series for further development into CDK-specific chemical probes or therapeutics.


Subject(s)
Cyclin-Dependent Kinases/antagonists & inhibitors , Thiazoles/chemical synthesis , Thiazoles/pharmacology , Apoptosis/drug effects , Blotting, Western , Breast Neoplasms/drug therapy , Caspases/metabolism , Cell Cycle/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Coloring Agents , Computer Simulation , Cyclin-Dependent Kinase 2/antagonists & inhibitors , Cyclin-Dependent Kinases/chemistry , Drug Screening Assays, Antitumor , Female , Glycogen Synthase Kinase 3/antagonists & inhibitors , Glycogen Synthase Kinase 3 beta , High-Throughput Screening Assays , Humans , Indicators and Reagents , Male , Models, Molecular , Phosphorylation , Structure-Activity Relationship , Testicular Neoplasms/drug therapy , Testicular Neoplasms/pathology , Tetrazolium Salts
6.
Adv Pharmacol ; 65: 143-89, 2012.
Article in English | MEDLINE | ID: mdl-22959026

ABSTRACT

Multiple myeloma (MM) is an incurable hematological cancer involving proliferation of abnormal plasma cells that infiltrate the bone marrow (BM) and secrete monoclonal antibodies. The disease is clinically characterized by bone lesions, anemia, hypercalcemia, and renal failure. MM is presently treated with conventional therapies like melphalan, doxorubicin, and prednisone; or novel therapies like thalidomide, lenalidomide, and bortezomib; or with procedures like autologous stem cell transplantation. Unfortunately, these therapies fail to eliminate the minimal residual disease that remains persistent within the confines of the BM of MM patients. Mounting evidence indicates that components of the BM-including extracellular matrix, cytokines, chemokines, and growth factors-provide a sanctuary for subpopulations of MM. This co-dependent development of the disease in the context of the BM not only ensures the survival and growth of the plasma cells but contributes to de novo drug resistance. In addition, by fostering homing, angiogenesis, and osteolysis, this crosstalk plays a critical role in the progression of the disease. Not surprisingly then, over the past decade, several strategies have been developed to disrupt this communication between the plasma cells and the BM components including antibodies, peptides, and inhibitors of signaling pathways. Ultimately, the goal is to use these therapies in combination with the existing antimyeloma agents in order to further reduce or abolish minimal residual disease and improve patient outcomes.


Subject(s)
Molecular Targeted Therapy , Multiple Myeloma/drug therapy , Multiple Myeloma/pathology , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Cell Adhesion/drug effects , Disease Progression , Humans , Signal Transduction/drug effects
7.
Mol Cancer Ther ; 10(12): 2257-66, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21980133

ABSTRACT

We recently reported that the ß1 integrin antagonist, referred to as HYD1, induces necrotic cell death in myeloma cell lines as a single agent using in vitro and in vivo models. In this article, we sought to delineate the determinants of sensitivity and resistance toward HYD1-induced cell death. To this end, we developed an HYD1 isogenic resistant myeloma cell line by chronically exposing H929 myeloma cells to increasing concentrations of HYD1. Our data indicate that the acquisition of resistance toward HYD1 correlates with reduced levels of the cleaved α4 integrin subunit. Consistent with reduced VLA-4 (α4ß1) expression, the resistant variant showed ablated functional binding to fibronectin, VCAM-1, and the bone marrow stroma cell line HS-5. The reduction in binding of the resistant cell line to HS-5 cells translated to a compromised cell adhesion-mediated drug resistant phenotype as shown by increased sensitivity to melphalan- and bortezomib-induced cell death in the bone marrow stroma coculture model of drug resistance. Importantly, we show that HYD1 is more potent in relapsed myeloma specimens than newly diagnosed patients, a finding that correlated with α4 integrin expression. Collectively, these data indicate that this novel d-amino acid peptide may represent a good candidate for pursuing clinical trials in relapsed myeloma and in particular patients with high levels of α4 integrin. Moreover, our data provide further rationale for continued preclinical development of HYD1 and analogues of HYD1 for the treatment of multiple myeloma and potentially other tumors that home and/or metastasize to the bone.


Subject(s)
Drug Resistance, Neoplasm/genetics , Integrin alpha4/genetics , Multiple Myeloma/pathology , Oligopeptides/therapeutic use , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Bone Neoplasms/genetics , Bone Neoplasms/metabolism , Bone Neoplasms/secondary , Cell Adhesion/drug effects , Cell Adhesion/genetics , Cell Line, Tumor , Down-Regulation/genetics , Drug Evaluation, Preclinical , Drug Resistance, Neoplasm/physiology , Gene Expression Regulation, Neoplastic/drug effects , Humans , Integrin alpha4/metabolism , Integrin beta Chains/metabolism , Multiple Myeloma/drug therapy , Multiple Myeloma/genetics , Multiple Myeloma/metabolism , Oligopeptides/pharmacology , Phenotype , Protein Isoforms/genetics , Protein Isoforms/metabolism
8.
Dig Dis Sci ; 55(3): 739-46, 2010 Mar.
Article in English | MEDLINE | ID: mdl-19294505

ABSTRACT

PURPOSE: To replicate the association of IL23R R381Q (rs11209026) with inflammatory bowel disease (IBD), examine the effect of the two nonsynonymous variations, Q3H and L310P, on IBD, and to study gender distribution of these variants in IBD patients. RESULTS: IL23R R381Q was associated with Crohn's disease (CD) (P = 0.010), but not with ulcerative colitis (UC); L310P was associated with UC (P = 0.004), but not with CD; no association was observed for Q3H with CD or UC. A female-specific association of R381Q with CD (P = 0.041), and of L310P with UC (P = 0.008) was observed. CONCLUSION: We replicated the association of IL23R R381Q with CD but not UC, and we observed an association of L310P with UC, but not CD, in a central Pennsylvania population. Further analysis of the distribution of IL23R variants revealed that these effects were largely female-specific. The results suggest that IL23R R381Q confers protection against CD and that L310P confers protection against UC in females.


Subject(s)
Inflammatory Bowel Diseases/genetics , Colitis, Ulcerative/etiology , Crohn Disease/genetics , Female , Genotype , Humans , Male , Sex Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...