Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
1.
Blood ; 119(15): e131-8, 2012 Apr 12.
Article in English | MEDLINE | ID: mdl-22289890

ABSTRACT

Conventional assays evaluating antitumor activity of immune effector cells have limitations that preclude their high-throughput application. We adapted the recently developed Compartment-Specific Bioluminescence Imaging (CS-BLI) technique to perform high-throughput quantification of innate antitumor activity and to show how pharmacologic agents (eg, lenalidomide, pomalidomide, bortezomib, and dexamethasone) and autologous BM stromal cells modulate that activity. CS-BLI-based screening allowed us to identify agents that enhance or inhibit innate antitumor cytotoxicity. Specifically, we identified compounds that stimulate immune effector cells against some tumor targets but suppressed their activity against other tumor cells. CS-BLI offers rapid, simplified, and specific evaluation of multiple conditions, including drug treatments and/or cocultures with stromal cells and highlights that immunomodulatory pharmacologic responses can be heterogeneous across different types of tumor cells. This study provides a framework to identify novel immunomodulatory agents and to prioritize compounds for clinical development on the basis of their effect on antitumor immunity.


Subject(s)
High-Throughput Screening Assays/methods , Immunity, Innate/physiology , Luminescent Measurements/methods , Neoplasms/diagnosis , Neoplasms/immunology , Animals , CD56 Antigen/metabolism , Cell Compartmentation/genetics , Cell Compartmentation/immunology , Cell Compartmentation/physiology , Cell Survival , Diagnostic Imaging/instrumentation , Diagnostic Imaging/methods , High-Throughput Screening Assays/instrumentation , Humans , Immunotherapy/methods , K562 Cells , Leukocytes, Mononuclear/metabolism , Leukocytes, Mononuclear/pathology , Luminescent Measurements/instrumentation , Neoplasms/pathology , Neoplasms/therapy , Substrate Specificity/immunology , Tumor Cells, Cultured
2.
Blood ; 116(4): 593-602, 2010 Jul 29.
Article in English | MEDLINE | ID: mdl-20382846

ABSTRACT

Using a combination high-throughput screening technology, multiple classes of drugs and targeted agents were identified that synergize with dexamethasone (Dex) in multiple myeloma (MM) cells. Performing combination screening with these enhancers, we discovered an unexpected synergistic interaction between adenosine receptor agonists and phosphodiesterase (PDE) inhibitors that displays substantial activity in a panel of MM and diffuse large B-cell lymphoma (DLBCL) cell lines and tumor cells from MM patients. We have used selective adenosine receptor agonists, antagonists, and PDE inhibitors as well as small interfering RNAs targeting specific molecular isoforms of these proteins to dissect the molecular mechanism of this synergy. The adenosine A2A receptor and PDE2, 3, 4, and 7 are important for activity. Drug combinations induce cyclic AMP (cAMP) accumulation and up-regulate PDE4B. We also observe rigorous mathematical synergy in 3-way combinations containing A2A agonists, PDE inhibitors, and Dex at multiple concentrations and ratios. Taken together, these data suggest that A2A agonist/PDE inhibitor combinations may be attractive as an adjunctive to clinical glucocorticoid containing regiments for patients with MM or DLBCL and confer benefit in both glucocorticoid-sensitive and -resistant populations.


Subject(s)
Adenosine A2 Receptor Agonists , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Drug Screening Assays, Antitumor/methods , Hematologic Neoplasms/drug therapy , Phosphodiesterase Inhibitors/administration & dosage , B-Lymphocytes/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Dexamethasone/administration & dosage , Dexamethasone/pharmacology , Drug Delivery Systems , Drug Synergism , Glucocorticoids/administration & dosage , Glucocorticoids/pharmacology , Hematologic Neoplasms/metabolism , Hematologic Neoplasms/pathology , High-Throughput Screening Assays/methods , Humans , Lymphoma, Large B-Cell, Diffuse/drug therapy , Lymphoma, Large B-Cell, Diffuse/pathology , Multiple Myeloma/drug therapy , Multiple Myeloma/pathology , Phosphodiesterase Inhibitors/isolation & purification , Phosphodiesterase Inhibitors/pharmacology , Validation Studies as Topic
3.
J Nat Prod ; 75(4): 793-7, 2012 Apr 27.
Article in English | MEDLINE | ID: mdl-22458669

ABSTRACT

CR1642D, an endophytic isolate of Penicillium sp. collected from a Costa Rican rainforest, was identified through a high-throughput approach to identify natural products with enhanced antitumor activity in the context of tumor-stromal interactions. Bioassay-guided separation led to the identification of five xanthones (1-5) from CR1642D. The structures of the xanthone dimer penexanthone A (1) and monomer penexanthone B (2) were elucidated on the basis of spectroscopic analyses, including 2D NMR experiments. All of the compounds were tested against a panel of tumor cell lines in the presence and absence of bone marrow stromal cells. Compound 3 was the most active, with IC(50) values of 1-17 µM, and its activity was enhanced 2-fold against tumor cell line RPMI8226 in the presence of stromal cells (IC(50) 1.2 µM, but 2.4 µM without stromal cells).


Subject(s)
Penicillium/chemistry , Xanthones/isolation & purification , Xanthones/pharmacology , Costa Rica , Humans , Molecular Sequence Data , Molecular Structure , Nuclear Magnetic Resonance, Biomolecular , Stromal Cells/drug effects , Xanthones/chemistry
4.
Br J Haematol ; 152(4): 420-32, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21223249

ABSTRACT

Cell cycle regulators, such as cyclin-dependent kinases (CDKs), are appealing targets for multiple myeloma (MM) therapy given the increased proliferative rates of tumour cells in advanced versus early stages of MM. We hypothesized that a multi-targeted CDK inhibitor with a different spectrum of activity compared to existing CDK inhibitors could trigger distinct molecular sequelae with therapeutic implications for MM. We therefore studied the small molecule heterocyclic compound NVP-LCQ195/AT9311 (LCQ195), which inhibits CDK1, CDK2 and CDK5, as well as CDK3 and CDK9. LCQ195 induced cell cycle arrest and eventual apoptotic cell death of MM cells, even at sub-µmol/l concentrations, spared non-malignant cells, and overcame the protection conferred to MM cells by stroma or cytokines of the bone marrow milieu. In MM cells, LCQ195 triggered decreased amplitude of transcriptional signatures associated with oncogenesis, drug resistance and stem cell renewal, including signatures of activation of key transcription factors for MM cells e.g. myc, HIF-1α, IRF4. Bortezomib-treated MM patients whose tumours had high baseline expression of genes suppressed by LCQ195 had significantly shorter progression-free and overall survival than those with low levels of these transcripts in their MM cells. These observations provide insight into the biological relevance of multi-targeted CDK inhibition in MM.


Subject(s)
Antineoplastic Agents/pharmacology , Cyclin-Dependent Kinases/antagonists & inhibitors , Multiple Myeloma/pathology , Apoptosis/drug effects , Boronic Acids/therapeutic use , Bortezomib , Cell Cycle/drug effects , Cell Survival/drug effects , Coculture Techniques , Cyclin-Dependent Kinases/metabolism , Cytokines/antagonists & inhibitors , Cytokines/pharmacology , Dose-Response Relationship, Drug , Down-Regulation/drug effects , Drug Combinations , Drug Interactions , Drug Screening Assays, Antitumor , Gene Expression Regulation, Neoplastic/drug effects , Humans , Multiple Myeloma/drug therapy , Multiple Myeloma/enzymology , Multiple Myeloma/genetics , Pyrazines/therapeutic use , Signal Transduction/drug effects , Stromal Cells/physiology , Survival Analysis , Transcription, Genetic , Treatment Outcome , Tumor Cells, Cultured
5.
Haematologica ; 96(8): 1170-9, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21712538

ABSTRACT

BACKGROUND: Isothiocyanates, a family of phytochemicals found in cruciferous vegetables, have cytotoxic effects against several types of tumor cells. Multiple myeloma is a fatal disease characterized by clonal proliferation of plasma cells in the bone marrow. The growing body of preclinical information on the anti-cancer activity of isothiocyanates led us to investigate their anti-myeloma properties. DESIGN AND METHODS: We evaluated the anti-myeloma activity of the isothiocyanates, sulforaphane and phenethyl isothiocyanate, on a panel of human myeloma cell lines as well as primary myeloma tumor cells. Cell viability, apoptosis, cell cycle alterations and cell proliferation were then analyzed in vitro and in a xenograft mouse model in vivo. The molecular sequelae of isothiocyanate treatment in multiple myeloma cells were evaluated by multiplex analyses using bead arrays and western blotting. RESULTS: We observed that sulforaphane and phenylethyl isothiocyanate have activity against myeloma cell lines and patients' myeloma cells both in vitro and in vivo using a myeloma xenograft mouse model. Isothiocyanates induced apoptotic death of myeloma cells; depletion of mitochondrial membrane potential; cleavage of PARP and caspases-3 and -9; as well as down-regulation of anti-apoptotic proteins including Mcl-1, X-IAP, c-IAP and survivin. Isothiocyanates induced G(2)/M cell cycle arrest accompanied by mitotic phosphorylation of histone H3. Multiplex analysis of phosphorylation of diverse components of signaling cascades revealed changes in MAPK activation; increased phosphorylation of c-jun and HSP27; as well as changes in the phosphorylation of Akt, and GSK3α/ß and p53. Isothiocyanates suppressed proliferation of myeloma cells alone and when co-cultured with HS-5 stromal cells. Sulforaphane and phenylethyl isothiocyanate enhanced the in vitro anti-myeloma activity of several conventional and novel therapies used in multiple myeloma. CONCLUSIONS: Our study shows that isothiocyanates have potent anti-myeloma activities and may enhance the activity of other anti-multiple myeloma agents. These results indicate that isothiocyanates may have therapeutic potential in multiple myeloma and provide the preclinical framework for future clinical studies of isothiocyanates in multiple myeloma.


Subject(s)
Antineoplastic Agents/pharmacology , Isothiocyanates/pharmacology , Multiple Myeloma/drug therapy , Signal Transduction/drug effects , Thiocyanates/pharmacology , Animals , Antineoplastic Agents/therapeutic use , Antineoplastic Agents/toxicity , Apoptosis/drug effects , Cell Cycle/drug effects , Cell Line , Cell Proliferation/drug effects , Cell Survival/drug effects , Disease Models, Animal , Dose-Response Relationship, Drug , Humans , Isothiocyanates/therapeutic use , Isothiocyanates/toxicity , Mice , Mice, SCID , Multiple Myeloma/metabolism , Stromal Cells/drug effects , Sulfoxides , Thiocyanates/therapeutic use , Thiocyanates/toxicity , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
6.
J Immunol ; 183(12): 7799-809, 2009 Dec 15.
Article in English | MEDLINE | ID: mdl-19933853

ABSTRACT

We investigated the roles of specific subsets of donor APCs purified from bone marrow in donor T cell activation and graft-vs-leukemia (GvL) activity in murine models of hemopoietic stem cell transplantation. Lineage(-)CD11c(+) APC precursors were separated from donor bone marrow based on expression of CD11b. Transplanting lineage(-)CD11c(+)CD11b(-) APC (CD11b(-) APC) in combination with c-kit(+)Sca-1(+)lineage(-) hemopoietic stem cells (HSC) and congenic donor T cells led to increased donor CD4(+) and CD8(+) T cell proliferation and higher donor T cell chimerism than with transplanting grafts containing HSC, T cells, and lineage(-)CD11c(+)CD11b(+) APCs (CD11b(+) APC), or grafts containing only HSC and T cells. Transplanting CD11b(-) APCs induced Th1/type 1 cytotoxic T lymphocyte donor T cell immune polarization and enhanced GvL activity of donor T cells without increased graft-vs-host disease in both MHC- and minor histocompatibility Ag-mismatched murine hemopoietic stem cell transplantation models, whereas CD11b(+) APCs led to Th2/type 2 cytotoxic T lymphocyte donor T cell immune polarization. Donor CD11b(-) APCs were plasmacytoid dendritic cell progenitors (>90% CD317; PDCA-1(+)) and up-regulated CD80, CD86, and IL-12 during alloantigen presentation, whereas CD11b(+) APCs expressed Gr-1 and up-regulated expression of programmed death ligands-1 and 2 after activation. These results are the first to show that manipulation of the content of donor APCs in allogeneic HSC grafts can regulate donor T cell immunity and enhance GvL without increasing graft-vs-host disease activity.


Subject(s)
Antigen-Presenting Cells/immunology , Bone Marrow Cells/immunology , Cell Polarity/immunology , Graft vs Leukemia Effect/immunology , Hematopoietic Stem Cell Transplantation , Lymphocyte Activation/immunology , T-Lymphocyte Subsets/immunology , Animals , Antigen-Presenting Cells/metabolism , Antigen-Presenting Cells/transplantation , Bone Marrow Cells/metabolism , Cell Line, Tumor , Graft Enhancement, Immunologic/methods , Hematopoietic Stem Cell Transplantation/methods , Isoantigens/administration & dosage , Isoantigens/biosynthesis , Isoantigens/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Transgenic , T-Lymphocyte Subsets/metabolism , T-Lymphocyte Subsets/transplantation
7.
Semin Hematol ; 46(2): 166-75, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19389500

ABSTRACT

The successful clinical development of thalidomide, bortezomib, and lenalidomide not only transformed the therapeutic management of multiple myeloma (MM) but also catalyzed a renewed interest in the development of additional classes of novel agents for this disease. This review focuses on a series of new therapeutics that have shown promising preclinical results, as well as encouraging safety profiles and early evidence of anti-MM activity in clinical studies, either alone or in combination with other, conventional or novel, anti-MM treatments. These agents include second-generation proteasome inhibitors and immunomodulatory agents, as well as members of other therapeutic classes, such as histone deacetylase inhibitors (HDAC), heat shock protein 90 (Hsp90) inhibitors, and the alkylphospholipid Akt inhibitor perifosine.


Subject(s)
Antineoplastic Agents/therapeutic use , Enzyme Inhibitors/therapeutic use , Multiple Myeloma/drug therapy , Animals , Boronic Acids/therapeutic use , Bortezomib , Drug Screening Assays, Antitumor , HSP90 Heat-Shock Proteins/antagonists & inhibitors , HSP90 Heat-Shock Proteins/metabolism , Histone Deacetylase Inhibitors , Histone Deacetylases/metabolism , Humans , Immunologic Factors/therapeutic use , Multiple Myeloma/metabolism , Phosphorylcholine/analogs & derivatives , Phosphorylcholine/therapeutic use , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/metabolism , Pyrazines/therapeutic use
8.
Br J Haematol ; 147(5): 672-6, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19751238

ABSTRACT

This study characterized the preclinical anti-myeloma activity of VE465, a low molecular weight pan-Aurora kinase inhibitor. After 96-h drug exposure, several multiple myeloma (MM) cell lines were more sensitive to VE465 compared to non-malignant cells. The anti-MM activity of VE465 was maintained in the presence of interleukin-6 and, interestingly, enhanced by co-culture with stromal cells. However, primary MM cells were less responsive than cell lines. Combinations with dexamethasone (Dex), doxorubicin (Doxo) and bortezomib showed no antagonism. Our study highlights the potential role of the tumour microenvironment in modulating the activity of this drug class.


Subject(s)
Antineoplastic Agents/pharmacology , Multiple Myeloma/pathology , Piperazines/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Apoptosis/drug effects , Aurora Kinases , Cell Communication/drug effects , Cell Cycle/drug effects , Coculture Techniques , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor/methods , Hematopoietic Stem Cells/cytology , Humans , Tumor Cells, Cultured
9.
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
11.
Mol Cancer Ther ; 6(3): 1070-8, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17363500

ABSTRACT

B-Raf is an important mediator of cell proliferation and survival signals transduced via the Ras-Raf-MEK-ERK cascade. BRAF mutations have been detected in several tumors, including papillary thyroid carcinoma, but the precise role of B-Raf as a therapeutic target for thyroid carcinoma is still under investigation. We analyzed a panel of 93 specimens and 14 thyroid carcinoma cell lines for the presence of BRAF mutations and activation of the mitogen-activated protein/ERK kinase (MEK)/extracellular signal-regulated kinase (ERK) pathway. We also compared the effect of a B-Raf small inhibitory RNA construct and the B-Raf kinase inhibitor AAL881 on both B-Raf wild-type and mutant thyroid carcinoma cell lines. We found a high prevalence of the T1799A (V600E) mutation in papillary and anaplastic carcinoma specimens and cell lines. There was no difference in patient age, B-Raf expression, Ki67 immunostaining, or clinical stage at presentation between wild-type and BRAF(V600E) specimens. Immunodetection of phosphorylated and total forms of MEK and ERK revealed no difference in their phosphorylation between wild-type and BRAF(V600E) patient specimens or cell lines. Furthermore, a small inhibitory RNA construct targeting the expression of both wild-type B-Raf and B-Raf(V600E) induced a comparable reduction of viability in both wild-type and BRAF(V600E) mutant cancer cells. Interestingly, AAL881 inhibited MEK and ERK phosphorylation and induced apoptosis preferentially in BRAF(V600E)-harboring cells than wild-type ones, possibly because of better inhibitory activity against B-Raf(V600E). We conclude that B-Raf is important for the pathophysiology of thyroid carcinomas irrespective of mutational status. Small molecule inhibitors that selectively target B-Raf(V600E) may provide clinical benefit for patients with thyroid cancer.


Subject(s)
Adenocarcinoma, Follicular/genetics , Carcinoma, Papillary/genetics , Isoquinolines/pharmacology , Mutation/genetics , Proto-Oncogene Proteins B-raf/genetics , Thyroid Neoplasms/genetics , Adenocarcinoma, Follicular/pathology , Adenocarcinoma, Follicular/prevention & control , Adult , Aged , Apoptosis/drug effects , Carcinoma/genetics , Carcinoma/pathology , Carcinoma/prevention & control , Carcinoma, Papillary/pathology , Carcinoma, Papillary/prevention & control , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/pathology , Carcinoma, Squamous Cell/prevention & control , Cell Proliferation/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Humans , Male , Middle Aged , Mitogen-Activated Protein Kinases/metabolism , Phosphorylation/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects , Thyroid Neoplasms/pathology , Thyroid Neoplasms/prevention & control , Tumor Cells, Cultured/drug effects
12.
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
13.
J Clin Endocrinol Metab ; 92(12): 4845-52, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17848408

ABSTRACT

CONTEXT: The Bcl-2 family of proteins regulates apoptosis in various models and may represent a promising therapeutic target in human malignancies. OBJECTIVE/METHODS: We evaluated the sensitivity of thyroid carcinoma cell lines (two papillary, one follicular, two anaplastic, three medullary) in vitro to BH3I-1 and BH3I-2', two cell-permeable inhibitors of the Bcl-2 homology (BH)-3 domain-mediated interaction between proapoptotic and antiapoptotic Bcl-2 family members. The thyroid carcinoma cell line FRO was stably transfected with cDNA for Bcl-2 or constitutively active Akt and evaluated for sensitivity to BH3-domain inhibition. RESULTS: BH3-domain inhibition disrupted the mitochondrial membrane potential in thyroid carcinoma cells, induced caspase-dependent apoptosis, and potently sensitized them to sublethal concentrations of doxorubicin and the proteasome inhibitor bortezomib (Velcade). Overexpression of constitutively active Akt suppressed BH3I-1-induced cell death. Bcl-2-overexpressing FRO cells were more resistant to conventional chemotherapeutic agents (such as doxorubicin) but significantly more sensitive to BH3I-1 than control cells and were found to overexpress caspase-9, caspase-8, Bmf, Bok, and Bik transcripts and express less A1, BRaf, and FLIP transcripts. CONCLUSIONS: Bcl-2 expression protects thyroid carcinomas against chemotherapy-induced apoptosis. Nevertheless, overexpression of Bcl-2 may result in "oncogene addiction" of the cancer cell, which can be exploited by using BH3-domain inhibitors alone or in combination with other agents, including conventional chemotherapeutics (such as doxorubicin) or novel targeted therapies (such as the proteasome inhibitor bortezomib), for the treatment of aggressive thyroid cancer, including the medullary and anaplastic types.


Subject(s)
Carcinoma, Medullary/genetics , Carcinoma/genetics , Genes, bcl-2/genetics , Thyroid Neoplasms/genetics , Antibiotics, Antineoplastic/pharmacology , Antineoplastic Agents/pharmacology , BH3 Interacting Domain Death Agonist Protein/genetics , Blotting, Western , Boronic Acids/pharmacology , Bortezomib , Cell Line, Tumor , Colorimetry , Doxorubicin/pharmacology , Humans , Membrane Potentials/physiology , Oncogene Protein v-akt/genetics , Protease Inhibitors/pharmacology , Pyrazines/pharmacology , RNA, Neoplasm/biosynthesis , RNA, Neoplasm/genetics , Tetrazolium Salts , Thiazoles , Transcription, Genetic
14.
Hematol Oncol Clin North Am ; 21(6): 1007-34, vii-viii, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17996586

ABSTRACT

Multiple myeloma (MM) is viewed as a prototypic disease state for the study of how neoplastic cells interact with their local bone marrow (BM) microenvironment. This interaction reflects not only the osteo-tropic clinical behavior of MM and the clinical impact of the lytic bone lesions caused by its tumor cells but also underlines the broadly accepted notion that nonneoplastic cells of the BM can attenuate the activity of cytotoxic chemotherapy and glucocorticoids. This article summarizes the recent progress in characterization, at the molecular and cellular levels, of how the BM milieu interacts with MM cells and modifies their biologic behavior.


Subject(s)
Bone Marrow/physiology , Cell Communication/physiology , Multiple Myeloma/physiopathology , Humans , Multiple Myeloma/therapy , Osteoclasts/physiology , Stromal Cells/physiology
15.
Hum Gene Ther ; 17(8): 798-806, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16942440

ABSTRACT

Combining chemotherapy and immunotherapy is problematic because chemotherapy can ablate the immune responses initiated by modulators of the immune system. We hypothesized that protection of immunocompetent cells from the toxic effects of chemotherapy, using drug resistance gene therapy strategies, would allow the combined use of chemotherapy and immunotherapy. In wild-type mice, the antitumor effectiveness of an immunotherapy regimen employing an agonistic anti-CD137 antibody is diminished with escalating doses of the antifolate trimetrexate (TMTX). Using retroviral gene transfer of a mutant form of dihydrofolate reductase (L22Y-DHFR), hematopoietic stem cells were genetically engineered to withstand the toxic effects of TMTX. Mice transplanted with L22Y-DHFR-modified bone marrow were then challenged with AG104 sarcoma cells and treated with TMTX only, anti-CD137 only, or a combination of chemotherapy and immunotherapy. Although tumor burden was transiently decreased during TMTX administration, no mice treated with TMTX alone survived the tumor challenge, whereas approximately 40% of transplanted mice treated with anti-CD137 alone survived. However, 100% of mice survived with complete tumor regression after transplantation with L22Y-DHFR-transduced bone marrow followed by combined treatment with TMTX and anti-CD137. In addition, adoptive transfer of splenocytes from cured mice extended the survival of tumor- bearing animals by approximately 3 weeks compared with controls. Therefore, protection of the hematopoietic system can allow for the combined administration of chemotherapy and immunotherapy, which results in complete tumor clearance.


Subject(s)
Antigens, CD , Bone Marrow Transplantation , Drug Resistance/genetics , Genetic Therapy , Hematopoietic Stem Cells/enzymology , Neoplasms, Experimental/therapy , Point Mutation , Receptors, Nerve Growth Factor , Receptors, Tumor Necrosis Factor , Sarcoma/therapy , Tetrahydrofolate Dehydrogenase/genetics , Adoptive Transfer/methods , Animals , Antibodies/immunology , Antibodies/pharmacology , Antigens, CD/immunology , Antimetabolites, Antineoplastic/administration & dosage , Combined Modality Therapy/methods , Drug Resistance/drug effects , Drug Resistance/immunology , Genetic Therapy/methods , Humans , Mice , Neoplasms, Experimental/enzymology , Neoplasms, Experimental/genetics , Neoplasms, Experimental/immunology , Receptors, Nerve Growth Factor/immunology , Receptors, Tumor Necrosis Factor/immunology , Sarcoma/enzymology , Sarcoma/genetics , Sarcoma/immunology , Tetrahydrofolate Dehydrogenase/immunology , Transplantation, Homologous , Trimetrexate/pharmacology , Tumor Necrosis Factor Receptor Superfamily, Member 9
16.
ACS Chem Biol ; 9(5): 1086-91, 2014 May 16.
Article in English | MEDLINE | ID: mdl-24556163

ABSTRACT

BTK is a member of the TEC family of non-receptor tyrosine kinases whose deregulation has been implicated in a variety of B-cell-related diseases. We have used structure-based drug design in conjunction with kinome profiling and cellular assays to develop a potent, selective, and irreversible BTK kinase inhibitor, QL47, which covalently modifies Cys481. QL47 inhibits BTK kinase activity with an IC50 of 7 nM, inhibits autophosphorylation of BTK on Tyr223 in cells with an EC50 of 475 nM, and inhibits phosphorylation of a downstream effector PLCγ2 (Tyr759) with an EC50 of 318 nM. In Ramos cells QL47 induces a G1 cell cycle arrest that is associated with pronounced degradation of BTK protein. QL47 inhibits the proliferation of B-cell lymphoma cancer cell lines at submicromolar concentrations.


Subject(s)
Lymphoma, B-Cell/drug therapy , Lymphoma, B-Cell/enzymology , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Protein-Tyrosine Kinases/antagonists & inhibitors , Protein-Tyrosine Kinases/metabolism , Agammaglobulinaemia Tyrosine Kinase , B-Lymphocytes/drug effects , B-Lymphocytes/enzymology , B-Lymphocytes/pathology , Cell Line, Tumor , Drug Discovery , Humans , Lymphoma, B-Cell/pathology , Molecular Docking Simulation , Phosphorylation/drug effects , Protein-Tyrosine Kinases/chemistry , Signal Transduction/drug effects
17.
Nat Rev Drug Discov ; 12(3): 217-28, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23449307

ABSTRACT

The role of stromal cells and the tumour microenvironment in general in modulating tumour sensitivity is increasingly becoming a key consideration for the development of active anticancer therapeutics. Here, we discuss how these tumour-stromal interactions affect tumour cell signalling, survival, proliferation and drug sensitivity. Particular emphasis is placed on the ability of stromal cells to confer - to tumour cells - resistance or sensitization to different classes of therapeutics, depending on the specific microenvironmental context. The mechanistic understanding of these microenvironmental interactions can influence the evaluation and selection of candidate agents for various cancers, in both the primary site as well as the metastatic setting. Progress in in vitro screening platforms as well as orthotopic and 'orthometastatic' xenograft mouse models has enabled comprehensive characterization of the impact of the tumour microenvironment on therapeutic efficacy. These recent advances can hopefully bridge the gap between preclinical studies and clinical trials of anticancer agents.


Subject(s)
Antineoplastic Agents/pharmacology , Neoplasms/drug therapy , Neoplasms/physiopathology , Stromal Cells/drug effects , Stromal Cells/physiology , Animals , Drug Discovery , Drug Evaluation, Preclinical , Drug Resistance, Neoplasm , Drug Screening Assays, Antitumor , Humans , Precision Medicine
18.
Clin Cancer Res ; 19(20): 5591-601, 2013 Oct 15.
Article in English | MEDLINE | ID: mdl-24004671

ABSTRACT

PURPOSE: Cellular immunotherapy frequently fails to induce sustained remissions in patients with multiple myeloma, indicating the ability of multiple myeloma cells to evade cellular immunity. Toward a better understanding and effective therapeutic modulation of multiple myeloma immune evasion mechanisms, we here investigated the role of the tumor microenvironment in rendering multiple myeloma cells resistant to the cytotoxic machinery of T cells. EXPERIMENTAL DESIGN: Using a compartment-specific, bioluminescence imaging-based assay system, we measured the lysis of luciferase-transduced multiple myeloma cells by CD4(+) or CD8(+) CTLs in the presence versus absence of adherent accessory cells of the bone marrow microenvironment. We simultaneously determined the level of CTL activation by measuring the granzyme B release in culture supernatants. RESULTS: Bone marrow stromal cells from patients with multiple myeloma and healthy individuals, as well as vascular endothelial cells, significantly inhibited the lysis of multiple myeloma cells in a cell-cell contact-dependent manner and without substantial T-cell suppression, thus showing the induction of a cell adhesion-mediated immune resistance (CAM-IR) against CTL lysis. Further analyses revealed that adhesion to accessory cells downregulated Fas and upregulated the caspase-3 inhibitor survivin in multiple myeloma cells. Reconstitution of Fas expression with bortezomib enhanced the CTL-mediated lysis of multiple myeloma cells. Repressing survivin with the small-molecule YM155 synergized with CTLs and abrogated CAM-IR in vitro and in vivo. CONCLUSION: These results reveal the cell adhesion-mediated induction of apoptosis resistance as a novel immune escape mechanism and provide a rationale to improve the efficacy of cellular therapies by pharmacologic modulation of CAM-IR.


Subject(s)
Cytotoxicity, Immunologic , Multiple Myeloma/immunology , Multiple Myeloma/pathology , T-Lymphocytes, Cytotoxic/immunology , Tumor Microenvironment/immunology , Animals , Antineoplastic Agents/therapeutic use , Cell Adhesion/immunology , Cell Communication , Cell Line, Tumor , Combined Modality Therapy , Disease Models, Animal , Humans , Imidazoles/therapeutic use , Immunomodulation , Immunotherapy, Adoptive , Mice , Multiple Myeloma/metabolism , Multiple Myeloma/therapy , Naphthoquinones/therapeutic use , Xenograft Model Antitumor Assays , fas Receptor/metabolism
19.
Oncoimmunology ; 1(8): 1406-1408, 2012 Nov 01.
Article in English | MEDLINE | ID: mdl-23243609

ABSTRACT

The clinical success of immunomodulatory thalidomide derivatives has renewed the general interest in immunomodulatory anticancer compounds and prompted us to develop a high-throughput system to quantify immune effector-cell activity. We documented that the interaction between cancer cells, their stroma, anticancer agents and cells from the innate system are critical for determining the response of tumors to immunomodulatory strategies.

20.
J Breast Cancer ; 15(3): 273-82, 2012 Sep.
Article in English | MEDLINE | ID: mdl-23091539

ABSTRACT

PURPOSE: Tumor-specific delivery of tumor necrosis factor-related apoptosis-inducing ligand (TRAIL), an apoptosis-inducing peptide, at effective doses remains challenging. Herein we demonstrate the utility of a scaffold-based delivery system for sustained therapeutic cell release that capitalizes on the tumor-homing properties of mesenchymal stem cells (MSCs) and their ability to express genetically-introduced therapeutic genes. METHODS: Implants were formed from porous, biocompatible silk scaffolds seeded with full length TRAIL-expressing MSCs (FLT-MSCs). under a doxycycline inducible promoter. In vitro studies with FLT-MSCs demonstrated TRAIL expression and antitumor effects on breast cancer cells. Next, FLT-MSCs were administered to mice using three administration routes (mammary fat pad co-injections, tail vein injections, and subcutaneous implantation on scaffolds). RESULTS: In vitro cell-specific bioluminescent imaging measured tumor cell specific growth in the presence of stromal cells and demonstrated FLT-MSC inhibition of breast cancer growth. FLT-MSC implants successfully decreased bone and lung metastasis, whereas liver metastasis decreased only with tail vein and co-injection administration routes. Average tumor burden was decreased when doxycycline was used to induce TRAIL expression for co-injection and scaffold groups, as compared to controls with no induced TRAIL expression. CONCLUSION: This implant-based therapeutic delivery system is an effective and completely novel method of anticancer therapy and holds great potential for clinical applications.

SELECTION OF CITATIONS
SEARCH DETAIL