Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 175
Filter
1.
Pharmacol Res ; 158: 104682, 2020 08.
Article in English | MEDLINE | ID: mdl-32035162

ABSTRACT

Advanced systemic mastocytosis is a rare and still untreatable disease. Blocking antibodies against inhibitory receptors, also known as "immune checkpoints", have revolutionized anti-cancer treatment. Inhibitory receptors are expressed not only on normal immune cells, including mast cells but also on neoplastic cells. Whether activation of inhibitory receptors through monoclonal antibodies can lead to tumor growth inhibition remains mostly unknown. Here we show that the inhibitory receptor Siglec-7 is expressed by primary neoplastic mast cells in patients with systemic mastocytosis and by mast cell leukemia cell lines. Activation of Siglec-7 by anti-Siglec-7 monoclonal antibody caused phosphorylation of Src homology region 2 domain-containing phosphatase-1 (SHP-1), reduced phosphorylation of KIT and induced growth inhibition in mast cell lines. In SCID-beige mice injected with either the human mast cell line HMC-1.1 and HMC-1.2 or with Siglec-7 transduced B cell lymphoma cells, anti-Siglec-7 monoclonal antibody reduced tumor growth by a mechanism involving Siglec-7 cytoplasmic domains in "preventive" and "treatment" settings. These data demonstrate that activation of Siglec-7 on mast cell lines can inhibit their growth in vitro and in vivo. This might pave the way to additional treatment strategies for mastocytosis.


Subject(s)
Lectins/agonists , Leukemia, Mast-Cell/drug therapy , Adult , Aged , Aged, 80 and over , Animals , Antibodies, Monoclonal/therapeutic use , Antigens, Differentiation, Myelomonocytic , Cell Line, Tumor , Cell Survival/drug effects , Female , Genes, src/drug effects , Humans , Leukemia, Mast-Cell/pathology , Lymphoma, B-Cell/drug therapy , Lymphoma, B-Cell/pathology , Male , Mastocytosis/drug therapy , Mice , Mice, SCID , Middle Aged , Phosphorylation , Protein Tyrosine Phosphatase, Non-Receptor Type 6/drug effects , Proto-Oncogene Proteins c-kit/drug effects , Xenograft Model Antitumor Assays
2.
Br J Haematol ; 187(4): 488-501, 2019 11.
Article in English | MEDLINE | ID: mdl-31309543

ABSTRACT

Mutations in two type-3 receptor tyrosine kinases (RTKs), KIT and FLT3, are common in both acute myeloid leukaemia (AML) and systemic mastocytosis (SM) and lead to hyperactivation of key signalling pathways. A large number of tyrosine kinase inhibitors (TKIs) have been developed that target either FLT3 or KIT and significant clinical benefit has been demonstrated in multiple clinical trials. Given the structural similarity of FLT3 and KIT, it is not surprising that some of these TKIs inhibit both of these receptors. This is typified by midostaurin, which has been approved by the US Food and Drug Administration for mutant FLT3-positive AML and for KIT D816V-positive SM. Here, we compare the in vitro activities of the clinically available FLT3 and KIT inhibitors with those of midostaurin against a panel of cells expressing a variety of oncogenic FLT3 or KIT receptors, including wild-type (wt) FLT3, FLT3-internal tandem duplication (ITD), FLT3 D835Y, the resistance mutant FLT3-ITD+ F691L, KIT D816V, and KIT N822K. We also examined the effects of these inhibitors in vitro and in vivo on cells expressing mutations in c-CBL found in AML that result in hypersensitization of RTKs, such as FLT3 and KIT. The results show a wide spectrum of activity of these various mutations to these clinically available TKIs.


Subject(s)
Antineoplastic Agents/pharmacology , Hematologic Neoplasms/drug therapy , Mutant Proteins/drug effects , Protein Kinase Inhibitors/pharmacology , Aniline Compounds/pharmacology , Aniline Compounds/therapeutic use , Antineoplastic Agents/therapeutic use , Benzimidazoles/pharmacology , Benzimidazoles/therapeutic use , Benzothiazoles/pharmacology , Benzothiazoles/therapeutic use , Cell Line, Tumor , Drug Screening Assays, Antitumor , Hematologic Neoplasms/genetics , Humans , Phenylurea Compounds/pharmacology , Phenylurea Compounds/therapeutic use , Piperidines/pharmacology , Piperidines/therapeutic use , Protein Kinase Inhibitors/therapeutic use , Proto-Oncogene Proteins c-cbl/drug effects , Proto-Oncogene Proteins c-cbl/genetics , Proto-Oncogene Proteins c-kit/drug effects , Proto-Oncogene Proteins c-kit/genetics , Pyrazines/pharmacology , Pyrazines/therapeutic use , Pyrazoles/pharmacology , Pyrazoles/therapeutic use , Pyrroles/pharmacology , Pyrroles/therapeutic use , Sorafenib/pharmacology , Sorafenib/therapeutic use , Staurosporine/analogs & derivatives , Staurosporine/pharmacology , Staurosporine/therapeutic use , Triazines/pharmacology , Triazines/therapeutic use , fms-Like Tyrosine Kinase 3/drug effects , fms-Like Tyrosine Kinase 3/genetics
3.
Circ Res ; 114(11): 1700-1712, 2014 May 23.
Article in English | MEDLINE | ID: mdl-24718482

ABSTRACT

RATIONALE: Sorafenib is an effective treatment for renal cell carcinoma, but recent clinical reports have documented its cardiotoxicity through an unknown mechanism. OBJECTIVE: Determining the mechanism of sorafenib-mediated cardiotoxicity. METHODS AND RESULTS: Mice treated with sorafenib or vehicle for 3 weeks underwent induced myocardial infarction (MI) after 1 week of treatment. Sorafenib markedly decreased 2-week survival relative to vehicle-treated controls, but echocardiography at 1 and 2 weeks post MI detected no differences in cardiac function. Sorafenib-treated hearts had significantly smaller diastolic and systolic volumes and reduced heart weights. High doses of sorafenib induced necrotic death of isolated myocytes in vitro, but lower doses did not induce myocyte death or affect inotropy. Histological analysis documented increased myocyte cross-sectional area despite smaller heart sizes after sorafenib treatment, further suggesting myocyte loss. Sorafenib caused apoptotic cell death of cardiac- and bone-derived c-kit+ stem cells in vitro and decreased the number of BrdU+ (5-bromo-2'-deoxyuridine+) myocytes detected at the infarct border zone in fixed tissues. Sorafenib had no effect on infarct size, fibrosis, or post-MI neovascularization. When sorafenib-treated animals received metoprolol treatment post MI, the sorafenib-induced increase in post-MI mortality was eliminated, cardiac function was improved, and myocyte loss was ameliorated. CONCLUSIONS: Sorafenib cardiotoxicity results from myocyte necrosis rather than from any direct effect on myocyte function. Surviving myocytes undergo pathological hypertrophy. Inhibition of c-kit+ stem cell proliferation by inducing apoptosis exacerbates damage by decreasing endogenous cardiac repair. In the setting of MI, which also causes large-scale cell loss, sorafenib cardiotoxicity dramatically increases mortality.


Subject(s)
Antineoplastic Agents/adverse effects , Antineoplastic Agents/pharmacology , Heart/drug effects , Myocardial Infarction/mortality , Niacinamide/analogs & derivatives , Phenylurea Compounds/adverse effects , Phenylurea Compounds/pharmacology , Animals , Apoptosis/drug effects , Cats , Cell Proliferation/drug effects , Cells, Cultured , Disease Models, Animal , Dose-Response Relationship, Drug , In Vitro Techniques , Male , Metoprolol/pharmacology , Mice , Mice, Inbred C57BL , Myocardial Infarction/pathology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/pathology , Niacinamide/adverse effects , Niacinamide/pharmacology , Proto-Oncogene Proteins c-kit/drug effects , Proto-Oncogene Proteins c-kit/metabolism , Sorafenib
4.
J Nat Prod ; 79(2): 293-9, 2016 Feb 26.
Article in English | MEDLINE | ID: mdl-26807861

ABSTRACT

Although stem cell factor receptor (c-KIT) kinase is responsible for various malignant human cancers, the presence of constitutively active gain-of-function mutants has made it difficult to discover new anticancer agents using c-KIT as the target protein. To identify the common inhibitors of wild-type c-KIT and the most abundant gain-of-function mutant (D816V), the virtual screening of natural products was performed for the two target proteins in parallel with the scoring function improved by implementing a sophisticated solvation free energy term. As a result, four common inhibitors of natural origin are found with biochemical potencies ranging from low micromolar to submicromolar levels. The results of extensive docking simulations show that although the natural-product inhibitors establish weaker hydrophobic interactions with the D816V mutant than with the wild type, they exhibit a little higher inhibitory activity for the former than the latter by strengthening the hydrogen-bond interactions to a sufficient extent. Of the four natural-product inhibitors, (Z)-6-hydroxy-2-(4-methoxybenzylidene)benzofuran-3(2H)-one (3) is anticipated to serve as a new molecular core for the structure-activity relationship studies to optimize the biochemical potencies because it exhibits good inhibitory activity against both the wild type and D816V mutant despite its low molecular weight (268.3 amu).


Subject(s)
Antineoplastic Agents/pharmacology , Benzofurans/pharmacology , Biological Products/pharmacology , Proto-Oncogene Proteins c-kit/metabolism , Algorithms , Antineoplastic Agents/chemistry , Benzofurans/chemistry , Biological Products/chemistry , HEK293 Cells , Humans , Molecular Structure , Mutation/drug effects , Protein Kinase Inhibitors/chemistry , Proto-Oncogene Proteins c-kit/drug effects , Proto-Oncogene Proteins c-kit/genetics , Structure-Activity Relationship
5.
Reproduction ; 149(4): 329-38, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25737569

ABSTRACT

Prospermatogonia transition to type A spermatogonia, which provide the source for the spermatogonial stem cell (SSC) pool. A percentage of these type A spermatogonia then differentiate to enter meiosis as spermatocytes by ∼P10. It is currently unclear as to when these distinct populations are initially formed in the neonatal testis, and when the expression of markers both characteristic of and required for the adult undifferentiated and differentiating states is established. In this study, we compared expression of known spermatogonial cell fate markers during normal development and in response to the differentiation signal provided by retinoic acid (RA). We found that some markers for the undifferentiated state (ZBTB16/PLZF and CDH1) were expressed in nearly all spermatogonia from P1 through P7. In contrast, differentiation markers (STRA8 and KIT) appeared in a subset of spermatogonia at P4, coincident with the onset of RA signaling. GFRA1, which was present in nearly all prospermatogonia at P1, was only retained in STRA8/KIT- spermatogonia. From P4 through P10, there was a great deal of heterogeneity in the male germ cell population in terms of expression of markers, as markers characteristic of the undifferentiated (except GFRA1) and differentiating states were co-expressed through this interval. After P10, these fate markers diverged to mark distinct populations of undifferentiated and differentiating spermatogonia, and this pattern was maintained in juvenile (P18) and adult (P>60) testes. Taken together, these results reveal that the spermatogonia population is heterogeneous during the first wave of spermatogenesis, and indicate that neonatal spermatogonia may not serve as an ideal substitute for studying the function of adult spermatogonia.


Subject(s)
Biomarkers/metabolism , Cell Differentiation/physiology , Spermatogenesis/physiology , Spermatogonia/cytology , Testis/cytology , Animals , Animals, Newborn , Antineoplastic Agents/pharmacology , Cdh1 Proteins/drug effects , Cell Differentiation/drug effects , Immunoenzyme Techniques , Kruppel-Like Transcription Factors/drug effects , Male , Mice , Mice, Inbred C57BL , Promyelocytic Leukemia Zinc Finger Protein , Proto-Oncogene Proteins c-kit/drug effects , Spermatogenesis/drug effects , Spermatogonia/drug effects , Spermatogonia/metabolism , Testis/drug effects , Testis/metabolism , Tretinoin/pharmacology
6.
Oncology ; 89(1): 1-13, 2015.
Article in English | MEDLINE | ID: mdl-25720422

ABSTRACT

Gastrointestinal stromal tumour (GIST), while relatively rare, is the most common mesenchymal tumour of the gastrointestinal tract. These tumours are largely resistant to cytotoxic chemotherapy and, in the past, were typically managed surgically. However, as a result of the identification of activating mutations in the proto-oncogene KIT and the development of compounds that inhibit the KIT receptor tyrosine kinase, GISTs have, in the last 14 years, become the archetype of a targeted agent-responsive tumour. Due to the almost continual emergence of new data from clinical trials and other studies on GIST diagnosis and treatment, the management of this disease requires regular review. The 2013 ArcheoloGIST summit was convened in Prague, Czech Republic. Interaction between attending physicians and the expert faculty was a core component of the summit. The current article is based on discussions held during two interactive sessions at ArcheoloGIST 2013 in which the authors aimed to: (1) reach a consensus on the current management of GIST and (2) provide a vision for the future diagnosis and treatment of this disease.


Subject(s)
Antineoplastic Agents/therapeutic use , Gastrointestinal Neoplasms/drug therapy , Gastrointestinal Stromal Tumors/drug therapy , Medical Oncology/trends , Molecular Targeted Therapy/methods , Mutation , Protein Kinase Inhibitors/therapeutic use , Protein-Tyrosine Kinases/antagonists & inhibitors , Proto-Oncogene Proteins c-kit/genetics , Antineoplastic Agents/pharmacology , Benzamides/therapeutic use , Chemotherapy, Adjuvant , Clinical Trials as Topic , Congresses as Topic , Consensus , Drug Administration Schedule , Drug Resistance, Neoplasm , Gastrointestinal Neoplasms/genetics , Gastrointestinal Neoplasms/pathology , Gastrointestinal Neoplasms/surgery , Gastrointestinal Stromal Tumors/genetics , Gastrointestinal Stromal Tumors/pathology , Gastrointestinal Stromal Tumors/surgery , Humans , Imatinib Mesylate , Mutation/drug effects , Piperazines/therapeutic use , Proto-Oncogene Mas , Proto-Oncogene Proteins c-kit/drug effects , Pyrimidines/therapeutic use , Receptor, Platelet-Derived Growth Factor alpha/genetics , Treatment Outcome
7.
PLoS Comput Biol ; 10(7): e1003749, 2014 Jul.
Article in English | MEDLINE | ID: mdl-25079768

ABSTRACT

Receptor tyrosine kinase KIT controls many signal transduction pathways and represents a typical allosterically regulated protein. The mutation-induced deregulation of KIT activity impairs cellular physiological functions and causes serious human diseases. The impact of hotspots mutations (D816H/Y/N/V and V560G/D) localized in crucial regulatory segments, the juxtamembrane region (JMR) and the activation (A-) loop, on KIT internal dynamics was systematically studied by molecular dynamics simulations. The mutational outcomes predicted in silico were correlated with in vitro and in vivo activation rates and drug sensitivities of KIT mutants. The allosteric regulation of KIT in the native and mutated forms is described in terms of communication between the two remote segments, JMR and A-loop. A strong correlation between the communication profile and the structural and dynamical features of KIT in the native and mutated forms was established. Our results provide new insight on the determinants of receptor KIT constitutive activation by mutations and resistance of KIT mutants to inhibitors. Depiction of an intra-molecular component of the communication network constitutes a first step towards an integrated description of vast communication pathways established by KIT in physiopathological contexts.


Subject(s)
Mutation/genetics , Proto-Oncogene Proteins c-kit/chemistry , Proto-Oncogene Proteins c-kit/genetics , Allosteric Regulation/genetics , Antineoplastic Agents/pharmacology , Cytoplasm/chemistry , Cytoplasm/metabolism , Humans , Hydrogen Bonding , Molecular Dynamics Simulation , Protein Structure, Tertiary , Proto-Oncogene Proteins c-kit/drug effects , Proto-Oncogene Proteins c-kit/metabolism , Signal Transduction
8.
J Surg Res ; 183(2): 598-605, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23608618

ABSTRACT

BACKGROUND: To investigate the effects of the c-kit blocker imatinib mesylate (Glivec) on the bladders of animals with suprasacral cord injury (SSCI) and sacral cord injury (SCI). MATERIALS AND METHODS: We randomized 60 female Sprague-Dawley rats into control, sham, SSCI (T8/9 transection), and SCI (S1-3 transection) groups. Six weeks later, we evaluated the effects of stepwise Glivec administrations on urinary bladder contraction using cystometry and the detrusor strip stretch-test. We investigated spontaneous calcium transients of kit-positive interstitial cells of Cajal (ICCs) with the preloaded Ca(2+) indicator fluo-3AM. The expression levels of c-kit and the number of ICCs in those bladders were determined using Western blot and fluorescence staining analyses, respectively. RESULTS: Bladder capacity and compliance were decreased in SSCI bladders and increased in SCI bladders (P<0.05). The amplitude and frequency of spontaneous contractions of detrusor strips, the frequency and relative fluorescence intensity of the spontaneous Ca(2+) waves, and c-kit expression in the bladder were significantly increased in the SSCI group and decreased in the SCI group compared with the control and sham groups (P<0.05). The dose-dependent effects of Glivec also confirmed consistent functional variations in bladder activity. CONCLUSIONS: The expressions and effects of Glivec were enhanced in SSCI bladders and inhibited in SCI bladders, which may indicate potential roles of ICCs for the c-kit signaling pathway in the pathogenesis of SSCI and SCI bladder.


Subject(s)
Benzamides/pharmacology , Benzamides/therapeutic use , Piperazines/pharmacology , Piperazines/therapeutic use , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-kit/metabolism , Pyrimidines/pharmacology , Pyrimidines/therapeutic use , Spinal Cord Injuries/drug therapy , Urinary Bladder/drug effects , Urinary Bladder/physiopathology , Animals , Calcium/metabolism , Dose-Response Relationship, Drug , Female , Imatinib Mesylate , Interstitial Cells of Cajal/metabolism , Interstitial Cells of Cajal/pathology , Models, Animal , Muscle Contraction/drug effects , Muscle Contraction/physiology , Protein Kinase Inhibitors/therapeutic use , Proto-Oncogene Proteins c-kit/antagonists & inhibitors , Proto-Oncogene Proteins c-kit/drug effects , Rats , Rats, Sprague-Dawley , Sacrum , Signal Transduction/drug effects , Signal Transduction/physiology , Spinal Cord Injuries/metabolism , Spinal Cord Injuries/physiopathology , Urinary Bladder/pathology
9.
Cancer Cell ; 8(6): 467-78, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16338660

ABSTRACT

The erythroleukemia developed by spi-1/PU.1 transgenic mice is a multistage process characterized by an early arrest of the proerythroblast differentiation followed later on by malignant transformation. Herein, we report the presence of acquired mutations in the SCF receptor gene (Kit) in 86% of tumors isolated during the late stage of the disease. Kit mutations affect codon 814 or 818. Ectopic expression of Kit mutants in nonmalignant proerythroblasts confers erythropoietin independence and tumorigenicity to cells. Using PP1, PP2, and imatinib mesylate, we show that Kit mutants are responsible for the autonomous expansion of malignant cells via Erk1/2 and PI3K/Akt activations. These findings represent a proof of principle for oncogenic cooperativity between one proliferative and one differentiation blocking event for the development of an overt leukemia.


Subject(s)
Leukemia, Erythroblastic, Acute/genetics , Promoter Regions, Genetic , Proto-Oncogene Proteins c-kit/genetics , Proto-Oncogene Proteins/biosynthesis , Trans-Activators/biosynthesis , Animals , Benzamides , Cell Differentiation/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Disease Progression , Gene Expression Regulation, Neoplastic/drug effects , Humans , Imatinib Mesylate , Leukemia, Erythroblastic, Acute/metabolism , Mice , Mice, Transgenic , Mutation , Piperazines/pharmacology , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-kit/drug effects , Proto-Oncogene Proteins c-kit/metabolism , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Time Factors , Trans-Activators/genetics
10.
Cancer Cell ; 1(5): 421-32, 2002 Jun.
Article in English | MEDLINE | ID: mdl-12124172

ABSTRACT

Up to 30% of acute myelogenous leukemia (AML) patients harbor an activating internal tandem duplication (ITD) within the juxtamembrane domain of the FLT3 receptor, suggesting that it may be a target for kinase inhibitor therapy. For this purpose we have developed CT53518, a potent antagonist that inhibits FLT3, platelet-derived growth factor receptor (PDGFR), and c-Kit (IC(50) approximately 200 nM), while other tyrosine or serine/threonine kinases were not significantly inhibited. In Ba/F3 cells expressing different FLT3-ITD mutants, CT53518 inhibited IL-3-independent cell growth and FLT3-ITD autophosphorylation with an IC(50) of 10-100 nM. In human FLT3-ITD-positive AML cell lines, CT53518 induced apoptosis and inhibited FLT3-ITD phosphorylation, cellular proliferation, and signaling through the MAP kinase and PI3 kinase pathways. Therapeutic efficacy of CT53518 was demonstrated both in a nude mouse model and in a murine bone marrow transplant model of FLT3-ITD-induced disease.


Subject(s)
Antineoplastic Agents/pharmacology , Enzyme Inhibitors/pharmacology , Leukemia, Myeloid, Acute/drug therapy , Piperazines/pharmacology , Proto-Oncogene Proteins/antagonists & inhibitors , Quinazolines/pharmacology , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Animals , Apoptosis/drug effects , Bone Marrow Cells/enzymology , Bone Marrow Cells/pathology , Bone Marrow Transplantation , Flow Cytometry , Humans , Immunoblotting , Interleukin-3/metabolism , Leukemia, Myeloid, Acute/enzymology , Leukemia, Myeloid, Acute/genetics , Mice , Mice, Inbred BALB C , Mice, Nude , Mutation , Phosphorylation , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-kit/drug effects , Proto-Oncogene Proteins c-kit/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Receptors, Cell Surface/antagonists & inhibitors , Receptors, Platelet-Derived Growth Factor/antagonists & inhibitors , Tandem Repeat Sequences , Transfection , Tumor Cells, Cultured/drug effects , fms-Like Tyrosine Kinase 3
11.
Br J Dermatol ; 164(2): 372-86, 2011 Feb.
Article in English | MEDLINE | ID: mdl-20846303

ABSTRACT

BACKGROUND: Keloids are fibroproliferative disorders characterized by increased deposition of extracellular matrix components. Stem cell factor (SCF) and its receptor c-KIT are expressed in a wide variety of cells and have also been demonstrated to be important modulators of the wound healing process. OBJECTIVES: To examine the role of the SCF/c-KIT system in keloid pathogenesis. METHODS: Immunohistochemical staining and Western blot analyses were used to examine localization and expression of SCF and c-KIT in keloid and normal skin tissue. This was followed by the detection of SCF and c-KIT expression in fibroblasts cultured in vitro and fibroblasts exposed to serum. To investigate the effect of epithelial-mesenchymal interactions, a two-chamber system was employed in which keratinocytes on membrane inserts were cocultured with the fibroblasts. SCF and c-KIT expression levels in all cell extracts and conditioned media were assayed by Western blotting. In another set of experiments, the effect of imatinib (Glivec(®), Gleevec(®); Novartis Pharma AG, Basel, Switzerland) on keloid fibroblasts was examined. RESULTS: SCF and c-KIT were upregulated in keloid scar tissue and in cultured fibroblasts stimulated with serum, highlighting their importance in the initial phase of wound healing. We further demonstrated that epithelial-mesenchymal interactions, mimicked by coculture of keratinocytes and fibroblasts in vitro, not only stimulated secretion of the soluble form of SCF in keloid cocultures but also brought about shedding of the extracellular domain of c-KIT perhaps by upregulation of tumour necrosis factor-α converting enzyme which was also upregulated in keloid scars in vivo and keloid cocultures in vitro. In addition keloid cocultures expressed increased levels of phosphorylated c-KIT highlighting an activation of the SCF/c-KIT system. Finally, we demonstrated that imatinib, a tyrosine kinase inhibitor, may be a possible therapeutic agent for keloids. CONCLUSION: These data indicate that the SCF/c-KIT system plays an important role in scar pathogenesis, and underscore the role of imatinib as a key therapeutic agent in keloid scars.


Subject(s)
Fibroblasts/drug effects , Keloid/drug therapy , Piperazines/pharmacology , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-kit/metabolism , Pyrimidines/pharmacology , Stem Cell Factor/metabolism , Benzamides , Blotting, Western , Cell Communication/drug effects , Cells, Cultured , Coculture Techniques , Fibroblasts/metabolism , Humans , Imatinib Mesylate , Immunohistochemistry , Keloid/etiology , Keloid/metabolism , Proto-Oncogene Proteins c-kit/drug effects , Skin/metabolism , Stem Cell Factor/drug effects , Up-Regulation
12.
Curr Probl Cancer ; 45(3): 100705, 2021 06.
Article in English | MEDLINE | ID: mdl-33495025

ABSTRACT

Nilotinib has been used as a third-line drug for gastrointestinal stromal tumors (GISTs) after a failure of sunitinib. In this study, we aimed to evaluate the efficacy of nilotinib in different genomic subtypes of GISTs. We searched the English articles through EMBASE, Cochrane Library and PubMed Database regarding to the use of nilotinib on GISTs, which published up to February 15, 2019. Inclusion criteria were: GISTs patients received nilotinib in a clinical trial and had detailed genetic subtype records (such as KIT exon 9, KIT exon 11, or PDGFRA mutations, or wild-type). The clinical benefit rate was used to assess the efficacy of nilotinib. A total of 3 studies involving 218 GISTs were included in this meta-analysis. The overall OR (KIT group vs WT group) was 3.26 (95% CI: 1.14-9.28; P = 0.027, Pheterogeneity = 0.613). The overall OR in KIT exon 11 group vs WT group was 5.30 (95% CI: 1.79-15.68; P = 0.003, Pheterogeneity = 0.409). The overall OR in KIT exon 9 group vs WT group was 0.13 (95% CI: 0.02-0.86; P = 0.035, Pheterogeneity = 0.229). The overall OR in KIT exon 11 group vs exon 9 group was 9.96 (95% CI: 0.39-254.66; P < 0.0001, Pheterogeneity = 0.024). Different genotypes of GISTs showed different responses to nilotinib, and KIT exon 11-mutant GISTs mostly benefited from nilotinib, followed by KIT exon 9-mutant or WT one.


Subject(s)
Gastrointestinal Neoplasms/drug therapy , Gastrointestinal Neoplasms/genetics , Gastrointestinal Stromal Tumors/drug therapy , Gastrointestinal Stromal Tumors/genetics , Pyrimidines/pharmacology , Adolescent , Adult , Aged , Aged, 80 and over , Female , Gastrointestinal Neoplasms/pathology , Gastrointestinal Stromal Tumors/pathology , Genotype , Humans , Male , Middle Aged , Proto-Oncogene Proteins c-kit/drug effects , Proto-Oncogene Proteins c-kit/genetics , Receptor, Platelet-Derived Growth Factor alpha/drug effects , Receptor, Platelet-Derived Growth Factor alpha/genetics , Treatment Outcome , Young Adult
13.
Am J Clin Pathol ; 155(2): 239-266, 2021 02 04.
Article in English | MEDLINE | ID: mdl-33313644

ABSTRACT

OBJECTIVES: The 2019 Workshop of the Society for Hematopathology/European Association for Haematopathology received and reviewed cases covering the spectrum of mastocytosis and related diseases, including morphologic mimics, focusing on recent updates and relevant findings for pathologists. METHODS: The workshop panel reviewed 99 cases of cutaneous and systemic mastocytosis (SM) and SM and associated hematologic neoplasms (SM-AHN). RESULTS: Despite a common theme of KIT mutation (particularly D816V), mastocytosis is a heterogeneous neoplasm with a wide variety of presentations. This spectrum, including rare subtypes and extramedullary organ involvement, is discussed and illustrated by representative cases. CONCLUSIONS: In the age of targeted treatment aimed at KIT, the accurate diagnosis and classification of mastocytosis has major implications for therapy and further interventions. Understanding the clinical, pathologic, and genetic findings of mastocytosis is crucial for selecting the proper tests to perform and subsequent arrival at a correct diagnosis in this rare disease.


Subject(s)
Mastocytosis , Adolescent , Adult , Aged , Antineoplastic Agents/therapeutic use , Child , Diagnosis, Differential , Female , Genetic Testing , Humans , Immunohistochemistry , Infant , Leukemia, Myeloid, Acute/diagnosis , Leukemia, Myelomonocytic, Chronic/diagnosis , Male , Mast Cells/pathology , Mastocytosis/diagnosis , Mastocytosis/drug therapy , Mastocytosis/genetics , Mastocytosis/pathology , Mastocytosis, Systemic/drug therapy , Mastocytosis, Systemic/pathology , Middle Aged , Mutation , Oncogenes , Prognosis , Proto-Oncogene Proteins c-kit/drug effects , Proto-Oncogene Proteins c-kit/genetics , Tryptases/isolation & purification , Young Adult
14.
Theranostics ; 11(1): 93-106, 2021.
Article in English | MEDLINE | ID: mdl-33391463

ABSTRACT

Rationale: Acute myeloid leukemia (AML) is a common type of haematological malignancy. Several studies have shown that neoplasia in AML is enhanced by tyrosine kinase pathways. Recently, given that aberrant activation of Fms-like tyrosine receptor kinase 3 (FLT3) acts as a critical survival signal for cancer cells in 20‒30% patients with AML, inhibition of FLT3 may be a potential therapeutic strategy. Therefore, we identified LT-171-861, a novel kinase inhibitor with remarkable inhibitory activity against FLT3, in preclinical models of AML. Methods: We determined the inhibitory effects of LT-171-861 in vitro using AML cell lines and transformed BaF3 cells. Target engagement assays were used to verify the interaction between LT-171-861 and FLT3. Finally, a subcutaneous model and a bone marrow engrafted model were used to evaluate the antitumor effects of LT­171­861 in vivo. Results: Our data demonstrated that LT-171-861 had high affinity for FLT3 protein. We also showed that LT-171-861 had an inhibitory effect on FLT3 mutants in cellular assays. Moreover, LT-171-861 had a growth-inhibitory effect on human AML cell lines harboring FLT3 internal tandem duplications (FLT3-ITD) such as FLT3-D835Y, FLT3­ITD-N676D, FLT3-ITD-D835Y, FLT3-ITD-F691L, FLT3-ITD-Y842C and AML blasts from patients with FLT3-ITD. Furthermore, LT-171-861 showed potent antileukemic efficacy against AML cells. We also show the efficacy of LT­171-861 in a subcutaneous implantation model and a bone marrow engrafted model in vivo, where administration of LT-171-861 led to almost complete tumor regression and increased survival. Conclusions: Overall, this study not only identifies LT-171-861 as a potent FLT3 inhibitor, but also provides a rationale for the upcoming clinical trial of LT-171-861 in patients with AML and FLT3-ITD mutations.


Subject(s)
Apoptosis/drug effects , Cell Proliferation/drug effects , Heterocyclic Compounds/pharmacology , Leukemia, Myeloid, Acute/drug therapy , Piperazines/pharmacology , Protein Kinase Inhibitors/pharmacology , Purines/pharmacology , fms-Like Tyrosine Kinase 3/antagonists & inhibitors , Animals , CSK Tyrosine-Protein Kinase/drug effects , Cell Line , Cell Line, Tumor , Extracellular Signal-Regulated MAP Kinases/drug effects , Female , Heterocyclic Compounds/therapeutic use , Humans , Inhibitory Concentration 50 , Janus Kinases/drug effects , Leukemia, Myeloid, Acute/genetics , Mice , Mice, Nude , Mice, SCID , Neoplasm Transplantation , Piperazines/therapeutic use , Protein Kinase Inhibitors/therapeutic use , Proto-Oncogene Proteins c-kit/drug effects , Purines/therapeutic use , THP-1 Cells , fms-Like Tyrosine Kinase 3/genetics
15.
Prostate ; 70(14): 1555-62, 2010 Oct 01.
Article in English | MEDLINE | ID: mdl-20687229

ABSTRACT

BACKGROUND: Benign prostatic hyperplasia (BPH) is associated with the proliferation of prostate tissue and an increase in smooth muscle tone. However, the way in which the hormonal environment affects cell proliferation and prostatic interstitial cells (PIC) responsible for the maintenance of the smooth muscle tone is not clear. The present study investigated the effect of estrogen supplementation on cell proliferation, androgen/estrogen ratio, and expression and/or distribution of PIC. METHODS: Male Sprague-Dawley rats were anesthetized with isoflurane/oxygen breathing mixture and subcutaneously implanted with silastic capsules. These capsules were either empty or filled with a 10 or 20 mg of crystalline estrogen. RESULTS: Estrogen exerted a potent effect on ventral prostate weight, which was manifested as a significant decrease between controls and the E(10)- and E(20)-treated rats. Active cell proliferation detected as Ki67-positive nuclei was observed in the stromal and epithelial cells of the ventral prostatic lobes from estrogen-treated rats and controls. Estrogen supplementation caused a significant and dose-dependent increase in prostatic estradiol and 5alpha-dihydrotestosterone (DHT) concentration but the ratios of either DHT/E(2) or E(2)/DHT were not significantly affected. PIC were observed in the region between the fibromuscular stroma and the glandular epithelium in all three experimental groups. E(20)-treated rats showed a higher expression of PIC than controls and E(10)-treated rats. CONCLUSIONS: The present study provides novel information regarding the synergistic role of estrogens and androgens in the prostate: estrogen may prevent prostatic hyperplasia via mechanisms other than affecting cell proliferation or DHT/estrogen ratio.


Subject(s)
Estrogens/pharmacology , Prostate/cytology , Prostate/physiology , Proto-Oncogene Proteins c-kit/genetics , Androgens/metabolism , Androgens/physiology , Animals , Body Weight/drug effects , Cell Division/drug effects , Dietary Supplements , Dihydrotestosterone/metabolism , Estradiol/metabolism , Estrogens/administration & dosage , Estrogens/metabolism , Estrogens/physiology , Ki-67 Antigen/drug effects , Ki-67 Antigen/metabolism , Leydig Cells/drug effects , Leydig Cells/physiology , Male , Organ Size/drug effects , Prostate/anatomy & histology , Prostate/drug effects , Prostatic Hyperplasia/pathology , Proto-Oncogene Proteins c-kit/drug effects , Proto-Oncogene Proteins c-kit/metabolism , Rats , Rats, Sprague-Dawley , Testosterone/metabolism
16.
Dig Dis Sci ; 55(12): 3315-27, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20725786

ABSTRACT

BACKGROUND: Gastrointestinal stromal tumors (GISTs) are relatively common mesenchymal tumors of the digestive tract characterized by c-KIT mutations. This is a comprehensive review of the current data of the literature on the various aspects of the diagnosis and treatment of these tumors. METHODS: The stomach is the most commonly involved site for these tumors in the digestive tract. Computed tomography and endoscopy can usually establish the diagnosis. The study of certain specific immunohistochemical markers may contribute to better characterization of these tumors. RESULTS: Surgical resection of GISTs has been the most effective therapy. In addition, targeted therapy with tyrosine kinase inhibitors may reduce the development of recurrence or decrease the disease progression in patients with metastatic disease. CONCLUSIONS: The introduction of tyrosine kinase inhibitors has resulted in significant improvement in the overall prognosis of these patients. Furthermore, preoperative imatinib can decrease tumor volume and is associated with complete surgical resection in locally advanced primary GISTs.


Subject(s)
Gastrointestinal Stromal Tumors/therapy , Antineoplastic Agents/therapeutic use , Benzamides , Biomarkers, Tumor , Diagnosis, Differential , Disease Progression , Endoscopy, Gastrointestinal , Gastrointestinal Stromal Tumors/diagnosis , Gastrointestinal Stromal Tumors/epidemiology , Gastrointestinal Stromal Tumors/metabolism , Gastrointestinal Stromal Tumors/pathology , Humans , Imatinib Mesylate , Immunohistochemistry , Neoplasm Recurrence, Local/epidemiology , Piperazines/therapeutic use , Proto-Oncogene Proteins c-kit/drug effects , Proto-Oncogene Proteins c-kit/physiology , Pyrimidines/therapeutic use , Receptor, Platelet-Derived Growth Factor alpha/drug effects , Receptor, Platelet-Derived Growth Factor alpha/physiology
17.
Przegl Lek ; 67(6): 413-6, 2010.
Article in Polish | MEDLINE | ID: mdl-21344772

ABSTRACT

Because of unsatisfactory treatment results in high risk neuroblastoma, it is necessary to find new, effective and safe, treatment options. Tyrosine kinase inhibitors, including imatinib, are one of the most profoundly examinated drugs. Its employment in neuroblastoma treatment is potentially possible because of expression of c-kit and PDGFR, which are cellular targets of imatinib. It was shown that this drug inhibits growth of neuroblastoma cell lines both in vivo and in vitro. The prognostic meaning of receptors expression is still not clear and depends on the method used for evaluation. The only found phase II study did not reveal the satisfactory response for imanitinib in treatment of resistant neuroblastoma, but the presence of receptors expression was not inclusion criteria. However, it seems that assuming the correct patient's eligibility criteria and proper dosage schedule, possible in combination with chemotherapy, imatinib may become safe and effective treatment modality.


Subject(s)
Biomarkers, Tumor/analysis , Neuroblastoma/diagnosis , Neuroblastoma/drug therapy , Piperazines/therapeutic use , Protein-Tyrosine Kinases/antagonists & inhibitors , Proto-Oncogene Proteins c-kit/analysis , Pyrimidines/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Benzamides , Cell Line, Tumor , Drug Resistance, Neoplasm , Humans , Imatinib Mesylate , Neuroblastoma/chemistry , Platelet-Derived Growth Factor/analysis , Platelet-Derived Growth Factor/drug effects , Prognosis , Proto-Oncogene Proteins c-kit/drug effects
18.
Aging (Albany NY) ; 13(1): 411-423, 2020 12 03.
Article in English | MEDLINE | ID: mdl-33290262

ABSTRACT

As the first clinical proteasome inhibitor, Bortezomib (BTZ) has been reported to improve the outcome of lymphoma. However, due to the unstable property, low bioavailability, and hydrophobic properties of BTZ, it is needed to develop effective drug delivery systems to deliver BTZ into targeted cells or organs. Here we developed a bortezomib (BTZ)-loaded HMSNs (BTZ@HMSNs) system, which can sustain the release of BTZ in targeted tissues. In vitro assays showed that BTZ@HMSNs limited cell proliferation and augmented apoptosis of lymphoma SNK-1 cells. Moreover, BTZ@HMSNs significantly diminished migration and invasion of SNK-1 cells as compared with BTZ. In contrast to the upregulation of SHP-1, BTZ@HMSNs decreased the mRNA levels of c-Kit, NF-κB, and JAK1, which elicit oncogenic role in lymphoma development. Importantly, lymphoma mice model showed that BTZ@HMSNs significantly activated p53 signaling and reduced tumor volume and weight compared with free BTZ. Our data thus demonstrate that BTZ@HMSNs manifests improved tumor-suppressing effect in vitro and in vivo compared to free BTZ. We believe that HMSNs is a promising strategy for delivering therapeutic agents for cancer treatment.


Subject(s)
Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Bortezomib/administration & dosage , Bortezomib/pharmacology , Cell Proliferation/drug effects , Lymphoma/drug therapy , Nanospheres , Animals , Cell Line, Tumor , Cell Movement/drug effects , Drug Carriers , Humans , In Vitro Techniques , Janus Kinase 1/drug effects , Janus Kinase 1/genetics , Lymphoma/genetics , Lymphoma/metabolism , Mice , Mice, Nude , NF-kappa B/drug effects , NF-kappa B/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 6/drug effects , Protein Tyrosine Phosphatase, Non-Receptor Type 6/genetics , Proto-Oncogene Proteins c-kit/drug effects , Proto-Oncogene Proteins c-kit/genetics , RNA, Messenger/drug effects , RNA, Messenger/metabolism , Silicon Dioxide , Tumor Suppressor Protein p53/drug effects , Tumor Suppressor Protein p53/metabolism , Xenograft Model Antitumor Assays
19.
Exp Hematol ; 36(10): 1266-77, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18657349

ABSTRACT

OBJECTIVE: Mutations of the receptor tyrosine kinase Kit occur in several human and canine cancers. While Kit inhibitors have activity in the clinical setting, they possess variable efficacy against particular forms of mutant Kit and drug resistance often develops over time. Inhibitors of heat shock protein 90 (HSP90), a chaperone for which Kit is a client protein, have demonstrated activity against human cancers and evidence suggests they downregulate several mutated and imatinib-resistant forms of Kit. The purpose of this study was to evaluate a novel HSP90 inhibitor, STA-9090, against wild-type (WT) and mutant Kit in canine bone marrow-derived cultured mast cells (BMCMCs), malignant mast cell lines, and fresh malignant mast cells. MATERIALS AND METHODS: BMCMCs, cell lines, and fresh malignant mast cells were treated with STA-9090, 17-AAG, and SU11654 and evaluated for loss in cell viability, cell death, alterations in HSP90 and Kit expression/signaling, and Kit mutation. STA-9090 activity was tested in a canine mastocytoma xenograft model. RESULTS: Treatment of BMCMCs, cell lines, and fresh malignant cells with STA-9090 induced growth inhibition, apoptosis that was caspase-3/7-dependent, and downregulation of phospho/total Kit and Akt, but not extracellular signal-regulated kinase (ERK) or phosphoinositide-3 kinase (PI-3K). Loss of Kit cell-surface expression was also observed. Furthermore, STA-9090 exhibited superior activity to 17-AAG and SU11654, and was effective against malignant mast cells expressing either WT or mutant Kit. Lastly, STA-9090 inhibited tumor growth in a canine mastocytoma mouse xenograft model. CONCLUSIONS: STA-9090 exhibits broad activity against mast cells expressing WT or mutant Kit, suggesting it may be an effective agent in the clinical setting against mast cell malignancies.


Subject(s)
Antineoplastic Agents/therapeutic use , HSP90 Heat-Shock Proteins/antagonists & inhibitors , Leukemia, Mast-Cell/drug therapy , Proto-Oncogene Proteins c-kit/genetics , Triazoles/therapeutic use , Animals , Apoptosis/drug effects , Cell Cycle/drug effects , Cell Line, Tumor , DNA Primers , Dog Diseases/pathology , Dogs , Mastocytoma/pathology , Mastocytoma/veterinary , Mice , Mice, SCID , Proto-Oncogene Proteins c-kit/drug effects , Reverse Transcriptase Polymerase Chain Reaction , Transplantation, Heterologous
20.
Medicine (Baltimore) ; 98(3): e14060, 2019 Jan.
Article in English | MEDLINE | ID: mdl-30653116

ABSTRACT

RATIONALE: Gastrointestinal stromal tumor (GIST) is the most common mesenchymal tumor of the gastrointestinal tract and is characterized by KIT mutations. Patientsresistant to 1st-line imatinib therapy are usually given sunitinib assecond-line treatment, which provides a median progression-free survival of 8 to 12 months. We report the 1st case of metastatic jejunum GIST with a KIT exon 11 deletion that showed complete response (CR) to sunitinib for more than 3 years. PATIENT CONCERNS: A 34-year-old man with advanced jejunum GIST was surgically treated upon initial diagnosis, and was histologically found to carry a high recurrence risk. Genetic testing revealed a KIT exon 11 deletion, and adjuvant therapy with imatinib was administered. The imatinib dose was escalated following recurrence in the abdomen, but the mass continued to grow. DIAGNOSIS: He was diagnosed with abdominal recurrence of GIST based on his medical history and histopathological results. INTERVENTION: Second-line sunitinib therapy was given. OUTCOMES: The mass disappeared, and CR was seen following 7 months of sunitinib therapy; this CR was sustained for more than 45 months. LESSONS: In cases of metastatic jejunum GIST with a KIT exon 11 deletion, sunitinib as second-line therapy can be used to achieve CR for more than 3 years.


Subject(s)
Antineoplastic Agents/administration & dosage , Gastrointestinal Stromal Tumors/drug therapy , Jejunal Neoplasms/drug therapy , Sunitinib/administration & dosage , Adult , Drug Administration Schedule , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , Exons , Gastrointestinal Stromal Tumors/genetics , Humans , Induction Chemotherapy , Jejunal Neoplasms/genetics , Male , Mutation , Proto-Oncogene Proteins c-kit/drug effects , Proto-Oncogene Proteins c-kit/genetics , Time Factors , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL