Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
1.
Br J Haematol ; 204(4): 1279-1287, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38131400

ABSTRACT

Primary vitreoretinal lymphoma (PVRL) is a rare subtype of malignant lymphoma with a poor prognosis because of high frequency of central nervous system (CNS) progression. Identification of factors associated with CNS progression is essential to improve the prognosis of patients with PVRL. We conducted a retrospective study of 54 patients diagnosed with PVRL and treated at our hospital to identify factors associated with CNS progression and prognosis. All patients were treated with intravitreal methotrexate (MTX) injections in the affected eyes until lesion resolution. Twenty-four patients were treated with systemic administration of high-dose MTX (systemic HD-MTX) every other week for a total of five cycles following intravitreal MTX injection. Of 24 patients, 20 completed five cycles of systemic HD-MTX. The 5-year cumulative incidence of CNS progression and overall survival (OS) rate were 78.0% and 69.0% respectively. By univariate and multivariate analyses, bilateral disease and the detection of B-cell clonality confirmed by flow cytometric analysis were risk factors associated with CNS progression. Moreover, systemic HD-MTX completion reduced the risk of CNS progression and was identified as a factor affecting OS. In this study, factors for CNS progression identified may potentially contribute to the optimized therapeutic stratification to improve the survival of patients with PVRL.


Subject(s)
Central Nervous System Neoplasms , Lymphoma , Retinal Neoplasms , Humans , Retinal Neoplasms/drug therapy , Retinal Neoplasms/pathology , Retrospective Studies , Vitreous Body/pathology , Lymphoma/drug therapy , Central Nervous System/pathology , Methotrexate
2.
Rinsho Ketsueki ; 64(11): 1426-1430, 2023.
Article in Japanese | MEDLINE | ID: mdl-38072429

ABSTRACT

A 43-year-old man presenting with oral bleeding was diagnosed with acute promyelocytic leukemia (APL). Induction chemotherapy consisting of all-trans retinoic acid and idarubicin was initiated, and disseminated intravascular coagulation (DIC) was treated with fresh frozen plasma and recombinant thrombomodulin infusions. The patient was free from neurological symptoms throughout the clinical course. However, cerebral hemorrhagic lesions were detected incidentally on magnetic resonance imaging performed to screen for leukemic central nervous system invasion at 2 weeks after treatment initiation. Imaging findings suggested subacute or later-phase cerebral hemorrhage. Platelet transfusions and other supportive care was provided. Serial imaging evaluations confirmed reduction of the hemorrhagic lesions. Hematological remission was achieved after induction chemotherapy, and no symptoms due to cerebral hemorrhage developed during the subsequent consolidation therapy. As patients with APL characteristically experience hemorrhagic events due to bleeding tendency caused by DIC, physicians should be aware of the possibility of asymptomatic cerebral hemorrhage in these patients.


Subject(s)
Disseminated Intravascular Coagulation , Hemorrhagic Disorders , Leukemia, Promyelocytic, Acute , Male , Humans , Adult , Leukemia, Promyelocytic, Acute/complications , Leukemia, Promyelocytic, Acute/drug therapy , Disseminated Intravascular Coagulation/etiology , Tretinoin/therapeutic use , Cerebral Hemorrhage/complications , Antineoplastic Combined Chemotherapy Protocols/therapeutic use
3.
Cancer Sci ; 114(9): 3719-3727, 2023 Sep.
Article in English | MEDLINE | ID: mdl-37365854

ABSTRACT

Methotrexate (MTX)-associated lymphoproliferative disorder (MTX-LPD) is a troublesome problem in patients receiving MTX for rheumatoid arthritis (RA). However, its incidence, prognosis, and risk factors remain unclear. In this retrospective study, we evaluated the actual incidence, prognostic impact, and risk factors of MTX-LPD. Of the 986 patients with RA treated with MTX, 90 patients experienced 95 new malignancies (NMs), with LPD as the most frequent in 26 patients. The cumulative LPD incidences were 1.3% and 4.7% at 5 and 10 years after MTX initiation, respectively. Among the 24 patients who discontinued MTX after developing LPD, 15 showed sustained regression, without difference in overall survival between patients with LPD and without NM. Inflammatory markers and absolute lymphocyte counts were not useful for early LPD development detection, but most of the patients with LPD had persistently elevated erythrocyte sedimentation ratios. Regarding concomitant drugs, tacrolimus increased the risk only if patients were not receiving biological disease-modifying antirheumatic drugs (bDMARDs). bDMARDs did not increase the risk for any of the drugs or the number of classes used. The number of LPD cases was lower in patients with IL-6A even after a long period after MTX, although with no statistically significant difference. Thus, approximately 1 in 20 patients with RA developed MTX-LPD over the 10 years of MTX treatment, but it did not affect the survival of patients with RA. Tacrolimus increased the risk of developing LPD for certain patients and should be used with caution.


Subject(s)
Antirheumatic Agents , Arthritis, Rheumatoid , Lymphoproliferative Disorders , Humans , Methotrexate/adverse effects , Retrospective Studies , Tacrolimus/therapeutic use , Arthritis, Rheumatoid/drug therapy , Arthritis, Rheumatoid/chemically induced , Arthritis, Rheumatoid/complications , Antirheumatic Agents/adverse effects , Lymphoproliferative Disorders/chemically induced , Lymphoproliferative Disorders/epidemiology
5.
Rinsho Ketsueki ; 63(4): 260-264, 2022.
Article in Japanese | MEDLINE | ID: mdl-35491214

ABSTRACT

Paroxysmal nocturnal hemoglobinuria (PNH) is characterized by hemolysis, thrombosis, and bone marrow failure. Infection, pregnancy, and surgical operation have the potential to evoke severe episodes of hemolysis and thrombosis. Therefore, the use of an antibody agent against complement component 5 (C5), eculizumab, one day before the operation is recommended. Ravulizumab is a newly approved long-acting antibody agent against C5. Thus, little is known about perioperative management with ravulizumab. We experienced a 43-year-old female patient who safely underwent laparoscopic cholecystectomy under ravulizumab treatment for PNH. Ravulizumab was administered one day before the operation. Laparoscopic cholecystectomy for cholelithiasis was performed under intravenous anesthesia, intermittent air compression of the lower extremities, and low pneumoperitoneum pressure. Additionally, heparin was administered, and the patient left the sickbed early without significant postoperative complications. Like eculizumab, complement inhibition by ravulizumab is also considered effective in the perioperative management of patients with PNH. However, close cooperation with surgeons and anesthesiologists and careful management based on clinical symptoms and laboratory data such as LDH, CH50, and D-dimer are essential.


Subject(s)
Cholecystectomy, Laparoscopic , Hemoglobinuria, Paroxysmal , Thrombosis , Adult , Antibodies, Monoclonal, Humanized , Cholecystectomy, Laparoscopic/adverse effects , Female , Hemoglobinuria, Paroxysmal/complications , Hemoglobinuria, Paroxysmal/drug therapy , Hemolysis , Humans , Pregnancy , Thrombosis/etiology
6.
Nihon Hoshasen Gijutsu Gakkai Zasshi ; 78(5): 502-510, 2022 May 20.
Article in Japanese | MEDLINE | ID: mdl-35354699

ABSTRACT

This study aimed to investigate the feasibility of estimating functional ischemia information from coronary artery computed tomography (CACT) data (i.e., morphological information). Fifty-five suspected ischemic heart disease patients were included in this study. To calculate the ischemic myocardium percentage (LV myocardial territories volume of distal portion the stenotic lesion/total LV myocardial volume) from CACT data with "coronary territories analysis, Ziostation2", and compared with the ischemic LV myocardium percentage and the functional flow reserve (FFR). The results showed that ischemic LV myocardium percentage was correlated with the FFR (r=-0.57). The median ischemic LV myocardium percentage of the FFR-positive group (n=33) was 37.1% (interquartile range [IQR] 33, 41.4%) and that of the FFR-negative group (n=22) was 24.8% (IQR 19.6, 30.6%). The ischemic LV myocardium percentage was significantly higher in the FFR-positive group (p<0.01) than in the FFR-negative group. The receiver operating characteristic (ROC) curve showed that the cutoff value for the ischemic LV myocardium percentage was 30%, with the sensitivity of 90.9% and the specificity of 77.3%. In conclusion, myocardial ischemia to diagnosis of FFR may occur when ischemic LV myocardium percentage is over than 30% and is unlikely to occur when it is less than 30%. This study suggests that the analysis of CACT data may contribute to the diagnosis of functional ischemia.


Subject(s)
Coronary Artery Disease , Coronary Stenosis , Fractional Flow Reserve, Myocardial , Myocardial Ischemia , Coronary Angiography/methods , Heart Ventricles/diagnostic imaging , Humans , Ischemia , Myocardial Ischemia/diagnostic imaging , Myocardium , Predictive Value of Tests , ROC Curve , Severity of Illness Index
7.
Int J Mol Med ; 49(4)2022 Apr.
Article in English | MEDLINE | ID: mdl-35119085

ABSTRACT

Anti­CD19 chimeric antigen receptor (CAR)­T cell therapy against refractory B­cell malignancies shows excellent therapeutic effects. However, there are some obstacles to be overcome in this treatment. Since current CAR­T cells target a single cell­surface protein on tumor cells, the CAR­T cells also attack normal cells expressing the protein. This is one of the major adverse effects of this therapy. To improve target­cell­specificity of this therapy, we established a novel CAR system, in which T­cell activation was controlled by expression patterns of proteins on target cells. Our novel CAR­T cells had two distinct CARs consisting of a 'Signal­CAR', recognizing a protein on tumor cells, and a 'Scissors­CAR', recognizing another protein on normal cells. The signal­CAR had a peptide sequence which was cleaved by the Scissors­CAR, and functional domains for cellular activation. The Scissors­CAR had a protease domain that cleaved its recognition peptide sequence in the Signal­CAR. When tumor cells expressed only the protein recognized by the Signal­CAR, the tumor cells were attacked. By contrast, normal cells expressing both the proteins induced inactivation of the Signal­CAR through cleavage of the recognition site when getting in contact with the CAR­T cells. To establish this system, we invented a Scissors­CAR that was dominantly localized on cell membranes and was activated only when the CAR­T cells were in contact with the normal cells. Using a T­cell line, Jurkat, and two proteins, CD19 and HER2, as target proteins, we showed that the anti­CD19­Signal­CAR was cleaved by the anti­HER2­Scissors­CAR when the CAR­T cells were co­cultivated with cells expressing both the proteins, CD19 and HER2. Furthermore, we demonstrated that primary CAR­T cells expressing both the CARs showed attenuated cytotoxicity againsT cells with both the target proteins. Our novel system would improve safety of the CAR­T cell therapy, leading to expansion of treatable diseases by this immunotherapy.


Subject(s)
Receptors, Chimeric Antigen , Antigens, CD19/genetics , Antigens, CD19/metabolism , Membrane Proteins/metabolism , Peptide Hydrolases/metabolism , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/metabolism , Receptors, Chimeric Antigen/genetics , Receptors, Chimeric Antigen/metabolism , T-Lymphocytes/metabolism
8.
Clin Case Rep ; 10(2): e05372, 2022 Feb.
Article in English | MEDLINE | ID: mdl-35154721

ABSTRACT

We report a case of fever of unknown origin in a patient with MDS associated with IgM-MGUS. The patient was positive for MYD88 mutation, and chemotherapy for LPL/WM improved the fever. Analysis of MYD88 and the effect of chemotherapy on LPL/WM finally revealed the latent LPL/WM in this case.

9.
In Vivo ; 35(6): 3407-3411, 2021.
Article in English | MEDLINE | ID: mdl-34697176

ABSTRACT

BACKGROUND: Neurotoxicity is one of the dangerous complications of chimeric antigen receptor (CAR) T-cell therapy, while its pathophysiology remains to be fully understood. Motor weakness not associated with central nervous system (CNS) toxicity has rarely been reported after CAR T-cell therapy. CASE REPORT: A 42-year-old female with a refractory diffuse large B-cell lymphoma received tisagenlecleucel (tisa-cel) and developed cytokine release syndrome (CRS) on day 3. She was treated with tocilizumab and methylprednisolone, which resolved CRS promptly. On day 7, motor weakness in lower extremities appeared, and she gradually became unable to walk without showing any other symptoms attributed to CNS disturbances. Whereas dexamethasone and tocilizumab were ineffective, neuropathy improved after high dose chemotherapy followed by autologous stem cell transplantation. Nerve conduction study (NCS) in lower extremities showed a decline in compound muscle action potential amplitude along with worsening of motor weakness, which was restored after improvement of symptoms. Based on symptoms and NCS, her motor weakness was thought to be due to disturbance in peripheral nerves. CONCLUSION: This study reports a patient who developed severe motor weakness due to disturbance in peripheral nerves after tisa-cel therapy. Neurotoxicity of non-CNS origin should also be noted in CAR T-cell therapy.


Subject(s)
Immunotherapy, Adoptive/adverse effects , Muscle Weakness/chemically induced , Peripheral Nerves , Receptors, Antigen, T-Cell , Adult , Cytokine Release Syndrome/chemically induced , Female , Hematopoietic Stem Cell Transplantation , Humans , Peripheral Nerves/drug effects , Peripheral Nerves/physiopathology , Transplantation, Autologous
10.
Front Immunol ; 12: 618081, 2021.
Article in English | MEDLINE | ID: mdl-33692791

ABSTRACT

As hematopoietic progenitors supply a large number of blood cells, therapeutic strategies targeting hematopoietic progenitors are potentially beneficial to eliminate unwanted blood cells, such as leukemic cells and immune cells causing diseases. However, due to their pluripotency, targeting those cells may impair the production of multiple cell lineages, leading to serious side effects such as anemia and increased susceptibility to infection. To minimize those side effects, it is important to identify monopotent progenitors that give rise to a particular cell lineage. Monocytes and monocyte-derived macrophages play important roles in the development of inflammatory diseases and tumors. Recently, we identified human monocyte-restricted progenitors, namely, common monocyte progenitors and pre-monocytes, both of which express high levels of CD64, a well-known monocyte marker. Here, we introduce a dimeric pyrrolobenzodiazepine (dPBD)-conjugated anti-CD64 antibody (anti-CD64-dPBD) that selectively induces the apoptosis of proliferating human monocyte-restricted progenitors but not non-proliferating mature monocytes. Treatment with anti-CD64-dPBD did not affect other types of hematopoietic cells including hematopoietic stem and progenitor cells, neutrophils, lymphocytes and platelets, suggesting that its off-target effects are negligible. In line with these findings, treatment with anti-CD64-dPBD directly killed proliferating monocytic leukemia cells and prevented monocytic leukemia cell generation from bone marrow progenitors of chronic myelomonocytic leukemia patients in a patient-derived xenograft model. Furthermore, by depleting the source of monocytes, treatment with anti-CD64-dPBD ultimately eliminated tumor-associated macrophages and significantly reduced tumor size in humanized mice bearing solid tumors. Given the selective action of anti-CD64-dPBD on proliferating monocyte progenitors and monocytic leukemia cells, it should be a promising tool to target cancers and other monocyte-related inflammatory disorders with minimal side effects on other cell lineages.


Subject(s)
Antineoplastic Agents, Immunological/pharmacology , Immunoconjugates/pharmacology , Monocyte-Macrophage Precursor Cells/drug effects , Animals , Antineoplastic Agents, Immunological/therapeutic use , Humans , Immunoconjugates/therapeutic use , Immunophenotyping , Mice , Mice, Knockout , Mice, Transgenic , Monocyte-Macrophage Precursor Cells/metabolism , Monocytes/drug effects , Monocytes/metabolism , THP-1 Cells , Tumor-Associated Macrophages/drug effects , Tumor-Associated Macrophages/metabolism
11.
Intern Med ; 60(8): 1169-1174, 2021 Apr 15.
Article in English | MEDLINE | ID: mdl-33229801

ABSTRACT

Objective Patients with hematological malignancies, particularly those with multiple myeloma, often suffer from pathological vertebral compression fractures (VCFs). Consequent and significant spinal pain and paralysis impair the activities of daily living and quality of life and delay subsequent chemotherapy. Balloon kyphoplasty (BKP), which is less invasive than conventional therapies, is a type of percutaneous vertebroplasty in which cement is injected into the broken vertebrae to stabilize the spinal column. The present study assessed the effect of BKP on hematological tumors. Methods We retrospectively analyzed five myeloma patients and one lymphoma patient who underwent BKP for pathological VCFs in our institution. Results The median age was 74 years old. The spinal operation level ranged from T2 to L4. BKP was performed at the diagnosis in two cases, after first-line chemotherapy in one case, and after subsequent chemotherapy in three cases. After approximately 1 month, the patients' average Eastern Cooperative Oncology Group performance status score rapidly improved from 3.2 to 1.3. The numeric rating scale score decreased from 8.8 to 2.0, and the Karnofsky Performance Status score increased from 35 to 75. No severe complications were observed. All patients became able to walk unassisted and underwent early subsequent chemotherapy. Conclusion BKP can be a safe and effective treatment option for pathological VCFs in patients with hematological malignancies and allows for rapid induction with subsequent chemotherapy.


Subject(s)
Fractures, Compression , Hematologic Neoplasms , Kyphoplasty , Osteoporotic Fractures , Spinal Fractures , Activities of Daily Living , Aged , Fractures, Compression/etiology , Fractures, Compression/surgery , Hematologic Neoplasms/complications , Hematologic Neoplasms/therapy , Humans , Kyphoplasty/adverse effects , Quality of Life , Retrospective Studies , Spinal Fractures/etiology , Spinal Fractures/surgery , Treatment Outcome
12.
Cancers (Basel) ; 12(2)2020 Feb 10.
Article in English | MEDLINE | ID: mdl-32050632

ABSTRACT

JAK2-V617F plays a key role in the pathogenesis of myeloproliferative neoplasm. However, its inhibitor ruxolitinib has shown limited clinical efficacies because of the ruxolitinib-persistent proliferation of JAK2-V617F-positive cells. We here demonstrate that the USP9X inhibitor WP1130 or EOAI3402143 (G9) inhibited proliferation and induced apoptosis more efficiently in cells dependent on JAK2-V617F than on cytokine-activated JAK2. WP1130 preferentially downregulated activated and autophosphorylated JAK2-V617F by enhancing its K63-linked polyubiquitination and inducing its aggresomal translocation to block downstream signaling. Furthermore, JAK2-V617F associated physically with USP9X in leukemic HEL cells. Induction of apoptosis by inhibition of USP9X was mediated through the intrinsic mitochondria-mediated pathway, synergistically enhanced by BH3 mimetics, prevented by overexpression of Bcl-xL, and required oxidative stress to activate stress-related MAP kinases p38 and JNK as well as DNA damage responses in HEL cells. Although autophosphorylated JAK2-V617F was resistant to WP1130 in the ruxolitinib-persistent HEL-R cells, these cells expressed Bcl-2 and Bcl-xL at lower levels and showed an increased sensitivity to WP1130 as well as BH3 mimetics as compared with ruxolitinib-naive HEL cells. Thus, USP9X represents a promising target along with anti-apoptotic Bcl-2 family members for novel therapeutic strategies against JAK2-V617F-positive myeloproliferative neoplasms, particularly under the ruxolitinib persistence conditions.

13.
Cancers (Basel) ; 11(12)2019 Nov 20.
Article in English | MEDLINE | ID: mdl-31756944

ABSTRACT

FLT3-ITD is the most frequent tyrosine kinase mutation in acute myeloid leukemia (AML) associated with poor prognosis. We previously found that FLT3-ITD activates the mTORC1/S6K/4EBP1 pathway cooperatively through the STAT5/PIM and PI3K/AKT pathways to promote proliferation and survival by enhancing the eIF4F complex formation required for cap-dependent translation. Here, we show that, in contrast to BCR/ABL causing Ph-positive leukemias, FLT3-ITD distinctively activates the serine/threonine kinases RSK1/2 through activation of the MEK/ERK pathway and PDK1 to transduce signals required for FLT3-ITD-dependent, but not BCR/ABL-dependent, proliferation and survival of various cells, including MV4-11. Activation of the MEK/ERK pathway by FLT3-ITD and its negative feedback regulation by RSK were mediated by Gab2/SHP2 interaction. RSK1 phosphorylated S6RP on S235/S236, TSC2 on S1798, and eIF4B on S422 and, in cooperation with PIM, on S406, thus activating the mTORC1/S6K/4EBP1 pathway and eIF4B cooperatively with PIM. RSK1 also phosphorylated Bad on S75 and downregulated BIM-EL in cooperation with ERK. Furthermore, inhibition of RSK1 increased sensitivities to BH3 mimetics inhibiting Mcl-1 or Bcl-2 and induced activation of Bax, leading to apoptosis, as well as inhibition of proliferation synergistically with inhibition of PIM or PI3K. Thus, RSK1 represents a promising target, particularly in combination with PIM or PI3K, as well as anti-apoptotic Bcl-2 family members, for novel therapeutic strategies against therapy-resistant FLT3-ITD-positive AML.

14.
Cancer Lett ; 453: 84-94, 2019 07 01.
Article in English | MEDLINE | ID: mdl-30946869

ABSTRACT

FLT3-ITD and FLT3-TKD are the most frequent mutations in acute myeloid leukemia (AML) with the former associated with a poor prognosis. Here we show that inhibition of the deubiquitinase USP9X by its inhibitor WP1130 or EOAI3402143 (G9) induces apoptosis preferentially in cells transformed by these mutant kinases, including FLT3-ITD-positive AML cell line MV4-11 and primary AML cells. Mechanistically, WP1130 induced aggresomal translocation of the mutant kinases, particularly FLT3-ITD in its activated and autophosphorylated conformation, to block the downstream signaling events, which was aggravated by knock down of USP9X. Moreover, USP9X physically associated with FLT3-ITD to inhibit its K63-linked polyubiquitination, while FLT3-ITD induced tyrosine phosphorylation and degradation of USP9X through the ubiquitin/proteasome pathway. WP1130 or G9 also induced oxidative stress to stimulate stress-related MAP kinase pathways and DNA damage responses to activate in cooperation with inhibition of FLT3-ITD signaling the intrinsic mitochondria-mediated apoptotic pathway, which was synergistically enhanced by BH3 mimetics and prevented by overexpression of Bcl-xL or Mcl-1. Thus, USP9X represents a promising target for novel therapies against therapy-resistant FLT3-ITD-positive AML.


Subject(s)
Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/metabolism , Ubiquitin Thiolesterase/antagonists & inhibitors , fms-Like Tyrosine Kinase 3/metabolism , Apoptosis/drug effects , Cell Line, Tumor , Cyanoacrylates/pharmacology , Down-Regulation/drug effects , HL-60 Cells , Humans , K562 Cells , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/pathology , Mutation , Myeloid Cell Leukemia Sequence 1 Protein/metabolism , Oxidative Stress/physiology , Phosphorylation , Pyridines/pharmacology , Signal Transduction/drug effects , U937 Cells , Ubiquitin Thiolesterase/genetics , Ubiquitin Thiolesterase/metabolism , Ubiquitination
15.
Transl Oncol ; 12(2): 336-349, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30472492

ABSTRACT

FLT3-ITD and FLT3-TKD are the most frequent tyrosine kinase mutations in acute myeloid leukemia (AML), with the former conferring a poor prognosis. We have recently revealed that FLT3-ITD confers resistance to the PI3K/AKT pathway inhibitors by protecting the mTORC1/4EBP1/Mcl-1 pathway through Pim kinases induced by STAT5 activation in AML. The proteasome inhibitor bortezomib has recently been reported as a promising agent for treatment of AML. Here, we show that the proteasome inhibitor bortezomib as well as carfilzomib induces apoptosis through the intrinsic pathway more conspicuously in cells transformed by FLT3-TKD than FLT3-ITD. Mechanistically, bortezomib upregulated the stress-regulated protein REDD1 and induced downregulation of the mTORC1 pathway more distinctively in cells transformed by FLT3-TKD than FLT-ITD, while overexpression of Pim-1 partly prevented this downregulation and apoptosis in FLT3-TKD-transformed cells. Genetic enhancement of the REDD1 induction or pharmacological inhibition of STAT5, Pim kinases, mTORC1, or S6K by specific inhibitors, such as pimozide, AZD1208, PIM447, rapamycin, and PF-4708671, accelerated the downregulation of mTORC1/Mcl-1 pathway to enhance bortezomib-induced apoptosis in FLT3-ITD-expressing cells, including primary AML cells, while overexpression of Mcl-1 prevented induction of apoptosis. Thus, FLT3-ITD confers a resistance to the proteasome inhibitors on AML cells by protecting the mTORC1/Mcl-1 pathway through the STAT5/Pim axis, and inhibition of these signaling events remarkably enhances the therapeutic efficacy.

16.
Oncotarget ; 9(42): 26834-26851, 2018 Jun 01.
Article in English | MEDLINE | ID: mdl-29928488

ABSTRACT

The activated JAK2-V617F mutant is very frequently found in myeloproliferative neoplasms (MPNs), and its inhibitor ruxolitinib has been in clinical use, albeit with limited efficacies. Here, we examine the signaling mechanisms from JAK2-V617F and responses to ruxolitinib in JAK2-V617F-positive leukemic cell lines, including PVTL-2, newly established from a patient with post-MPN secondary acute myeloid leukemia, and the widely used model cell line HEL. We have found that ruxolitinib downregulated the mTORC1/S6K/4EBP1 pathway at least partly through inhibition of the STAT5/Pim-2 pathway with concomitant downregulation of c-Myc, MCL-1, and BCL-xL as well as induction of autophagy in these cells. Ruxolitinib very efficiently inhibited proliferation but only modestly induced apoptosis. However, inhibition of BCL-xL/BCL-2 by the BH3 mimetics ABT-737 and navitoclax or BCL-xL by A-1331852 induced caspase-dependent apoptosis involving activation of Bak and Bax synergistically with ruxolitinib in HEL cells. On the other hand, the putative pan-BH3 mimetic obatoclax as well as chloroquine and bafilomycin A1 inhibited autophagy at its late stage and induced apoptosis in PVTL-2 cells synergistically with ruxolitinib. The present study suggests that autophagy as well as the anti-apoptotic BCL-2 family members, regulated at least partly by the mTORC1 pathway downstream of STAT5/Pim-2, protects JAK2-V617F-positive leukemic cells from ruxolitinib-induced apoptosis depending on cell types and may contribute to development of new strategies against JAK2-V617F-positive neoplasms.

17.
Oncotarget ; 9(10): 8870-8886, 2018 Feb 06.
Article in English | MEDLINE | ID: mdl-29507660

ABSTRACT

FLT3-ITD is the most frequent tyrosine kinase mutation in acute myeloid leukemia (AML) associated with poor prognosis. We previously reported that activation of STAT5 confers resistance to PI3K/Akt inhibitors on the FLT3-ITD-positive AML cell line MV4-11 and 32D cells driven by FLT3-ITD (32D/ITD) but not by FLT3 mutated in the tyrosine kinase domain (32D/TKD). Here, we report the involvement of Pim kinases expressed through STAT5 activation in acquisition of this resistance. The specific pan-Pim kinase inhibitor AZD1208 as well as PIM447 in combination with the PI3K inhibitor GDC-0941 or the Akt inhibitor MK-2206 cooperatively downregulated the mTORC1/4EBP1 pathway, formation of the eIF4E/eIF4G complex, and Mcl-1 expression leading to activation of Bak and Bax to induce caspase-dependent apoptosis synergistically in these cells. These cooperative effects were enhanced or inhibited by knock down of mTOR or expression of its activated mutant, respectively. Overexpression of Mcl-1 conferred the resistance on 32D/ITD cells to combined inhibition of the PI3K/Akt pathway and Pim kinases, while the Mcl-1-specific BH3 mimetic A-1210477 conquered the resistance of MV4-11 cells to GDC-0941. Furthermore, overexpression of Pim-1 in 32D/TKD enhanced the mTORC1/Mcl-1 pathway and partially protected it from the PI3K/Akt inhibitors or the FLT3 inhibitor gilteritinib to confer the resistance to PI3K/Akt inhibitors. Finally, AZD1208 and GDC-0941 cooperatively inhibited the mTORC1/Mcl-1 pathway and reduced viable cell numbers of primary AML cells from some FLT3-ITD positive cases. Thus, Pim kinases may protect the mTORC1/4EBP1/Mcl-1 pathway to confer the resistance to the PI3K/Akt inhibitors on FLT3-ITD cells and represent promising therapeutic targets.

18.
Gan To Kagaku Ryoho ; 45(13): 2054-2056, 2018 Dec.
Article in Japanese | MEDLINE | ID: mdl-30692282

ABSTRACT

This study aimed to evaluate the surgical treatment outcome of patients with colorectal cancer having ulcerative colitis (UC). METHODS: We examined 112 patients who underwent the surgery from 1998 to 2016. Among the 112 patients, 14 were complicated with malignant tumor(9 with colorectal cancer and 5 with dysplasia-assciated lesion or mass)We performed group comparison between patients with(group A)and without(group B)colorectal cancer. RESULTS: The median age in group A was 58.1 years, which was significantly older than that in group B. The duration of disease in group A was 13.2 years, which was significantly longer than that in group B. Surgical procedures in group B were as follows; 5 cases had ileoanalanastomosis (IAA), 4 cases had ileo-analcanalanastomosis (IACA), 2 cases had ileorectal anastomosis(IRA), and 1 case had interspincteric resection(ISR) and abdominoperinealresection (APR)Laparoscopic sigmoid colectomy was performed in a patient aged 52 years because he was diagnosed with sporadic sigmoid colon cancer. Periodic endoscopic screening was not performed in 5 patients with pT2 colorectalcancer in group B. endoscopic submucosaldissection (ESD)was performed in 2 patients aged>70 years with rectal pTis cancer. CONCLUSION: Elderly patients with UC were often complicated with colorectalcancer, and these patients are indicated for surgicalprocedures other than IAA to preserve defecation function. Patients with UC should undergo periodic endoscopic screening for early detection of malignant tumor and definitive surgicalresection.


Subject(s)
Colitis, Ulcerative , Colorectal Neoplasms , Aged , Anastomosis, Surgical , Colectomy , Colitis, Ulcerative/complications , Colorectal Neoplasms/complications , Colorectal Neoplasms/surgery , Humans , Male , Middle Aged , Rectum , Retrospective Studies , Treatment Outcome
19.
Gan To Kagaku Ryoho ; 45(13): 2075-2077, 2018 Dec.
Article in Japanese | MEDLINE | ID: mdl-30692289

ABSTRACT

A 60s man with a history of laparoscopic distal gastrectomy(LDG)of gastric cancer in January 2015was followed up in an outpatient clinic. He remained healthy without recurrence for about 2 years, but in February 2017, colonoscopy identified an elevated lesion that covered one-third of the circumference in the ascending colon. Biopsy revealed that this lesion had poorly differentiated adenocarcinoma. No other metastatic lesions were noted on thoraco-abdominal CT, and tumor markers were not elevated in a blood test. Right hemicolectomy with D3 nodal dissection was performed on March 2017. Although regional lymph node metastasis was noted during surgery, distant metastasis was not identified. Histological examination revealed that the ascending colon tumor had poorly differentiated adenocarcinoma very similar to that of the primary gastric cancer, and the lesion was diagnosed as metastasis from previous gastric carcinoma. Pathological findings showed pPM0, pDM0, pRM0, pCY0, and definitive surgical treatment was confirmed. However, 2 months after the surgery, ascites caused by peritoneal and bilateral supraclavicular lymph node metastases were detected on CT. He passed away 6months after the last surgery. We report a case of metachronous ascending colon metastasis from gastric cancer that was difficult to preoperatively diagnose.


Subject(s)
Adenocarcinoma , Colonic Neoplasms , Stomach Neoplasms , Adenocarcinoma/secondary , Colon, Ascending , Colonic Neoplasms/secondary , Humans , Male , Middle Aged , Neoplasm Recurrence, Local , Stomach Neoplasms/pathology
20.
J Biol Chem ; 292(48): 19639-19655, 2017 12 01.
Article in English | MEDLINE | ID: mdl-28974577

ABSTRACT

Platelet endothelial cell adhesion molecule 1 (PECAM-1) is a cell adhesion protein involved in the regulation of cell adhesion and migration. Interestingly, several PECAM-1-deficient hematopoietic cells exhibit impaired chemotactic responses to stromal cell-derived factor 1 (SDF-1), a chemokine essential for B lymphopoiesis and bone marrow myelopoiesis. However, whether PECAM-1 is involved in SDF-1-regulated chemotaxis is unknown. We report here that SDF-1 induces tyrosine phosphorylation of PECAM-1 at its immunoreceptor tyrosine-based inhibition motifs in several hematopoietic cell lines via the Src family kinase Lyn, Bruton's tyrosine kinase, and JAK2 and that inhibition of these kinases reduced chemotaxis. Overexpression and knockdown of PECAM-1 enhanced and down-regulated, respectively, SDF-1-induced Gαi-dependent activation of the PI3K/Akt/mTORC1 pathway and small GTPase Rap1 in hematopoietic 32Dcl3 cells, and these changes in activation correlated with chemotaxis. Furthermore, pharmacological or genetic inhibition of the PI3K/Akt/mTORC1 pathway or Rap1, respectively, revealed that these pathways are independently activated and required for SDF-1-induced chemotaxis. When coexpressed in 293T cells, PECAM-1 physically associated with the SDF-1 receptor CXCR4. Moreover, PECAM-1 overexpression and knockdown reduced and enhanced SDF-1-induced endocytosis of CXCR4, respectively. Furthermore, when expressed in 32Dcl3 cells, an endocytosis-defective CXCR4 mutant, CXCR4-S324A/S325A, could activate the PI3K/Akt/mTORC1 pathway as well as Rap1 and induce chemotaxis in a manner similar to PECAM-1 overexpression. These findings suggest that PECAM-1 enhances SDF-1-induced chemotaxis by augmenting and prolonging activation of the PI3K/Akt/mTORC1 pathway and Rap1 and that PECAM-1, at least partly, exerts its activity by inhibiting SDF-1-induced internalization of CXCR4.


Subject(s)
Chemokine CXCL12/physiology , Leukocytes/physiology , Platelet Endothelial Cell Adhesion Molecule-1/physiology , Animals , Bone Marrow Cells/metabolism , Cell Line , Mice , Phosphorylation , Platelet Endothelial Cell Adhesion Molecule-1/chemistry , Protein-Tyrosine Kinases/metabolism , Receptors, CXCR4/metabolism , Tyrosine/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...