Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 192
Filter
2.
J Neurol ; 269(7): 3937-3958, 2022 Jul.
Article in English | MEDLINE | ID: mdl-35399125

ABSTRACT

Autologous haematopoietic stem cell transplantation (aHSCT) is gaining traction as a valuable treatment option for patients affected by severe multiple sclerosis (MS), particularly the relapsing-remitting form. We describe the current literature in terms of clinical trials, observational and retrospective studies, as well as immune reconstitution following transplantation, with a focus on the conditioning regimens used for transplantation. The evidence base predominantly consists of non-randomised, uncontrolled clinical trials or data from retrospective or observational cohorts, i.e. very few randomised or controlled trials. Most often, intermediate-intensity conditioning regimens are used, with promising results from both myeloablative and lymphoablative strategies, as well as from regimens that are low and high intensity. Efficacy of transplantation, which is likely secondary to immune reconstitution and restored immune tolerance, is, therefore, not clearly dependent on the intensity of the conditioning regimen. However, the conditioning regimen may well influence the immune response to transplantation. Heterogeneity of conditioning regimens among studies hinders synthesis of the articles assessing post-aHSCT immune system changes. Factors associated with better outcomes were lower Kurtzke Expanded Disability Status Scale, relapsing-remitting MS, younger age, and shorter disease duration at baseline, which supports the guidance for patient selection proposed by the European Society for Blood and Marrow Transplantation. Interestingly, promising outcomes were described for patients with secondary progressive MS by some studies, which may be worth taking into account when considering treatment options for patients with active, progressive disease. Of note, a significant proportion of patients develop autoimmune disease following transplantation, with alemtuzumab-containing regimens associated with the highest incidence.


Subject(s)
Hematopoietic Stem Cell Transplantation , Multiple Sclerosis , Hematopoietic Stem Cell Transplantation/adverse effects , Humans , Multiple Sclerosis/etiology , Multiple Sclerosis/surgery , Retrospective Studies , Transplantation, Autologous/methods , Treatment Outcome
3.
Life Sci ; 287: 120128, 2021 Dec 15.
Article in English | MEDLINE | ID: mdl-34774874

ABSTRACT

Glioblastoma is the most frequent and malignant brain tumor. The median survival for this disease is approximately 15 months, and despite all the available treatment strategies employed, it remains an incurable disease. Preclinical and clinical research have shown that the resistance process related to DNA damage repair pathways, glioma stem cells, blood-brain barrier selectivity, and dose-limiting toxicity of systemic treatment leads to poor clinical outcomes. In this context, the advent of drug delivery systems associated with localized treatment seems to be a promising and versatile alternative to overcome the failure of the current treatment approaches. In order to bypass therapeutic tumor resistance mechanisms, more effective combinatorial therapies should be identified, such as the use of cytotoxic drugs combined with the inhibition of DNA damage response (DDR)-related targets. Additionally, critical reasoning about the delivery approach and administration route in brain tumors treatment innovation is essential. The outcomes of future experimental studies regarding the association of delivery systems, alternative treatment routes, and DDR targets are expected to lead to the development of refined therapeutic interventions. Novel therapeutic approaches could improve the life's quality of glioblastoma patients and increase their survival rate.


Subject(s)
Antineoplastic Agents/administration & dosage , Brain Neoplasms/drug therapy , DNA Damage/drug effects , Drug Delivery Systems/trends , Drug Development/trends , Glioblastoma/drug therapy , Animals , Antineoplastic Agents/metabolism , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/metabolism , Brain Neoplasms/metabolism , DNA Damage/physiology , Drug Delivery Systems/methods , Drug Development/methods , Glioblastoma/metabolism , Humans
4.
Strahlenther Onkol ; 196(12): 1096-1102, 2020 12.
Article in English | MEDLINE | ID: mdl-33125504

ABSTRACT

PURPOSE: The coronavirus pandemic is affecting global health systems, endangering daily patient care. Hemato-oncological patients are particularly vulnerable to infection, requiring decisive recommendations on treatment and triage. The aim of this survey amongst experts on radiation therapy (RT) for lymphoma and leukemia is to delineate typical clinical scenarios and to provide counsel for high-quality care. METHODS: A multi-item questionnaire containing multiple-choice and free-text questions was developed in a peer-reviewed process and sent to members of the radiation oncology panels of the German Hodgkin Study Group and the German Lymphoma Alliance. Answers were assessed online and analyzed centrally. RESULTS: Omission of RT was only considered in a minority of cases if alternative treatment options were available. Hypofractionated regimens and reduced dosages may be used for indolent lymphoma and fractures due to multiple myeloma. Overall, there was a tendency to shorten RT rather than to postpone or omit it. Even in case of critical resource shortage, panelists agreed to start emergency RT for typical indications (intracranial pressure, spinal compression, superior vena cava syndrome) within 24 h. Possible criteria to consider for patient triage are the availability of (systemic) options, the underlying disease dynamic, and the treatment rationale (curative/palliative). CONCLUSION: RT for hemato-oncological patients receives high-priority and should be maintained even in later stages of the pandemic. Hypofractionation and shortened treatment schedules are feasible options for well-defined constellations, but have to be discussed in the clinical context.


Subject(s)
COVID-19/epidemiology , Lymphoma/radiotherapy , Multiple Myeloma/radiotherapy , Pandemics , Radiation Oncology/standards , SARS-CoV-2/isolation & purification , Triage/standards , Appointments and Schedules , COVID-19/complications , COVID-19/diagnosis , COVID-19/prevention & control , COVID-19 Testing , Cross Infection/prevention & control , Diagnosis, Differential , Dose Fractionation, Radiation , Humans , Hygiene/standards , Infection Control/methods , Infection Control/standards , Lymphoma/complications , Lymphoma/drug therapy , Multiple Myeloma/complications , Osteolysis/etiology , Osteolysis/radiotherapy , Personal Protective Equipment , Radiation Oncology/methods , Radiation Pneumonitis/diagnosis , Superior Vena Cava Syndrome/etiology , Superior Vena Cava Syndrome/radiotherapy , Surveys and Questionnaires , Time-to-Treatment , Whole-Body Irradiation
5.
Stem Cell Rev Rep ; 16(4): 776-791, 2020 08.
Article in English | MEDLINE | ID: mdl-32556945

ABSTRACT

Mesenchymal stromal cells (MSCs) are promising candidates for cell-based therapies, mainly due to their unique biological properties such as multipotency, self-renewal and trophic/immunomodulatory effects. However, clinical use has proven complex due to limitations such as high variability of MSCs preparations and high number of cells required for therapies. These challenges could be circumvented with cell immortalization through genetic manipulation, and although many studies show that such approaches are safe, little is known about changes in other biological properties and functions of MSCs. In this study, we evaluated the impact of MSCs immortalization with the TERT gene on the purinergic system, which has emerged as a key modulator in a wide variety of pathophysiological conditions. After cell immortalization, MSCs-TERT displayed similar immunophenotypic profile and differentiation potential to primary MSCs. However, analysis of gene and protein expression exposed important alterations in the purinergic signaling of in vitro cultured MSCs-TERT. Immortalized cells upregulated the CD39/NTPDase1 enzyme and downregulated CD73/NT5E and adenosine deaminase (ADA), which had a direct impact on their nucleotide/nucleoside metabolism profile. Despite these alterations, adenosine did not accumulate in the extracellular space, due to increased uptake. MSCs-TERT cells presented an impaired in vitro immunosuppressive potential, as observed in an assay of co-culture with lymphocytes. Therefore, our data suggest that MSCs-TERT have altered expression of key enzymes of the extracellular nucleotides/nucleoside control, which altered key characteristics of these cells and can potentially change their therapeutic effects in tissue engineering in regenerative medicine.


Subject(s)
Adenosine/metabolism , Immunosuppression Therapy , Mesenchymal Stem Cells/cytology , Telomerase/metabolism , 5'-Nucleotidase/metabolism , Adenosine Deaminase/metabolism , Adenosine Triphosphate/metabolism , Animals , Antigens, CD , Apyrase , Cell Differentiation , Cell Line, Transformed , Extracellular Space/chemistry , Gene Expression Regulation , Humans , Jurkat Cells , Rats, Wistar , Telomerase/genetics
6.
Cancer Immunol Immunother ; 69(9): 1801-1812, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32350590

ABSTRACT

Immunotherapy as an approach for cancer treatment is clinically promising. CD73, which is the enzyme that produces extracellular adenosine, favors cancer progression and protects the tumor from immune surveillance. While CD73 has recently been demonstrated to be a potential target for glioma treatment, its role in regulating the inflammatory tumor microenvironment has not yet been investigated. Thus, this study explores the immunotherapeutic value of the CD73 blockade in glioblastoma. The immuno-therapeutic value of the CD73 blockade was evaluated in vivo in immunocompetent pre-clinical glioblastoma model. As such, glioblastoma-bearing rats were nasally treated for 15 days with a siRNA CD73-loaded cationic-nanoemulsion (NE-siRNA CD73R). Apoptosis was determined by flow cytometry using Annexin-V staining and cell proliferation was analyzed by Ki67 expression by immunohistochemistry. The frequencies of the CD4+, CD8+, and CD4+CD25highCD39+ (Treg) T lymphocytes; CD11b+CD45high macrophages; CD11b+CD45low-microglia; and CD206+-M2-like phenotypes, along with expression levels of CD39 and CD73 in tumor and tumor-associated immune cells, were determined using flow cytometry, while inflammatory markers associated with tumor progression were evaluated using RT-qPCR. The CD73 blockade by NE-siRNA CD73 was found to induce tumor cell apoptosis. Meanwhile, the population of Tregs, microglia, and macrophages was significantly reduced in the tumor microenvironment, though IL-6, CCL17, and CCL22 increased. The treatment selectively decreased CD73 expression in the GB cells as well as in the tumor-associated-macrophages/microglia. This study indicates that CD73 knockdown using a nanotechnological approach to perform nasal delivery of siRNA-CD73 to CNS can potentially regulate the glioblastoma immune microenvironment and delay tumor growth by inducing apoptosis.


Subject(s)
5'-Nucleotidase/antagonists & inhibitors , 5'-Nucleotidase/immunology , Cell Proliferation/physiology , Glioblastoma/immunology , Glioblastoma/metabolism , Glioma/immunology , Glioma/metabolism , Adenosine/immunology , Adenosine/metabolism , Animals , Brain Neoplasms/immunology , Brain Neoplasms/metabolism , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cell Line, Tumor , Immunohistochemistry/methods , Immunotherapy/methods , Macrophages/immunology , Macrophages/metabolism , Microglia/immunology , Microglia/metabolism , Rats
7.
Cancer Chemother Pharmacol ; 85(6): 1177-1182, 2020 06.
Article in English | MEDLINE | ID: mdl-32417936

ABSTRACT

Glioblastoma is the most devastating primary brain tumor and effective therapies are not available. Treatment is based on surgery followed by radio and chemotherapy with temozolomide (TMZ), but TMZ increases patient survival only by 2 months. CD73, an enzyme responsible for adenosine production, emerges as a target for glioblastoma treatment. Indeed, adenosine causes tumor-promoting actions and CD73 inhibition increases sensitivity to TMZ in vitro. Here, a cationic nanoemulsion to nasal delivery of siRNA CD73 (NE-siRNA CD73) aiming glioblastoma treatment was employed alone or in combination with TMZ. In vitro, two glioblastoma cell lines (C6 and U138MG) with a chemo-resistant profile were used. Treatment alone with NE-siRNA CD73 reduced C6 and U138MG glioma cell viability by 70% and 25%, respectively. On the other hand, when NE-siRNA + TMZ combined treatment was employed, a reduction of 85% and 33% of cell viability was observed. Notably, treatment with NE-siRNA CD73 of glioma-bearing Wistar rats reduced tumor size by 80%, 60% more than the standard chemotherapy with TMZ, but no synergistic or additive effect was observed in vivo. Additionally, NE-siRNA CD73, TMZ or combined therapy decreased adenosine levels in liquor confirming the importance of this nucleoside on in vivo GB growth. Finally, no hemolytic potential was observed. These results suggest that nasal administration of NE-siRNA CD73 exhibits higher antiglioma effect when compared to TMZ. However, no synergistic or additive in vivo was promoted by the therapeutic regimen employed in this study.


Subject(s)
5'-Nucleotidase/antagonists & inhibitors , Brain Neoplasms/drug therapy , Drug Resistance, Neoplasm/genetics , Glioblastoma/drug therapy , RNA, Small Interfering/genetics , Temozolomide/pharmacology , 5'-Nucleotidase/genetics , Animals , Antineoplastic Agents, Alkylating/pharmacology , Apoptosis , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Cell Proliferation , Drug Evaluation, Preclinical , Glioblastoma/genetics , Glioblastoma/pathology , Humans , Male , RNA, Small Interfering/administration & dosage , Rats , Rats, Wistar , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
8.
Mol Neurobiol ; 57(2): 635-649, 2020 Feb.
Article in English | MEDLINE | ID: mdl-31407144

ABSTRACT

Glioblastoma is the most devastating primary brain tumor. Effective therapies are not available, mainly due to high tumor heterogeneity, chemoresistance, and the difficulties imposed by blood-brain barrier. CD73, an enzyme responsible for adenosine (ADO) production, is overexpressed in cancer cells and emerges as a target for glioblastoma treatment. Indeed, ADO causes a variety of tumor-promoting actions, particularly by inducing tumor immune escape, whereas CD73 inhibition impairs tumor progression. Here, a cationic nanoemulsion to deliver CD73siRNA (NE-siRNA CD73R) via nasal route aiming glioblastoma treatment was developed. NE-siRNA CD73R was uptaken by glioma cells in culture, resulting in a parallel 60-80% decrease in AMPase activity and 30-50% in cell viability. Upon nasal delivery, NE-siRNA CD73R was detected in rat brain and serum. Notably, treatment with CD73siRNA complexes of glioma-bearing Wistar rats reduced tumor growth by 60%. Additionally, NE-siRNA CD73R treatment decreased 95% ADO levels in liquor and tumor CD73 expression, confirming in vivo CD73 silencing. Finally, no toxicity was observed in either primary astrocytes or rats with this cationic nanoemulsion. These results suggest that nasal administration of cationic NE as CD73 siRNA delivery system represents a novel potential treatment for glioblastoma. Graphical Abstract Glioblastoma is the most common and devastating form of primary brain tumor. CD73, a protein involved in cell-cell adhesion and migration processes and also responsible for extracellular adenosine (ADO) production, is overexpressed by glioma cells and emerges as an important target for glioma treatment. Indeed, ADO participates in tumor immune escape, cell proliferation, and angiogenesis, and CD73 inhibition impairs those processes. Here, a cationic nanoemulsion to deliver CD73 siRNA (NE-siRNA CD73R) via nasal route aiming glioblastoma treatment was developed. NE-siRNA CD73R knockdown in vitro and in vivo CD73. Upon nasal delivery of NE-siRNA CD73R, the treatment markedly reduced tumor volume by 60% in a rat preclinical glioblastoma model. The treatment was well tolerated, and did not induce kidney, liver, lung, olfactory, bone marrow, or behavior alterations. These results indicate that the nasal administration of NE as a CD73 siRNA delivery system offered an efficient means of gene knockdown and may represent a potential alternative for glioblastoma treatment.


Subject(s)
5'-Nucleotidase/metabolism , Emulsions/administration & dosage , Gene Transfer Techniques , Glioblastoma/therapy , Nanoparticles/administration & dosage , RNA, Small Interfering/administration & dosage , Administration, Intranasal , Animals , Astrocytes/pathology , Brain Neoplasms/therapy , Cations , Cell Line, Tumor , Cell Proliferation , Cell Survival , GPI-Linked Proteins/metabolism , Glioblastoma/pathology , Humans , Male , Rats, Wistar
9.
Cancer Lett ; 428: 139-146, 2018 08 01.
Article in English | MEDLINE | ID: mdl-29709703

ABSTRACT

Over the last decade, autophagy has emerged as one of the critical cellular systems that control homeostasis. Besides management of normal homeostatic processes, autophagy can also be induced by tissue damage stress or by rapidly progressing tumors. During tumor progression, autophagy mediates a cellular reaction to the changes inside and outside of cells, which leads to tumor adaptation. Even though the regulation of autophagy seems universal and is a well-described process, its dysregulation and role in glioma progression remain an important topic of investigation. In this review, we summarize recent evidence of autophagy regulation in brain tumor tissues and possible interconnection between signaling pathways that govern cellular responses. This perspective may help to assess the qualitative differences and various outcomes in response to autophagy stimulation.


Subject(s)
Autophagy-Related Proteins/genetics , Autophagy/genetics , Brain Neoplasms/genetics , Gene Expression Regulation, Neoplastic , Glioma/genetics , Animals , Apoptosis/drug effects , Apoptosis/genetics , Autophagy/drug effects , Autophagy-Related Proteins/metabolism , Brain Neoplasms/mortality , Brain Neoplasms/pathology , Brain Neoplasms/therapy , Cell Survival/drug effects , Cell Survival/genetics , Chloroquine/pharmacology , Chloroquine/therapeutic use , Clinical Trials as Topic , Disease Models, Animal , Glioma/mortality , Glioma/pathology , Glioma/therapy , Humans , MicroRNAs/metabolism , Mitochondria/drug effects , Mitochondria/metabolism , Oncolytic Virotherapy , RNA, Long Noncoding/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics
10.
Leukemia ; 32(2): 510-519, 2018 02.
Article in English | MEDLINE | ID: mdl-28663580

ABSTRACT

The concept of arming antibodies with bioactive payloads for a site-specific therapy of cancer has gained considerable interest in recent years. However, a successful antibody-based targeting approach critically relies on the availability of a tumor-associated target that is not only preferentially expressed in the tumor tissue but is also easily accessible for antibody therapeutics coming from the bloodstream. Here, we perfused the vasculature of healthy and acute myeloid leukemia (AML)-bearing rats with a reactive ester derivative of biotin and subsequently quantified the biotinylated proteins to identify AML-associated bone marrow (BM) antigens accessible from the bloodstream. In total, >1400 proteins were identified. Overall, 181 proteins were >100-fold overexpressed in AML as compared with normal BM. Eleven of the most differentially expressed proteins were further validated by immunohistochemistry and confocal microscopic analyses, including novel antigens highly expressed in AML cells (for example, adaptor-related protein complex 3 ß2) and in the leukemia-modified extracellular matrix (ECM) (for example, collagen-VI-α-1). The presented atlas of targetable AML-associated BM proteins provides a valuable basis for the development of monoclonal antibodies that could be used as carriers for a site-specific pharmacodelivery of cytotoxic drugs, cytokines or radionuclides to the BM in AML.


Subject(s)
Antibodies, Monoclonal/pharmacology , Bone Marrow/metabolism , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/metabolism , Animals , Bone Marrow/drug effects , Cytokines/metabolism , Humans , Immunohistochemistry/methods , Male , Rats , Rats, Inbred BN
11.
Leukemia ; 31(3): 602-613, 2017 03.
Article in English | MEDLINE | ID: mdl-27538486

ABSTRACT

Apart from its unique histopathological appearance with rare tumor cells embedded in an inflammatory background of bystander cells, classical Hodgkin lymphoma (cHL) is characterized by an unusual activation of a broad range of signaling pathways involved in cellular activation. This includes constitutive high-level activity of nuclear factor-κB (NF-κB), Janus kinase/signal transducer and activator of transcription (JAK/STAT), activator protein-1 (AP-1) and interferon regulatory factor (IRF) transcription factors (TFs) that are physiologically only transiently activated. Here, we demonstrate that inactivation of the putative ubiquitin E3-ligase PDLIM2 contributes to this TF activation. PDLIM2 expression is lost at the mRNA and protein levels in the majority of cHL cell lines and Hodgkin and Reed-Sternberg (HRS) cells of nearly all cHL primary samples. This loss is associated with PDLIM2 genomic alterations, promoter methylation and altered splicing. Reconstitution of PDLIM2 in HRS cell lines inhibits proliferation, blocks NF-κB transcriptional activity and contributes to cHL-specific gene expression. In non-Hodgkin B-cell lines, small interfering RNA-mediated PDLIM2 knockdown results in superactivation of TFs NF-κB and AP-1 following phorbol 12-myristate 13-acetate (PMA) stimulation. Furthermore, expression of PDLIM2 is lost in anaplastic large cell lymphoma (ALCL) that shares key biological aspects with cHL. We conclude that inactivation of PDLIM2 is a recurrent finding in cHL and ALCL, promotes activation of inflammatory signaling pathways and thereby contributes to their pathogenesis.


Subject(s)
Gene Expression Regulation, Neoplastic , Hodgkin Disease/genetics , LIM Domain Proteins/genetics , Lymphoma, Large-Cell, Anaplastic/genetics , Microfilament Proteins/genetics , Base Sequence , Cell Line, Tumor , Cluster Analysis , DNA Methylation , Enzyme Activation , Female , Gene Silencing , Genetic Loci , Hodgkin Disease/metabolism , Humans , LIM Domain Proteins/metabolism , Lymphoma, Large-Cell, Anaplastic/metabolism , Male , Microfilament Proteins/metabolism , Mutation , NF-kappa B/metabolism , Promoter Regions, Genetic , Proteolysis , RNA Splice Sites , Transcription Factors , Ubiquitin-Protein Ligases
12.
Bone Marrow Transplant ; 51(11): 1441-1448, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27295269

ABSTRACT

Allogeneic stem cell transplantation (SCT) remains the best curative option for patients with refractory AML or with high-risk myelodysplastic syndrome (MDS). For decades, age alone had been widely used as the primary criterion to assess eligibility for allogeneic SCT; however, prospective studies to evaluate allogeneic SCT in elderly patients are still limited. A total of 187 patients (median age of 64 years, range 60-77 years) with AML (87%) or MDS (13%) transplanted between 1999 and 2014 were included in this retrospective analysis. Relapse-free survival (RFS) and overall survival (OS) at 3 years were 32% (95% confidence interval (CI): 25-39%) and 35% (95%CI: 27-42%), respectively. Overall survival was 49% (95%CI: 35-64%) in AML patients who were transplanted in first complete remission (CR1), but even patients with active disease did benefit from transplantation, showing an OS at 3 years of 30% (95%CI: 20-40%). Multivariate analysis revealed disease- and patient-specific risk indices as independent prognostic factors for OS and non-relapse mortality (NRM). In conclusion, our monocenter results indicate that patients should not be generally withheld from allogeneic SCT because of age or disease status only. Specific risk models incorporating disease status and disease-specific risk factors at the time of transplantation as well as existing comorbidities are helpful tools to assess transplantation-associated risk factors of elderly patients.


Subject(s)
Hematopoietic Stem Cell Transplantation/methods , Leukemia, Myeloid, Acute/therapy , Myelodysplastic Syndromes/therapy , Risk Assessment/methods , Age Factors , Aged , Female , Hematopoietic Stem Cell Transplantation/mortality , Humans , Leukemia, Myeloid, Acute/mortality , Male , Middle Aged , Myelodysplastic Syndromes/mortality , Retrospective Studies , Survival Analysis , Transplantation, Homologous , Treatment Outcome
13.
Oncogene ; 35(32): 4269-81, 2016 08 11.
Article in English | MEDLINE | ID: mdl-26776161

ABSTRACT

Constitutive activation of the antiapoptotic nuclear factor-κB (NF-κB) signaling pathway is a hallmark of the activated B-cell-like (ABC) subtype of diffuse large B-cell lymphomas (DLBCL). Recurrent oncogenic mutations are found in the scaffold protein CARMA1 (CARD11) that connects B-cell receptor (BCR) signaling to the canonical NF-κB pathway. We asked how far additional downstream processes are activated and contribute to the oncogenic potential of DLBCL-derived CARMA1 mutants. To this end, we expressed oncogenic CARMA1 in the NF-κB negative DLBCL lymphoma cell line BJAB. By a proteomic approach we identified recruitment of ß-catenin and its destruction complex consisting of APC, AXIN1, CK1α and GSK3ß to oncogenic CARMA1. Recruitment of the ß-catenin destruction complex was independent of CARMA1-BCL10-MALT1 complex formation or constitutive NF-κB activation and promoted the stabilization of ß-catenin. The ß-catenin destruction complex was also recruited to CARMA1 in ABC DLBCL cell lines, which coincided with elevated ß-catenin expression. In line, ß-catenin was frequently detected in non-GCB DLBCL biopsies that rely on chronic BCR signaling. Increased ß-catenin amounts alone were not sufficient to induce classical WNT target gene signatures, but could augment TCF/LEF-dependent transcriptional activation in response to WNT signaling. In conjunction with NF-κB, ß-catenin enhanced expression of immunosuppressive interleukin-10 and suppressed antitumoral CCL3, indicating that ß-catenin can induce a favorable tumor microenvironment. Thus, parallel activation of NF-κB and ß-catenin signaling by gain-of-function mutations in CARMA1 augments WNT stimulation and is required for regulating the expression of distinct NF-κB target genes to trigger cell-intrinsic and extrinsic processes that promote DLBCL lymphomagenesis.


Subject(s)
CARD Signaling Adaptor Proteins/genetics , CARD Signaling Adaptor Proteins/metabolism , Carcinogenesis , Guanylate Cyclase/genetics , Guanylate Cyclase/metabolism , Lymphoma, Large B-Cell, Diffuse/metabolism , NF-kappa B/metabolism , Signal Transduction , beta Catenin/metabolism , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Humans , Lymphoma, Large B-Cell, Diffuse/pathology , Mutation , Protein Stability , TCF Transcription Factors/metabolism , rho GTP-Binding Proteins/metabolism
15.
Leukemia ; 29(7): 1578-86, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25708834

ABSTRACT

Antibody drug conjugates (ADCs), in which cytotoxic drugs are linked to antibodies targeting antigens on tumor cells, represent promising novel agents for the treatment of malignant lymphomas. Pinatuzumab vedotin is an anti-CD22 ADC and polatuzumab vedotin an anti-CD79B ADC that are both linked to the microtubule-disrupting agent monomethyl auristatin E (MMAE). In the present study, we analyzed the activity of these agents in different molecular subtypes of diffuse large B-cell lymphoma (DLBCL) both in vitro and in early clinical trials. Both anti-CD22-MMAE and anti-CD79B-MMAE were highly active and induced cell death in the vast majority of activated B-cell-like (ABC) and germinal center B-cell-like (GCB) DLBCL cell lines. Similarly, both agents induced cytotoxicity in models with and without mutations in the signaling molecule CD79B. In line with these observations, relapsed and refractory DLBCL patients of both subtypes responded to these agents. Importantly, a strong correlation between CD22 and CD79B expression in vitro and in vivo was not detectable, indicating that patients should not be excluded from anti-CD22-MMAE or anti-CD79B-MMAE treatment because of low target expression. In summary, these studies suggest that pinatuzumab vedotin and polatuzumab vedotin are active agents for the treatment of patients with different subtypes of DLBCL.


Subject(s)
Antibodies, Monoclonal/pharmacology , CD79 Antigens/immunology , Immunoconjugates/pharmacology , Lymphoma, Large B-Cell, Diffuse/drug therapy , Lymphoma, Large B-Cell, Diffuse/immunology , Sialic Acid Binding Ig-like Lectin 2/immunology , Apoptosis/drug effects , Blotting, Western , CD79 Antigens/genetics , Cell Cycle/drug effects , Cell Proliferation/drug effects , Clinical Trials, Phase I as Topic , Cohort Studies , Flow Cytometry , Follow-Up Studies , Humans , Immunoenzyme Techniques , Lymphoma, Large B-Cell, Diffuse/classification , Lymphoma, Large B-Cell, Diffuse/pathology , Mutation/genetics , Neoplasm Staging , Prognosis , Sialic Acid Binding Ig-like Lectin 2/genetics , Tumor Cells, Cultured
16.
Cell Death Differ ; 20(11): 1444-54, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23933813

ABSTRACT

DNA lesions, constantly produced by endogenous and exogenous sources, activate the DNA damage response (DDR), which involves detection, signaling and repair of the damage. Autophagy, a lysosome-dependent degradation pathway that is activated by stressful situations such as starvation and oxidative stress, regulates cell fate after DNA damage and also has a pivotal role in the maintenance of nuclear and mitochondrial genomic integrity. Here, we review important evidence regarding the role played by autophagy in preventing genomic instability and tumorigenesis, as well as in micronuclei degradation. Several pathways governing autophagy activation after DNA injury and the influence of autophagy upon the processing of genomic lesions are also discussed herein. In this line, the mechanisms by which several proteins participate in both DDR and autophagy, and the importance of this crosstalk in cancer and neurodegeneration will be presented in an integrated fashion. At last, we present a hypothetical model of the role played by autophagy in dictating cell fate after genotoxic stress.


Subject(s)
Autophagy/genetics , Carcinogenesis/genetics , DNA Damage , DNA Repair , Genomic Instability , Animals , Carcinogenesis/metabolism , DNA, Mitochondrial/genetics , DNA, Mitochondrial/metabolism , Humans
17.
Leukemia ; 27(6): 1381-90, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23257783

ABSTRACT

Myeloid cell leukemia-1 (MCL1) is an anti-apoptotic member of the BCL2 family that is deregulated in various solid and hematological malignancies. However, its role in the molecular pathogenesis of diffuse large B-cell lymphoma (DLBCL) is unclear. We analyzed gene expression profiling data from 350 DLBCL patient samples and detected that activated B-cell-like (ABC) DLBCLs express MCL1 at significantly higher levels compared with germinal center B-cell-like DLBCL patient samples (P=2.7 × 10(-10)). Immunohistochemistry confirmed high MCL1 protein expression predominantly in ABC DLBCL in an independent patient cohort (n=249; P=0.001). To elucidate molecular mechanisms leading to aberrant MCL1 expression, we analyzed array comparative genomic hybridization data of 203 DLBCL samples and identified recurrent chromosomal gains/amplifications of the MCL1 locus that occurred in 26% of ABC DLBCLs. In addition, aberrant STAT3 signaling contributed to high MCL1 expression in this subtype. Knockdown of MCL1 as well as treatment with the BH3-mimetic obatoclax induced apoptotic cell death in MCL1-positive DLBCL cell lines. In summary, MCL1 is deregulated in a significant fraction of ABC DLBCLs and contributes to therapy resistance. These data suggest that specific inhibition of MCL1 might be utilized therapeutically in a subset of DLBCLs.


Subject(s)
Gene Expression Profiling , Lymphoma, Large B-Cell, Diffuse/genetics , Proto-Oncogene Proteins c-bcl-2/genetics , Antineoplastic Agents/therapeutic use , Comparative Genomic Hybridization , Humans , Immunohistochemistry , Lymphoma, Large B-Cell, Diffuse/drug therapy , Lymphoma, Large B-Cell, Diffuse/pathology , Myeloid Cell Leukemia Sequence 1 Protein , Real-Time Polymerase Chain Reaction
18.
Ann Hematol ; 91(11): 1765-72, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22926531

ABSTRACT

In most patients, mantle cell lymphoma (MCL) shows an aggressive clinical course with a continuous relapse pattern and a median survival of only 3-5 years. In the current study generation of the European MCL Network, the addition of high-dose Ara-C to R-CHOP (rituximab, cyclophosphamide, doxorubicin, vincristine and prednisone)-like regimen followed by myeloablative consolidation achieved a significant improvement of progression-free survival in younger patients. In elderly patients, rituximab maintenance led to a marked prolongation of remission duration. Emerging strategies include mammalian target of rapamycin (mTOR) inhibitors, proteasome inhibitors, immune modulatory drugs, Bruton's tyrosine kinase inhibitors and others, all based on the dysregulated control of cell cycle machinery and impairment of several apoptotic pathways. Combination strategies are currently being investigated in numerous trials, but their introduction into clinical practice and current treatment algorithms remains a challenge. In the current survey, the application of the molecular targeted compounds were collected and evaluated by a representative national network of 14 haematological institutions. Optimised strategies are recommended for clinical routine. Future studies will apply individualised approaches according to the molecular risk profile of the patient.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Evidence-Based Medicine , Lymphoma, Mantle-Cell/drug therapy , Adult , Age Factors , Aged , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Child , Consensus Development Conferences as Topic , Consolidation Chemotherapy/adverse effects , Consolidation Chemotherapy/methods , European Union , Health Care Surveys , Humans , Induction Chemotherapy/adverse effects , Induction Chemotherapy/methods , Lymphoma, Mantle-Cell/prevention & control , Neoplasm Recurrence, Local/drug therapy , Neoplasm Recurrence, Local/prevention & control , Survival Analysis
19.
Leukemia ; 26(5): 1064-72, 2012 May.
Article in English | MEDLINE | ID: mdl-22116552

ABSTRACT

The median survival of patients with mantle cell lymphoma (MCL) ranges from 3 to 5 years with current chemotherapeutic regimens. A common secondary genomic alteration detected in MCL is chromosome 13q31-q32 gain/amplification, which targets a microRNA (miRNA) cluster, miR-17∼92. On the basis of gene expression profiling, we found that high level expression of C13orf25, the primary transcript from which these miRNAs are processed, was associated with poorer survival in patients with MCL (P=0.021). We demonstrated that the protein phosphatase PHLPP2, an important negative regulator of the PI3K/AKT pathway, was a direct target of miR-17∼92 miRNAs, in addition to PTEN and BIM. These proteins were down-modulated in MCL cells with overexpression of the miR-17∼92 cluster. Overexpression of miR-17∼92 activated the PI3K/AKT pathway and inhibited chemotherapy-induced apoptosis in MCL cell lines. Conversely, inhibition of miR-17∼92 expression suppressed the PI3K/AKT pathway and inhibited tumor growth in a xenograft MCL mouse model. Targeting the miR-17∼92 cluster may therefore provide a novel therapeutic approach for patients with MCL.


Subject(s)
Antineoplastic Agents/pharmacology , Lymphoma, Mantle-Cell/drug therapy , MicroRNAs/genetics , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Animals , Apoptosis Regulatory Proteins/genetics , Bcl-2-Like Protein 11 , Cell Division/genetics , Cell Line , Down-Regulation , Drug Resistance, Neoplasm , Enzyme Activation , Female , Gene Expression Profiling , Humans , Lymphoma, Mantle-Cell/genetics , Lymphoma, Mantle-Cell/pathology , Membrane Proteins/genetics , Mice , Mice, SCID , PTEN Phosphohydrolase/genetics , Proto-Oncogene Proteins/genetics , Transplantation, Heterologous
20.
Oncogene ; 30(28): 3198-206, 2011 Jul 14.
Article in English | MEDLINE | ID: mdl-21478911

ABSTRACT

Multiple myeloma (MM) is a paradigm for a malignant disease that exploits external stimuli of the microenvironment for growth and survival. A thorough understanding of the complex interactions between malignant plasma cells and their surrounding requires a detailed analysis of the transcriptional response of myeloma cells to environmental signals. We determined the changes in gene expression induced by interleukin (IL)-6, tumor necrosis factor-α, IL-21 or co-culture with bone marrow stromal cells in myeloma cell lines. Among a limited set of genes that were consistently activated in response to growth factors, a prominent transcriptional target of cytokine-induced signaling in myeloma cells was the gene encoding the serine/threonine kinase serum/glucocorticoid-regulated kinase 1 (SGK1), which is a down-stream effector of PI3-kinase. We could demonstrate a rapid, strong and sustained induction of SGK1 in the cell lines INA-6, ANBL-6, IH-1, OH-2 and MM.1S as well as in primary myeloma cells. Pharmacologic inhibition of the Janus kinase/signal transducer and activator of transcription (JAK/STAT) pathway abolished STAT3 phosphorylation and SGK1 induction. In addition, small hairpin RNA (shRNA)-mediated knock-down of STAT3 reduced basal and induced SGK1 levels. Furthermore, downregulation of SGK1 by shRNAs resulted in decreased proliferation of myeloma cell lines and reduced cell numbers. On the molecular level, this was reflected by the induction of cell cycle inhibitory genes, for example, CDKNA1/p21, whereas positively acting factors such as CDK6 and RBL2/p130 were downregulated. Our results indicate that SGK1 is a highly cytokine-responsive gene in myeloma cells promoting their malignant growth.


Subject(s)
Cytokines/pharmacology , Immediate-Early Proteins/genetics , Multiple Myeloma/genetics , Multiple Myeloma/pathology , Protein Serine-Threonine Kinases/genetics , Transcription, Genetic/drug effects , Transcription, Genetic/genetics , Bone Marrow Cells/drug effects , Bone Marrow Cells/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Cell Survival/genetics , Cytokines/metabolism , Down-Regulation/genetics , Humans , Immediate-Early Proteins/deficiency , Janus Kinases/metabolism , Multiple Myeloma/metabolism , Protein Serine-Threonine Kinases/deficiency , RNA Interference , STAT3 Transcription Factor/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...