Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
1.
J Pharmacol Exp Ther ; 388(3): 774-787, 2024 02 15.
Article in English | MEDLINE | ID: mdl-38135509

ABSTRACT

This study provides a unique translational research opportunity to help both humans and dogs diagnosed with diseases that carry dismal prognoses in both species: histiocytic sarcoma (HS), hemangiosarcoma (HSA), and disseminated mastocytosis/mast cell tumor (MCT). Although exceedingly rare in humans, these so called "orphan diseases" are relatively more common in dogs. For these and other more commonplace cancers like lymphoma (Lym), dogs are an excellent translational model for human disease due to remarkably similar disease biology. In this study, assays were performed to assess the therapeutic potential of parthenolide (PTL), a known canonical nuclear factor kappa B (NF-κB) signaling inhibitor with additional mechanisms of antineoplastic activity, including alteration of cellular reduction-oxidation balance. Canine cell lines and primary cells are sensitive to PTL and undergo dose-dependent apoptosis after exposure to drug. PTL exposure also leads to glutathione depletion, reactive oxygen species generation, and NF-κB inhibition in canine cells. Standard-of-care therapeutics broadly synergize with PTL. In two canine HS cell lines, expression of NF-κB pathway signaling partners is downregulated with PTL therapy. Preliminary data suggest that PTL inhibits NF-κB activity of cells and extends survival time in a mouse model of disseminated canine HS. These data support further investigation of compounds that can antagonize canonical NF-κB pathway signaling in these cancers and pave the way for clinical trials of PTL in affected dogs. As dogs are an excellent natural disease model for these cancers, these data will ultimately improve our understanding of their human disease counterparts and hopefully improve care for both species. SIGNIFICANCE STATEMENT: Disseminated neoplasms in human and canine cancers are challenging to treat, and novel therapeutic approaches are needed to improve outcomes. Parthenolide is a promising treatment for histiocytic sarcoma, hemangiosarcoma, and mast cell neoplasia.


Subject(s)
Hemangiosarcoma , Histiocytic Sarcoma , Sesquiterpenes , Mice , Humans , Animals , Dogs , NF-kappa B/metabolism , Cell Line, Tumor , Histiocytic Sarcoma/drug therapy , Hemangiosarcoma/drug therapy , Sesquiterpenes/pharmacology , Sesquiterpenes/therapeutic use , Apoptosis
2.
Clin Cancer Res ; 25(13): 4079-4090, 2019 07 01.
Article in English | MEDLINE | ID: mdl-30940653

ABSTRACT

PURPOSE: Acute myeloid leukemia (AML) is a hematologic malignancy characterized by the accumulation of immature myeloid precursor cells. AML is poorly responsive to conventional chemotherapy and a diagnosis of AML is usually fatal. More effective and less toxic forms of therapy are desperately needed. AML cells are known to be highly dependent on the amino acid glutamine for their survival. These studies were directed at determining the effects of glutaminase inhibition on metabolism in AML and identifying general weaknesses that can be exploited therapeutically. EXPERIMENTAL DESIGN: AML cancer cell lines, primary AML cells, and mouse models of AML and acute lymphoblastic leukemia (ALL) were utilized. RESULTS: We show that blocking glutamine metabolism through the use of a glutaminase inhibitor (CB-839) significantly impairs antioxidant glutathione production in multiple types of AML, resulting in accretion of mitochondrial reactive oxygen species (mitoROS) and apoptotic cell death. Moreover, glutaminase inhibition makes AML cells susceptible to adjuvant drugs that further perturb mitochondrial redox state, such as arsenic trioxide (ATO) and homoharringtonine (HHT). Indeed, the combination of ATO or HHT with CB-839 exacerbates mitoROS and apoptosis, and leads to more complete cell death in AML cell lines, primary AML patient samples, and in vivo using mouse models of AML. In addition, these redox-targeted combination therapies are effective in eradicating ALL cells in vitro and in vivo. CONCLUSIONS: Targeting glutamine metabolism in combination with drugs that perturb mitochondrial redox state represents an effective and potentially widely applicable therapeutic strategy for treating multiple types of leukemia.


Subject(s)
Antineoplastic Agents/pharmacology , Energy Metabolism/drug effects , Glutamine/metabolism , Leukemia/metabolism , Oxidation-Reduction/drug effects , Animals , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Disease Models, Animal , Humans , Leukemia/drug therapy , Leukemia/pathology , Mice , Mice, Transgenic , Molecular Targeted Therapy/methods , Reactive Oxygen Species/metabolism , Xenograft Model Antitumor Assays
3.
Bioorg Med Chem ; 25(3): 1235-1241, 2017 02 01.
Article in English | MEDLINE | ID: mdl-28049618

ABSTRACT

A series of carbamate derivatives of the antileukemic sesquiterpene melampomagnolide B (MMB) has been synthesized utilizing a 1,2,4-triazole carbamate conjugate of MMB as an intermediate synthon. Five imidazole- and benzimidazole-carbamate analogs of MMB (8a-8e) were prepared and evaluated for anti-leukemic activity against cultured M9 ENL1 AML cells. All the analogs exhibited improved anti-leukemic activity (EC50=0.90-3.93µM) when compared to parthenolide and the parent sesquiterpene, MMB (EC50=7.0µM and 15.5µM, respectively). The imidazole carbamate analog, 8a (EC50=0.9µM), was 16 times more potent than MMB. The comparative bioavailabilities of 8a and MMB were determined in BALB/c mice following oral dosing of these compounds. It has been demonstrated that the absolute plasma bioavailabilities of MMB and 8a were 6.7±0.8%, and 45.5±2%, respectively. These results indicate that, compared to MMB, the PK parameters for 8a display significantly improved bioavailability and exposure after oral administration. Analog 8a is considered to be a potential clinical candidate for treatment of acute myelogenous leukemia.


Subject(s)
Antineoplastic Agents/pharmacology , Leukemia, Myeloid, Acute/drug therapy , Sesquiterpenes/pharmacology , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Cell Proliferation/drug effects , Cell Survival/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Humans , Leukemia, Myeloid, Acute/pathology , Male , Mice , Mice, Inbred BALB C , Molecular Structure , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/pathology , Sesquiterpenes/chemical synthesis , Sesquiterpenes/chemistry , Structure-Activity Relationship , Tumor Cells, Cultured
4.
Clin Cancer Res ; 21(6): 1360-72, 2015 Mar 15.
Article in English | MEDLINE | ID: mdl-25547679

ABSTRACT

PURPOSE: Although tyrosine kinase inhibitors (TKI) can be effective therapies for leukemia, they fail to fully eliminate leukemic cells and achieve durable remissions for many patients with advanced BCR-ABL(+) leukemias or acute myelogenous leukemia (AML). Through a large-scale synthetic lethal RNAi screen, we identified pyruvate dehydrogenase, the limiting enzyme for pyruvate entry into the mitochondrial tricarboxylic acid cycle, as critical for the survival of chronic myelogenous leukemia (CML) cells upon BCR-ABL inhibition. Here, we examined the role of mitochondrial metabolism in the survival of Ph(+) leukemia and AML upon TK inhibition. EXPERIMENTAL DESIGN: Ph(+) cancer cell lines, AML cell lines, leukemia xenografts, cord blood, and patient samples were examined. RESULTS: We showed that the mitochondrial ATP-synthase inhibitor oligomycin-A greatly sensitized leukemia cells to TKI in vitro. Surprisingly, oligomycin-A sensitized leukemia cells to BCR-ABL inhibition at concentrations of 100- to 1,000-fold below those required for inhibition of respiration. Oligomycin-A treatment rapidly led to mitochondrial membrane depolarization and reduced ATP levels, and promoted superoxide production and leukemia cell apoptosis when combined with TKI. Importantly, oligomycin-A enhanced elimination of BCR-ABL(+) leukemia cells by TKI in a mouse model and in primary blast crisis CML samples. Moreover, oligomycin-A also greatly potentiated the elimination of FLT3-dependent AML cells when combined with an FLT3 TKI, both in vitro and in vivo. CONCLUSIONS: TKI therapy in leukemia cells creates a novel metabolic state that is highly sensitive to particular mitochondrial perturbations. Targeting mitochondrial metabolism as an adjuvant therapy could therefore improve therapeutic responses to TKI for patients with BCR-ABL(+) and FLT3(ITD) leukemias.


Subject(s)
Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Mitochondrial Proton-Translocating ATPases/antagonists & inhibitors , Oligomycins/pharmacology , Protein Kinase Inhibitors/pharmacology , fms-Like Tyrosine Kinase 3/genetics , Animals , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Apoptosis/drug effects , Cell Line, Tumor , Dihydrolipoyllysine-Residue Acetyltransferase/genetics , Disease Models, Animal , Female , Fusion Proteins, bcr-abl/metabolism , Humans , Imatinib Mesylate/pharmacology , Ketone Oxidoreductases/metabolism , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Membrane Potential, Mitochondrial/drug effects , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, Knockout , Mitochondria/metabolism , Mitochondrial Proteins/genetics , Protein-Tyrosine Kinases/antagonists & inhibitors , RNA Interference , RNA, Small Interfering , Superoxides/metabolism , fms-Like Tyrosine Kinase 3/metabolism
5.
Cancer Res ; 74(14): 3890-901, 2014 Jul 15.
Article in English | MEDLINE | ID: mdl-24872387

ABSTRACT

Eradicating cancer stem-like cells (CSC) may be essential to fully eradicate cancer. Metabolic changes in CSC could hold a key to their targeting. Here, we report that the dietary micronutrient selenium can trigger apoptosis of CSC derived from chronic or acute myelogenous leukemias when administered at supraphysiologic but nontoxic doses. In leukemia CSC, selenium treatment activated ATM-p53-dependent apoptosis accompanied by increased intracellular levels of reactive oxygen species. Importantly, the same treatment did not trigger apoptosis in hematopoietic stem cells. Serial transplantation studies with BCR-ABL-expressing CSC revealed that the selenium status in mice was a key determinant of CSC survival. Selenium action relied upon the endogenous production of the cyclooxygenase-derived prostaglandins Δ(12)-PGJ2 and 15d-PGJ2. Accordingly, nonsteroidal anti-inflammatory drugs and NADPH oxidase inhibitors abrogated the ability of selenium to trigger apoptosis in leukemia CSC. Our results reveal how selenium-dependent modulation of arachidonic acid metabolism can be directed to trigger apoptosis of primary human and murine CSC in leukemia.


Subject(s)
Eicosanoids/metabolism , Leukemia/metabolism , Selenium/pharmacology , Animals , Apoptosis/drug effects , Arachidonic Acids/metabolism , Cell Transformation, Neoplastic/drug effects , Cytochrome P-450 Enzyme System/metabolism , Humans , Leukemia/pathology , Macrophages/drug effects , Macrophages/metabolism , Male , Mice , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Oxidative Stress/drug effects , Reactive Oxygen Species/metabolism , Selenium/administration & dosage , Signal Transduction/drug effects , Splenomegaly , Tumor Suppressor Protein p53/metabolism
6.
Best Pract Res Clin Haematol ; 25(4): 415-8, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23200537

ABSTRACT

There are a number of approaches for selective targeting of leukemic stem cells (LSCs). These include targeting stem-cell properties, such as self-renewal, inducing cycling of quiescent LSCs to sensitize them to conventional agents, employing or inducing immune-based mechanisms, and targeting tumor-specific physiology. Agents such as parthenolide inhibit the ability of leukemic stem cells to respond to oxidative stress and make leukemic stem cells and bulk leukemic cells susceptible to cell death, while normal stem cells remain relatively unharmed by these agents. The major mechanism of action of these small molecules appears to revolve around the aberrant glutathione metabolism pathway found in leukemic cells.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Drug Delivery Systems , Leukemia/drug therapy , Leukemia/metabolism , Neoplastic Stem Cells/metabolism , Sesquiterpenes/therapeutic use , Animals , Cell Death/drug effects , Glutathione/metabolism , Humans , Leukemia/pathology , Oxidative Stress/drug effects
7.
Cancer Invest ; 29(7): 439-50, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21740082

ABSTRACT

In this work, effects of bortezomib on apoptosis, clonal progenitor growth, cytokine production, and NF-κB expression in patients with MDS with cytopenias requiring transfusion support are examined. Bortezomib increased apoptosis in marrow mononuclear cells but had no effects on CFU-GM, BFU-E, or CFU-L content. No consistent effects on NF-κB activation in vivo were noted. To further define the role of bortezomib in AML and MDS, we examined it in combination with several targeted agents and chemotherapeutic agents in vitro. Combinations with arsenic trioxide, sorafenib, and cytarabine demonstrated synergistic in vitro effects in AML cell lines.


Subject(s)
Antineoplastic Agents/therapeutic use , Boronic Acids/therapeutic use , Leukemia, Myeloid, Acute/drug therapy , Myelodysplastic Syndromes/drug therapy , Protease Inhibitors/therapeutic use , Proteasome Inhibitors , Pyrazines/therapeutic use , Apoptosis/drug effects , Arsenic Trioxide , Arsenicals/pharmacology , Azacitidine/pharmacology , Benzenesulfonates/pharmacology , Boronic Acids/pharmacology , Bortezomib , Cell Line, Tumor , Cell Proliferation/drug effects , Cytarabine/pharmacology , Cytokines/blood , Farnesyltranstransferase/antagonists & inhibitors , Hematopoietic Stem Cells/drug effects , Humans , Leukemia, Myeloid, Acute/pathology , Myelodysplastic Syndromes/pathology , Niacinamide/analogs & derivatives , Oxides/pharmacology , Phenylurea Compounds , Pyrazines/pharmacology , Pyridines/pharmacology , Sorafenib
8.
Br J Haematol ; 151(1): 70-83, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20701602

ABSTRACT

Interactions between the nuclear factor (NF)-κB inhibitor parthenolide and the pan-histone deacetylase inhibitors (HDACIs) vorinostat and LBH589 were investigated in human acute myeloid leukaemia (AML) cells, including primary AML blasts. Co-administration of parthenolide blocked HDACI-mediated phosphorylation/activation of IKK and RelA/p65 in association with increased JNK1 activation in various AML cell types. These events were accompanied by an increase in apoptosis in multiple AML cell lines (e.g. U937, HL-60, NB4, MV-4-11, and MOLM-13). Significantly, parthenolide also increased HDACI-mediated cell death in haematopoietic cells transduced with the MLL-MLLT1 fusion gene, which exhibit certain leukaemia-initiating cell characteristics, as well as primary AML blasts. Exposure to parthenolide/HDACI regimens clearly inhibited the growth of AML-colony-forming units but was relatively sparing toward normal haematopoietic progenitors. Notably, blockade of c-Jun N-terminal kinase (JNK) signalling by either pharmacological inhibitors or genetic means (e.g. dominant-negative JNK1 or JNK1 shRNA) diminished parthenolide/HDACI-mediated lethality. Moreover, dominant-negative MKK7, but not dominant-negative MKK4/SEK1, blocked JNK1 activation and apoptosis induced by parthenolide/HDACI regimens. Together, these findings indicate that parthenolide potentiates HDACI lethality in human AML cells through a process involving NF-κB inhibition and subsequent MKK7-dependent activation of the SAPK/JNK pathway. They also raise the possibility that this strategy may target leukaemic progenitor cells.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Leukemia, Myeloid, Acute/pathology , NF-kappa B/antagonists & inhibitors , Apoptosis/drug effects , Drug Evaluation, Preclinical/methods , Drug Synergism , Enzyme Activation/drug effects , Histone Deacetylase Inhibitors/administration & dosage , Histone Deacetylase Inhibitors/pharmacology , Humans , JNK Mitogen-Activated Protein Kinases/metabolism , Leukemia, Myeloid, Acute/metabolism , Sesquiterpenes/administration & dosage , Sesquiterpenes/pharmacology , Signal Transduction/drug effects , Tumor Cells, Cultured
9.
Best Pract Res Clin Haematol ; 20(1): 13-8, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17336250

ABSTRACT

Malignant stem cells have recently been described as the source of several types of human cancer. These unique cell types are typically rare and possess properties that are distinct from most other tumor cells. The properties of leukemic stem cells indicate that current chemotherapy drugs will not be effective. The use of current cytotoxic agents is not effective in leukemia because the agents target both the leukemic and normal stem cell populations. Consequently, new strategies are required that specifically and preferentially target the malignant stem cell population, while sparing normal stem cells. Several well known agents are lethal for the leukemic stem cell in preclinical testing. They include parthenolide, commonly known as feverfew, and TDZD-8. They have undergone various levels of preclinical development, but have not been used in patients as yet in the cancer setting. These drugs and combinations of existing therapies that target the leukemic stem cell population may provide a cure in this disease. This article summarizes recent findings in the leukemic stem cell field and discusses new directions for therapy.


Subject(s)
Leukemia/drug therapy , Neoplastic Stem Cells/pathology , Antineoplastic Agents/therapeutic use , Drug Evaluation, Preclinical , Humans , Leukemia/pathology , Neoplastic Stem Cells/cytology , Neoplastic Stem Cells/drug effects , Sesquiterpenes/therapeutic use , Thiadiazoles/therapeutic use
10.
Expert Opin Biol Ther ; 5(9): 1147-52, 2005 Sep.
Article in English | MEDLINE | ID: mdl-16120045

ABSTRACT

Malignant stem cells are central to the pathogenesis and perpetuation of acute myelogenous leukaemia (AML). Despite their crucial role, standard chemotherapy often does not target these critical cells and, thus, the 'root' of leukaemic disease is not eradicated. To derive better therapies, unique molecular features of malignant stem cells have been characterised for AML and evaluated with regard to ablation of disease. In the course of such studies, the compound parthenolide, which is derived from the medicinal plant feverfew, has recently been shown to preferentially induce AML stem cells to undergo apoptosis. Importantly, parthenolide had no discernable effect on normal blood cells. Thus, this naturally occurring agent may provide new avenues of investigation for the treatment of leukaemia. In this article, characteristics of parthenolide are reviewed.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Leukemia, Myeloid, Acute/drug therapy , Neoplastic Stem Cells/drug effects , Sesquiterpenes/pharmacology , Tanacetum parthenium/chemistry , Animals , Antineoplastic Agents, Phytogenic/isolation & purification , Antineoplastic Agents, Phytogenic/therapeutic use , Apoptosis , Cell Line, Tumor , Humans , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/pathology , NF-kappa B/antagonists & inhibitors , NF-kappa B/metabolism , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Plants, Medicinal/chemistry , Sesquiterpenes/isolation & purification , Sesquiterpenes/therapeutic use , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL