Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
1.
Blood ; 141(21): 2599-2614, 2023 05 25.
Article in English | MEDLINE | ID: mdl-36630605

ABSTRACT

PSMD4/Rpn10 is a subunit of the 19S proteasome unit that is involved with feeding target proteins into the catalytic machinery of the 26S proteasome. Because proteasome inhibition is a common therapeutic strategy in multiple myeloma (MM), we investigated Rpn10 and found that it is highly expressed in MM cells compared with normal plasma cells. Rpn10 levels inversely correlated with overall survival in patients with MM. Inducible knockout or knockdown of Rpn10 decreased MM cell viability both in vitro and in vivo by triggering the accumulation of polyubiquitinated proteins, cell cycle arrest, and apoptosis associated with the activation of caspases and unfolded protein response-related pathways. Proteomic analysis revealed that inhibiting Rpn10 increased autophagy, antigen presentation, and the activation of CD4+ T and natural killer cells. We developed an in vitro AlphaScreen binding assay for high-throughput screening and identified a novel Rpn10 inhibitor, SB699551 (SB). Treating MM cell lines, leukemic cell lines, and primary cells from patients with MM with SB decreased cell viability without affecting the viability of normal peripheral blood mononuclear cells. SB inhibited the proliferation of MM cells even in the presence of the tumor-promoting bone marrow milieu and overcame proteasome inhibitor (PI) resistance without blocking the 20S proteasome catalytic function or the 19S deubiquitinating activity. Rpn10 blockade by SB triggered MM cell death via similar pathways as the genetic strategy. In MM xenograft models, SB was well tolerated, inhibited tumor growth, and prolonged survival. Our data suggest that inhibiting Rpn10 will enhance cytotoxicity and overcome PI resistance in MM, providing the basis for further optimization studies of Rpn10 inhibitors for clinical application.


Subject(s)
Multiple Myeloma , Proteasome Endopeptidase Complex , Humans , Proteasome Endopeptidase Complex/metabolism , Ubiquitin/metabolism , Multiple Myeloma/drug therapy , Multiple Myeloma/genetics , Proteomics , Leukocytes, Mononuclear/metabolism , Carrier Proteins/genetics , Proteins/metabolism , RNA-Binding Proteins
2.
Teach Learn Med ; 33(1): 78-88, 2021.
Article in English | MEDLINE | ID: mdl-32917107

ABSTRACT

Problem: Effective clinical workplace learning depends on interprofessional and multidisciplinary learning. However, traditional patient wards are centered around patient care and not so much around education. Other barriers such as time constraints also contribute to suboptimal interprofessional and multidisciplinary learning. Intervention: Six formal and informal learning activities that aimed at stimulation of interprofessional and multidisciplinary learning were designed and introduced in our patient ward to enable optimal integration of clinical practice and learning. Context: The study took place in an internal medicine inpatient ward where daily patient care is performed by specialized teams consisting of different healthcare professionals from the departments of Endocrinology, Nephrology, and Infectious Diseases. In the traditional ward setting, interprofessional and multidisciplinary learning mostly takes place during shared clinical activities. In this article, we describe the development and implementation of a Clinical Teaching Unit to support learning between different healthcare professionals. Impact: The intervention was evaluated with an online questionnaire among 108 nurses, student nurses, clerks, residents, supervising clinicians, and managers. Open-ended questions (response rate 65%) were used to determine the changes in the workplace experienced by the participants since the introduction of the Clinical Teaching Unit and what influenced their learning process and motivation to learn. Closed questions (response rate 46%) aimed to measure the effect of our intervention on collaboration, learning, and the quality of care and education. The results of the open-ended questions showed that participants experienced more interprofessional collaboration and learning. This took place in a less hierarchical, safer work climate which also resulted in perceptions of a better quality of patient care and education. The closed-ended questions showed that the intervention resulted in perceptions of improved collaboration, work culture, quality of care, education, and learning conditions. Lessons Learned: The findings imply that implementation of a Clinical Teaching Unit not only facilitates the integration of patient care and education but also the integration of different professions working together. From the intervention, we also learned that a successful Clinical Teaching Unit requires investment of time and staff, clear communication between healthcare professionals, and dedication of teachers within all professions.


Subject(s)
Cooperative Behavior , Curriculum/standards , Interdisciplinary Communication , Internal Medicine/education , Interprofessional Relations , Professional Competence/standards , Communication , Humans , Interdisciplinary Studies , Students, Medical/statistics & numerical data
3.
Br J Haematol ; 174(3): 397-409, 2016 08.
Article in English | MEDLINE | ID: mdl-27098276

ABSTRACT

Our prior study utilized both in vitro and in vivo multiple myeloma (MM) xenograft models to show that a novel alkylator melphalan-flufenamide (Melflufen) is a more potent anti-MM agent than melphalan and overcomes conventional drug resistance. Here we examined whether this potent anti-MM activity of melflufen versus melphalan is due to their differential effect on DNA damage and repair signalling pathways via γ-H2AX/ATR/CHK1/Ku80. Melflufen-induced apoptosis was associated with dose- and time-dependent rapid phosphorylation of γ-H2AX. Melflufen induces γ-H2AX, ATR, and CHK1 as early as after 2 h exposure in both melphalan-sensitive and -resistant cells. However, melphalan induces γ-H2AX in melphalan-sensitive cells at 6 h and 24 h; no γ-H2AX induction was observed in melphalan-resistant cells even after 24 h exposure. Similar kinetics was observed for ATR and CHK1 in meflufen- versus melphalan-treated cells. DNA repair is linked to melphalan-resistance; and importantly, we found that melphalan, but not melflufen, upregulates Ku80 that repairs DNA double-strand breaks. Washout experiments showed that a brief (2 h) exposure of MM cells to melflufen is sufficient to initiate an irreversible DNA damage and cytotoxicity. Our data therefore suggest that melflufen triggers a rapid, robust, and an irreversible DNA damage which may account for its ability to overcome melphalan-resistance in MM cells.


Subject(s)
Apoptosis/drug effects , DNA Damage , Melphalan/analogs & derivatives , Multiple Myeloma/pathology , Phenylalanine/analogs & derivatives , Antineoplastic Agents, Alkylating/pharmacology , DNA Repair/drug effects , Drug Resistance, Neoplasm/drug effects , Histones/metabolism , Humans , Kinetics , Melphalan/pharmacology , Multiple Myeloma/drug therapy , Phenylalanine/pharmacology , Phosphorylation/drug effects , Signal Transduction/drug effects , Tumor Cells, Cultured
4.
Blood ; 123(5): 706-16, 2014 Jan 30.
Article in English | MEDLINE | ID: mdl-24319254

ABSTRACT

Proteasome inhibitors have demonstrated that targeting protein degradation is effective therapy in multiple myeloma (MM). Here we show that deubiquitylating enzymes (DUBs) USP14 and UCHL5 are more highly expressed in MM cells than in normal plasma cells. USP14 and UCHL5 short interfering RNA knockdown decreases MM cell viability. A novel 19S regulatory particle inhibitor b-AP15 selectively blocks deubiquitylating activity of USP14 and UCHL5 without inhibiting proteasome activity. b-AP15 decreases viability in MM cell lines and patient MM cells, inhibits proliferation of MM cells even in the presence of bone marrow stroma cells, and overcomes bortezomib resistance. Anti-MM activity of b-AP15 is associated with growth arrest via downregulation of CDC25C, CDC2, and cyclin B1 as well as induction of caspase-dependent apoptosis and activation of unfolded protein response. In vivo studies using distinct human MM xenograft models show that b-AP15 is well tolerated, inhibits tumor growth, and prolongs survival. Combining b-AP15 with suberoylanilide hydroxamic acid, lenalidomide, or dexamethasone induces synergistic anti-MM activity. Our preclinical data showing efficacy of b-AP15 in MM disease models validates targeting DUBs in the ubiquitin proteasomal cascade to overcome proteasome inhibitor resistance and provides the framework for clinical evaluation of USP14/UCHL5 inhibitors to improve patient outcome in MM.


Subject(s)
Antineoplastic Agents/pharmacology , Boronic Acids/pharmacology , Multiple Myeloma/drug therapy , Piperidones/pharmacology , Protease Inhibitors/pharmacology , Pyrazines/pharmacology , Ubiquitin Thiolesterase/antagonists & inhibitors , Animals , Apoptosis/drug effects , Bortezomib , Cell Line, Tumor , Drug Resistance, Neoplasm/drug effects , Endoplasmic Reticulum Stress/drug effects , Female , Gene Expression Regulation, Neoplastic , Humans , Mice , Multiple Myeloma/genetics , Multiple Myeloma/pathology , Ubiquitin Thiolesterase/genetics , Up-Regulation
5.
Br J Haematol ; 171(5): 798-812, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26456076

ABSTRACT

The proteasome inhibitor bortezomib is an effective therapy for the treatment of relapsed and refractory multiple myeloma (RRMM); however, prolonged treatment can be associated with toxicity, peripheral neuropathy and drug resistance. Our earlier studies showed that the novel proteasome inhibitor marizomib is distinct from bortezomib in its chemical structure, mechanisms of action and effects on proteasomal activities, and that it can overcome bortezomib resistance. Pomalidomide, like lenalidomide, has potent immunomodulatory activity and has been approved by the US Food and Drug Administration for the treatment of RRMM. Here, we demonstrate that combining low concentrations of marizomib with pomalidomide induces synergistic anti-MM activity. Marizomib plus pomalidomide-induced apoptosis is associated with: (i) activation of caspase-8, caspase-9, caspase-3 and PARP cleavage, (ii) downregulation of cereblon (CRBN), IRF4, MYC and MCL1, and (iii) suppression of chymotrypsin-like, caspase-like, and trypsin-like proteasome activities. CRBN-siRNA attenuates marizomib plus pomalidomide-induced MM cells death. Furthermore, marizomib plus pomalidomide inhibits the migration of MM cells and tumour-associated angiogenesis, as well as overcomes cytoprotective effects of bone marrow microenvironment. In human MM xenograft model studies, the combination of marizomib and pomalidomide is well tolerated, inhibits tumour growth and prolongs survival. These preclinical studies provide the rationale for on-going clinical trials of combined marizomib and pomalidomide to improve outcome in patients with RRMM.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Multiple Myeloma/drug therapy , Adaptor Proteins, Signal Transducing , Angiogenesis Inhibitors/pharmacology , Animals , Apoptosis/drug effects , Caspases/metabolism , Cell Line, Tumor , Down-Regulation , Drug Resistance, Neoplasm/drug effects , Drug Synergism , Humans , Lactones/administration & dosage , Lactones/pharmacology , Mice, SCID , Peptide Hydrolases/metabolism , Poly(ADP-ribose) Polymerases/metabolism , Proteasome Inhibitors/pharmacology , Pyrroles/administration & dosage , Pyrroles/pharmacology , RNA, Small Interfering/metabolism , Thalidomide/administration & dosage , Thalidomide/analogs & derivatives , Thalidomide/pharmacology , Transplantation, Heterologous , Ubiquitin-Protein Ligases
6.
Cancer Cell ; 7(2): 129-41, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15710326

ABSTRACT

The Bcr-Abl tyrosine kinase oncogene causes chronic myelogenous leukemia (CML) and Philadelphia chromosome-positive (Ph+) acute lymphoblastic leukemia (ALL). We describe a novel selective inhibitor of Bcr-Abl, AMN107 (IC50 <30 nM), which is significantly more potent than imatinib, and active against a number of imatinib-resistant Bcr-Abl mutants. Crystallographic analysis of Abl-AMN107 complexes provides a structural explanation for the differential activity of AMN107 and imatinib against imatinib-resistant Bcr-Abl. Consistent with its in vitro and pharmacokinetic profile, AMN107 prolonged survival of mice injected with Bcr-Abl-transformed hematopoietic cell lines or primary marrow cells, and prolonged survival in imatinib-resistant CML mouse models. AMN107 is a promising new inhibitor for the therapy of CML and Ph+ ALL.


Subject(s)
Antineoplastic Agents/pharmacology , Fusion Proteins, bcr-abl/antagonists & inhibitors , Fusion Proteins, bcr-abl/genetics , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Pyrimidines/chemistry , Pyrimidines/pharmacology , Animals , Benzamides , Bone Marrow Cells/cytology , Cell Line , Cell Line, Tumor , Cell Survival , Crystallography, X-Ray , Dose-Response Relationship, Drug , Drug Resistance, Neoplasm , Hematopoietic Stem Cells/cytology , Imatinib Mesylate , Inhibitory Concentration 50 , Mice , Models, Biological , Models, Chemical , Mutation , Mycoplasma/metabolism , Phosphorylation , Piperazines/pharmacology , Retroviridae/genetics , Time Factors
7.
Blood ; 115(21): 4206-16, 2010 May 27.
Article in English | MEDLINE | ID: mdl-20299508

ABSTRACT

Many clinically validated kinases, such as BCR-ABL, c-Kit, PDGFR, and EGFR, become resistant to adenosine triphosphate-competitive inhibitors through mutation of the so-called gatekeeper amino acid from a threonine to a large hydrophobic amino acid, such as an isoleucine or methionine. We have developed a new class of adenosine triphosphate competitive inhibitors, exemplified by HG-7-85-01, which is capable of inhibiting T315I- BCR-ABL (clinically observed in chronic myeloid leukemia), T670I-c-Kit (clinically observed in gastrointestinal stromal tumors), and T674I/M-PDGFRalpha (clinically observed in hypereosinophilic syndrome). HG-7-85-01 is unique among all currently reported kinase inhibitors in having the ability to accommodate either a gatekeeper threonine, present in the wild-type forms of these kinases, or a large hydrophobic amino acid without becoming a promiscuous kinase inhibitor. The distinctive ability of HG-7-85-01 to simultaneously inhibit both wild-type and mutant forms of several kinases of clinical relevance is an important step in the development of the next generation of tyrosine kinase inhibitors.


Subject(s)
Fusion Proteins, bcr-abl/antagonists & inhibitors , Mutant Proteins/antagonists & inhibitors , Piperazines/chemistry , Piperazines/pharmacology , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-kit/antagonists & inhibitors , Receptor, Platelet-Derived Growth Factor alpha/antagonists & inhibitors , Thiazoles/chemistry , Thiazoles/pharmacology , src-Family Kinases/antagonists & inhibitors , Amino Acid Substitution , Animals , Antineoplastic Agents/pharmacology , Benzamides , Cell Line, Tumor , Drug Discovery , Drug Resistance, Neoplasm/genetics , Humans , Imatinib Mesylate , In Vitro Techniques , Molecular Structure , Mutation, Missense , Pyrimidines/pharmacology , Threonine/genetics
8.
Blood Cancer J ; 12(4): 50, 2022 04 01.
Article in English | MEDLINE | ID: mdl-35365613

ABSTRACT

Interaction of plasmacytoid dendritic cells (pDCs) with multiple myeloma (MM) cells, T- or NK-effector cells in the bone marrow (BM) microenvironment induces tumor cell growth, as well as inhibits innate and adaptive immune responses. Defining pDC-MM interaction-triggered immunosuppressive mechanism(s) will enable design of interventional therapies to augment anti-MM immunity. In the present study, we show that pDC-MM interactions induce metabolic enzyme Ecto-5' Nucleotidase/CD73 in both pDCs and MM cells. Gene expression database from MM patients showed that CD73 levels inversely correlate with overall survival. Using our pDC-MM coculture models, we found that blockade of CD73 with anti-CD73 Abs: decreases adenosine levels; activates MM patient pDCs; triggers cytotoxic T lymphocytes (CTL) activity against autologous patient MM cells. Combination of anti-CD73 Abs and an immune-stimulating agent TLR-7 agonist enhances autologous MM-specific CD8+ CTL activity. Taken together, our preclinical data suggest that the therapeutic targeting of CD73, alone or in combination with TLR-7 agonist, represents a promising novel strategy to restore host anti-MM immunity.


Subject(s)
Multiple Myeloma , 5'-Nucleotidase/metabolism , 5'-Nucleotidase/therapeutic use , Dendritic Cells/metabolism , Humans , Immunotherapy , Killer Cells, Natural , Multiple Myeloma/drug therapy , Tumor Microenvironment
9.
J Bone Jt Infect ; 7(6): 241-248, 2022.
Article in English | MEDLINE | ID: mdl-36532293

ABSTRACT

The aim of this study was to review the available literature concerning Madura foot ("mycetoma") caused by Madurella mycetomatis in immunocompromised patients. With a systematic literature search, we identified only three papers, describing a total of three immunocompromised patients. Hence, the clinical presentation and prognosis of the disease in this patient population have not yet been well described. In addition, we present a case from our institution, illustrating the complexity of the treatment of this rare disease. Although very rare in non-endemic countries, we emphasize that mycetoma should be included in the differential diagnoses of (immunocompromised) patients who have been residing in a geographical area where the disease is endemic and presenting with soft tissue inflammation of one of the extremities.

10.
Br J Haematol ; 155(5): 588-98, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21950728

ABSTRACT

SIRT1 belongs to the silent information regulator 2 (Sir2) protein family of enzymes and functions as a NAD(+) -dependent class III histone deacetylase. Here, we examined the anti-multiple myeloma (MM) activity of a novel oral agent, SRT1720, which targets SIRT1. Treatment of MM cells with SRT1720 inhibited growth and induced apoptosis in MM cells resistant to conventional and bortezomib therapies without significantly affecting the viability of normal cells. Mechanistic studies showed that anti-MM activity of SRT1720 is associated with: (i) activation of caspase-8, caspase-9, caspase-3, poly(ADP) ribose polymerase; (ii) increase in reactive oxygen species; (iii) induction of phosphorylated ataxia telangiectasia mutated/checkpoint kinase 2 signalling; (iv) decrease in vascular endothelial growth factor-induced migration of MM cells and associated angiogenesis; and (v) inhibition of nuclear factor-κB. Blockade of ATM attenuated SRT1720-induced MM cell death. In animal tumour model studies, SRT1720 inhibited MM tumour growth. Finally, SRT1720 enhanced the cytotoxic activity of bortezomib or dexamethasone. Our preclinical studies provide the rationale for novel therapeutics targeting SIRT1 in MM.


Subject(s)
Heterocyclic Compounds, 4 or More Rings/pharmacology , Multiple Myeloma/drug therapy , Animals , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Boronic Acids/administration & dosage , Bortezomib , Cell Line, Tumor , Dexamethasone/administration & dosage , Female , Heterocyclic Compounds, 4 or More Rings/administration & dosage , Heterocyclic Compounds, 4 or More Rings/chemistry , Humans , Immunohistochemistry , Mice , Multiple Myeloma/metabolism , Multiple Myeloma/pathology , Pyrazines/administration & dosage , Random Allocation , Xenograft Model Antitumor Assays
11.
Leukemia ; 35(2): 550-561, 2021 02.
Article in English | MEDLINE | ID: mdl-32424294

ABSTRACT

Our prior study showed that inhibition of 19S proteasome-associated ubiquitin receptor Rpn13 can overcome bortezomib resistance in MM cells. Here, we performed proteomic analysis of Rpn13 inhibitor (RA190)-treated MM cells and identified an antioxidant enzyme superoxide dismutase (SOD1) as a mediator of Rpn13 signaling. SOD1 levels are higher in MM patient cells versus normal PBMCs; and importantly, SOD1 expression correlates with the progression of disease and shorter survival. Functional validation studies show that RA190-induced cytotoxicity in bortezomib-sensitive and -resistant MM cells is associated with decrease in SOD1 levels; conversely, forced expression of SOD1 inhibits RA190-induced cell death. Genetic knockdown and biochemical blockade of SOD1 with LCS-1 sensitizes bortezomib-resistant MM cells to bortezomib. SOD1 inhibitor LCS-1 decreases viability in MM cell lines and patient cells. LCS-1-induced cell death is associated with: (1) increase in superoxide and ROS levels; (2) activation of caspases, and p53/p21 signaling; (3) decrease in MCL-1, BCLxL, CDC2, cyclin-B1, and c-Myc; (4) ER stress response; and (5) inhibition of proteasome function. In animal model studies, LCS-1 inhibits xenografted bortezomib-resistant human MM cell growth and prolongs host survival. Our studies therefore show that targeting Rpn13 overcomes bortezomib resistance by decreasing cellular SOD1 levels, and provide the rationale for novel therapeutics targeting SOD1 to improve patient outcome in MM.


Subject(s)
Biomarkers, Tumor/metabolism , Bortezomib/pharmacology , Drug Resistance, Neoplasm , Gene Expression Regulation, Neoplastic/drug effects , Intracellular Signaling Peptides and Proteins/metabolism , Multiple Myeloma/drug therapy , Proteome/analysis , Animals , Antineoplastic Agents/pharmacology , Apoptosis , Caspases/metabolism , Cell Proliferation , Female , Humans , Intracellular Signaling Peptides and Proteins/genetics , Mice , Mice, SCID , Multiple Myeloma/metabolism , Multiple Myeloma/pathology , Prognosis , Proteasome Endopeptidase Complex/drug effects , Survival Rate , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
12.
Blood ; 112(13): 5161-70, 2008 Dec 15.
Article in English | MEDLINE | ID: mdl-18820131

ABSTRACT

An attractive target for therapeutic intervention is constitutively activated, mutant FLT3, which is expressed in a subpopulation of patients with acute myelocyic leukemia (AML) and is generally a poor prognostic indicator in patients under the age of 65 years. PKC412 is one of several mutant FLT3 inhibitors that is undergoing clinical testing, and which is currently in late-stage clinical trials. However, the discovery of drug-resistant leukemic blast cells in PKC412-treated patients with AML has prompted the search for novel, structurally diverse FLT3 inhibitors that could be alternatively used to override drug resistance. Here, we report the potent and selective antiproliferative effects of the novel mutant FLT3 inhibitor NVP-AST487 on primary patient cells and cell lines expressing FLT3-ITD or FLT3 kinase domain point mutants. NVP-AST487, which selectively targets mutant FLT3 protein kinase activity, is also shown to override PKC412 resistance in vitro, and has significant antileukemic activity in an in vivo model of FLT3-ITD(+) leukemia. Finally, the combination of NVP-AST487 with standard chemotherapeutic agents leads to enhanced inhibition of proliferation of mutant FLT3-expressing cells. Thus, we present a novel class of FLT3 inhibitors that displays high selectivity and potency toward FLT3 as a molecular target, and which could potentially be used to override drug resistance in AML.


Subject(s)
Antineoplastic Agents/pharmacology , Carbanilides/pharmacology , Drug Resistance, Neoplasm , Protein Kinase Inhibitors/pharmacology , fms-Like Tyrosine Kinase 3/antagonists & inhibitors , Animals , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Cell Line , Leukemia, Myeloid, Acute/drug therapy , Mice , Mutant Proteins/antagonists & inhibitors , Protein Kinase C/antagonists & inhibitors , Staurosporine/analogs & derivatives
13.
Leukemia ; 34(2): 567-577, 2020 02.
Article in English | MEDLINE | ID: mdl-31462737

ABSTRACT

Our prior studies showed that dysfunctional plasmacytoid dendritic cells (pDCs) contribute to multiple myeloma (MM) pathogenesis. Specifically, pDC interactions with tumor and T/NK effector cells in the bone marrow (BM) milieu induce immune suppression and MM cell proliferation. Delineation of the mechanism(s) mediating pDC-MM-T-NK cell interactions will identify novel therapeutic targets to both enhance cytotoxicity and anti-MM immunity. Here, we utilized gene expression profiling (GEP) to show that pDC-MM interactions trigger upregulation of immunosuppressive tryptophan catabolic kynurenine (Kyn) pathway. In particular, we show that Kyn pathway enzyme kynurenine-3-monooxygenase (KMO) is upregulated during pDC-MM interactions. Using our coculture models of patient autologous pDC-T-NK-MM cells, we show that pharmacological blockade of KMO activates pDCs and triggers both MM-specific cytotoxic T-cell lymphocytes (CTL) and NK cells cytolytic activity against tumor cells. Furthermore, we show that simultaneous inhibition of Kyn pathway and immune checkpoint PD-L1 enhances antitumor immunity and cytotoxicity in MM. Our preclinical data therefore provide the basis for novel immune-based therapeutic approaches targeting Kyn metabolic pathway enzyme KMO, alone or in combination with anti-PD-L1 Ab, to restore anti-MM immune responses in MM.


Subject(s)
Kynurenine/metabolism , Multiple Myeloma/metabolism , Tryptophan/metabolism , B7-H1 Antigen/metabolism , Bone Marrow/metabolism , Cell Line, Tumor , Dendritic Cells/metabolism , Humans , Killer Cells, Natural/metabolism , Multiple Myeloma/immunology , Multiple Myeloma/therapy , T-Lymphocytes/metabolism
14.
Oncogene ; 39(13): 2786-2796, 2020 03.
Article in English | MEDLINE | ID: mdl-32024967

ABSTRACT

Bone marrow plasmacytoid dendritic cells (pDCs) in patients with multiple myeloma (MM) promote tumor growth, survival, drug resistance, and immune suppression. Understanding the molecular signaling crosstalk among the tumor cells, pDCs and immune cells will identify novel therapeutic approaches to enhance anti-MM immunity. Using oligonucleotide arrays, we found that pDC-MM interactions induce metabolic enzyme Alpha-Enolase (ENO1) in both pDCs and MM cells. Analysis of MM patient gene expression profiling database showed that ENO1 expression inversely correlates with overall survival. Protein expression analysis showed that ENO1 is expressed in pDC and MM cells; and importantly, that pDC-MM coculture further increases ENO1 expression in both MM cells and pDCs. Using our coculture models of patient autologous pDC-T-NK-MM cells, we examined whether targeting ENO1 can enhance anti-MM immunity. Biochemical inhibition of ENO1 with ENO1 inhibitor (ENO1i) activates pDCs, as well as increases pDC-induced MM-specific CD8+ CTL and NK cell activity against autologous tumor cells. Combination of ENO1i and anti-PD-L1 Ab or HDAC6i ACY-241 enhances autologous MM-specific CD8+ CTL activity. Our preclinical data therefore provide the basis for novel immune-based therapeutic approaches targeting ENO1, alone or in combination with anti-PD-L1 Ab or ACY241, to restore anti-MM immunity, enhance MM cytotoxicity, and improve patient outcome.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Biomarkers, Tumor/antagonists & inhibitors , Cell Communication/drug effects , DNA-Binding Proteins/antagonists & inhibitors , Dendritic Cells/drug effects , Multiple Myeloma/drug therapy , Phosphopyruvate Hydratase/antagonists & inhibitors , Tumor Suppressor Proteins/antagonists & inhibitors , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , B7-H1 Antigen/antagonists & inhibitors , B7-H1 Antigen/immunology , Biomarkers, Tumor/metabolism , Cell Communication/immunology , Cell Culture Techniques , Coculture Techniques , DNA-Binding Proteins/metabolism , Dendritic Cells/immunology , Dendritic Cells/metabolism , Gene Expression Profiling , Glycolysis/drug effects , Glycolysis/immunology , Histone Deacetylase 6/antagonists & inhibitors , Histone Deacetylase 6/metabolism , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylase Inhibitors/therapeutic use , Humans , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Lymphocyte Activation/drug effects , Multiple Myeloma/immunology , Multiple Myeloma/metabolism , Multiple Myeloma/mortality , Phosphopyruvate Hydratase/metabolism , Primary Cell Culture , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/metabolism , Tumor Cells, Cultured , Tumor Suppressor Proteins/metabolism
15.
Sci Rep ; 10(1): 5324, 2020 03 24.
Article in English | MEDLINE | ID: mdl-32210275

ABSTRACT

Ubiquitin specific peptidase 7 (USP7) is a deubiquitinating enzyme (DUB) that removes ubiquitin tags from specific protein substrates in order to alter their degradation rate and sub-cellular localization. USP7 has been proposed as a therapeutic target in several cancers because it has many reported substrates with a role in cancer progression, including FOXO4, MDM2, N-Myc, and PTEN. The multi-substrate nature of USP7, combined with the modest potency and selectivity of early generation USP7 inhibitors, has presented a challenge in defining predictors of response to USP7 and potential patient populations that would benefit most from USP7-targeted drugs. Here, we describe the structure-guided development of XL177A, which irreversibly inhibits USP7 with sub-nM potency and selectivity across the human proteome. Evaluation of the cellular effects of XL177A reveals that selective USP7 inhibition suppresses cancer cell growth predominantly through a p53-dependent mechanism: XL177A specifically upregulates p53 transcriptional targets transcriptome-wide, hotspot mutations in TP53 but not any other genes predict response to XL177A across a panel of ~500 cancer cell lines, and TP53 knockout rescues XL177A-mediated growth suppression of TP53 wild-type (WT) cells. Together, these findings suggest TP53 mutational status as a biomarker for response to USP7 inhibition. We find that Ewing sarcoma and malignant rhabdoid tumor (MRT), two pediatric cancers that are sensitive to other p53-dependent cytotoxic drugs, also display increased sensitivity to XL177A.


Subject(s)
Protease Inhibitors/pharmacology , Tumor Suppressor Protein p53/metabolism , Ubiquitin-Specific Peptidase 7/antagonists & inhibitors , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Humans , MCF-7 Cells , Protease Inhibitors/chemistry , Ubiquitin-Specific Peptidase 7/chemistry , Ubiquitin-Specific Peptidase 7/metabolism , Ubiquitination/drug effects
16.
Mol Cancer Ther ; 7(5): 1121-9, 2008 May.
Article in English | MEDLINE | ID: mdl-18445657

ABSTRACT

Clinical studies of patients with chronic myeloid leukemia revealed that a common pattern of response is a dramatic fall in the circulating population of blast cells, with a minimal or delayed decrease in marrow blasts, suggesting a protective environment. These observations suggest that a greater understanding of the interaction of stromal cells with leukemic cells is essential. Here, we present an in vivo system for monitoring relative tumor accumulation in leukemic mice and residual disease in leukemic mice treated with a tyrosine kinase inhibitor and an in vitro system for identifying integral factors involved in stromal-mediated cytoprotection. Using the in vivo model, we observed high tumor burden/residual disease in tissues characterized as significant sources of hematopoiesis-promoting stroma, with bone marrow stroma most frequently showing the highest accumulation of leukemia in untreated and nilotinib-treated mice as well as partial protection of leukemic cells from the inhibitory effects of nilotinib. These studies, which showed a pattern of leukemia distribution consistent with what is observed in imatinib- and nilotinib-treated chronic myeloid leukemia patients, were followed by a more in-depth analysis of stroma-leukemia cell interactions that lead to protection of leukemia cells from nilotinib-induced cytotoxicity. For the latter, we used the human BCR-ABL-positive cell line, KU812F, and the human bone marrow stroma cell line, HS-5, to more closely approximate the bone marrow-associated cytoprotection observed in drug-treated leukemia patients. This in vitro system helped to elucidate stromal-secreted viability factors that may play a role in stromal-mediated cytoprotection of tyrosine kinase inhibitor-treated leukemia cells.


Subject(s)
Antineoplastic Agents/toxicity , Fusion Proteins, bcr-abl/antagonists & inhibitors , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Protein Kinase Inhibitors/toxicity , Animals , Antineoplastic Agents/therapeutic use , Bone Marrow Cells/physiology , Cell Line, Tumor , Cell Proliferation/drug effects , Fusion Proteins, bcr-abl/metabolism , Humans , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/enzymology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Mice , Mice, Nude , Protein Kinase Inhibitors/metabolism , Pyrimidines/toxicity , Stromal Cells/physiology
17.
Leukemia ; 33(11): 2685-2694, 2019 11.
Article in English | MEDLINE | ID: mdl-30962579

ABSTRACT

Proteasome inhibition is an effective treatment for multiple myeloma (MM); however, targeting different components of the ubiquitin-proteasome system (UPS) remains elusive. Our RNA-interference studies identified proteasome-associated ubiquitin-receptor Rpn13 as a mediator of MM cell growth and survival. Here, we developed the first degrader of Rpn13, WL40, using a small-molecule-induced targeted protein degradation strategy to selectively degrade this component of the UPS. WL40 was synthesized by linking the Rpn13 covalent inhibitor RA190 with the cereblon (CRBN) binding ligand thalidomide. We show that WL40 binds to both Rpn13 and CRBN and triggers degradation of cellular Rpn13, and is therefore first-in-class in exploiting a covalent inhibitor for the development of degraders. Biochemical and cellular studies show that WL40-induced Rpn13 degradation is both CRBN E3 ligase- and Rpn13-dependent. Importantly, WL40 decreases viability in MM cell lines and patient MM cells, even those resistant to bortezomib. Mechanistically, WL40 interrupts Rpn13 function and activates caspase apoptotic cascade, ER stress response and p53/p21 signaling. In animal model studies, WL40 inhibits xenografted human MM cell growth and prolongs survival. Overall, our data show the development of the first UbR Rpn13 degrader with potent anti-MM activity, and provide proof of principle for the development of degraders targeting components of the UPS for therapeutic application.


Subject(s)
Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Intracellular Signaling Peptides and Proteins/metabolism , Multiple Myeloma/metabolism , Proteasome Inhibitors/pharmacology , Animals , Antineoplastic Agents/pharmacology , Apoptosis , Bortezomib/pharmacology , CRISPR-Cas Systems , Caspases/metabolism , Cell Line, Tumor , Cell Proliferation , Cell Survival , Dendritic Cells/cytology , Drug Evaluation, Preclinical , HCT116 Cells , Humans , Lenalidomide/pharmacology , Mice , Mice, SCID , Multiple Myeloma/therapy , Neoplasm Transplantation , Proteasome Endopeptidase Complex/metabolism , RNA Interference , Ubiquitin/chemistry
19.
Mol Cancer Ther ; 6(7): 1951-61, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17620426

ABSTRACT

Members of the inhibitor of apoptosis protein (IAP) family play a role in mediating apoptosis. Studies suggest that these proteins may be a viable target in leukemia because they have been found to be variably expressed in acute leukemias and are associated with chemosensitivity, chemoresistance, disease progression, remission, and patient survival. Another promising therapeutic target, FLT3, is mutated in about one third of acute myelogenous leukemia (AML) patients; promising results have recently been achieved in clinical trials investigating the effects of the protein tyrosine kinase inhibitor PKC412 on AML patients harboring mutations in the FLT3 protein. Of growing concern, however, is the development of drug resistance resulting from the emergence of point mutations in targeted tyrosine kinases used for treatment of acute leukemia patients. One approach to overriding resistance is to combine structurally unrelated inhibitors and/or inhibitors of different signaling pathways. The proapoptotic IAP inhibitor, LBW242, was shown in proliferation studies done in vitro to enhance the killing of PKC412-sensitive and PKC412-resistant cell lines expressing mutant FLT3 when combined with either PKC412 or standard cytotoxic agents (doxorubicin and Ara-c). In addition, in an in vivo imaging assay using bioluminescence as a measure of tumor burden, a total of 12 male NCr-nude mice were treated for 10 days with p.o. administration of vehicle, LBW242 (50 mg/kg/day), PKC412 (40 mg/kg/day), or a combination of LBW242 and PKC412; the lowest tumor burden was observed in the drug combination group. Finally, the combination of LBW242 and PKC412 was sufficient to override stromal-mediated viability signaling conferring resistance to PKC412.


Subject(s)
Antineoplastic Agents/pharmacology , Biomimetic Materials/pharmacology , Carrier Proteins , Leukemia/drug therapy , Mitochondrial Proteins , Mutant Proteins/metabolism , Oligopeptides/pharmacology , fms-Like Tyrosine Kinase 3/metabolism , Animals , Antineoplastic Agents/chemistry , Apoptosis/drug effects , Apoptosis Regulatory Proteins , Cell Line, Tumor , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Drug Resistance, Neoplasm/drug effects , Drug Synergism , Humans , Inhibitor of Apoptosis Proteins/metabolism , Mice , Mutant Proteins/genetics , Oligopeptides/chemistry , Staurosporine/analogs & derivatives , Staurosporine/pharmacology , Stromal Cells/drug effects , fms-Like Tyrosine Kinase 3/genetics
20.
Clin Cancer Res ; 23(15): 4280-4289, 2017 Aug 01.
Article in English | MEDLINE | ID: mdl-28270494

ABSTRACT

Purpose: The ubiquitin proteasome pathway is a validated therapeutic target in multiple myeloma. Deubiquitylating enzyme USP1 participates in DNA damage response and cellular differentiation pathways. To date, the role of USP1 in multiple myeloma biology is not defined. In the present study, we investigated the functional significance of USP1 in multiple myeloma using genetic and biochemical approaches.Experimental Design: To investigate the role of USP1 in myeloma, we utilized USP1 inhibitor SJB3-019A (SJB) for studies in myeloma cell lines and patient multiple myeloma cells.Results: USP1-siRNA knockdown decreases multiple myeloma cell viability. USP1 inhibitor SJB selectively blocks USP1 enzymatic activity without blocking other DUBs. SJB also decreases the viability of multiple myeloma cell lines and patient tumor cells, inhibits bone marrow plasmacytoid dendritic cell-induced multiple myeloma cell growth, and overcomes bortezomib resistance. SJB triggers apoptosis in multiple myeloma cells via activation of caspase-3, caspase-8, and caspase-9. Moreover, SJB degrades USP1 and downstream inhibitor of DNA-binding proteins as well as inhibits DNA repair via blockade of Fanconi anemia pathway and homologous recombination. SJB also downregulates multiple myeloma stem cell renewal/survival-associated proteins Notch-1, Notch-2, SOX-4, and SOX-2. Moreover, SJB induced generation of more mature and differentiated plasma cells. Combination of SJB and HDACi ACY-1215, bortezomib, lenalidomide, or pomalidomide triggers synergistic cytotoxicity.Conclusions: Our preclinical studies provide the framework for clinical evaluation of USP1 inhibitors, alone or in combination, as a potential novel multiple myeloma therapy. Clin Cancer Res; 23(15); 4280-9. ©2017 AACR.


Subject(s)
DNA-Binding Proteins/genetics , Drug Synergism , Multiple Myeloma/drug therapy , Neoplasm Proteins/genetics , Ubiquitin-Specific Proteases/genetics , Apoptosis/drug effects , Bortezomib/administration & dosage , Cell Line, Tumor , Cell Survival/drug effects , DNA Damage/drug effects , DNA Repair/drug effects , DNA-Binding Proteins/drug effects , Drug Resistance, Neoplasm/genetics , Gene Expression Regulation, Neoplastic/drug effects , Humans , Lenalidomide , Multiple Myeloma/genetics , Multiple Myeloma/pathology , Neoplasm Proteins/drug effects , Thalidomide/administration & dosage , Thalidomide/analogs & derivatives , Ubiquitin-Specific Proteases/antagonists & inhibitors , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL