Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
1.
Transpl Infect Dis ; 22(1): e13201, 2020 Feb.
Article in English | MEDLINE | ID: mdl-31643129

ABSTRACT

Adenovirus (HAdV) infections confer a high risk of morbidity and mortality for immunocompromised patients after stem cell transplantation (SCT). Treatment with standard antiviral drugs is of limited efficacy and associated with a high rate of adverse effects. HAdV-specific T cells are crucial for sustained viral elimination and the efficacy of adoptive T-cell therapy with donor-derived HAdV-specific T cells has been reported by several investigators. Here, we report our experience with the transfer of HAdV-specific T cells specific for penton, which was recently identified as an immunodominant target of T cells, and hexon in a 14-year-old boy after T-cell-depleted haploidentical SCT for myelodysplastic syndrome (MDS). He developed severe HAdV-associated enteritis complicated by acute graft-versus-host disease (GvHD). The patient received ten infusions of allogeneic HAdV-specific T cells manufactured from the haploidentical stem cell donor using the CliniMacs Interferon-γ (IFN-γ) cytokine capture and immunomagnetic selection. Initially, T cells were generated against the immunodominant target hexon and in subsequent transfers dual antigen-specific T cells against hexon and penton were applied. T-cell transfers were scheduled individually tailored to current immunosuppressive treatment. Each transfer was followed by reduction of HAdV load in peripheral blood and clinical improvement. Importantly, T-cell responses to both penton and hexon pools emerged in patient blood after repetitive transfers. Unfortunately, the patient experienced bacterial sepsis, and in this context, severe GvHD requiring intensive immunosuppression followed by secondary progression of HAdV infection. The patient succumbed to multiorgan failure 283 days after SCT. This case demonstrates the feasibility of HAdV-specific T-cell transfer even in the presence of immunosuppressive treatment. Targeting of multiple immunodominant viral proteins may prove valuable in patients with complicated HAdV infections.


Subject(s)
Adenovirus Infections, Human/therapy , Adoptive Transfer/methods , Capsid Proteins/immunology , Hematopoietic Stem Cell Transplantation/adverse effects , T-Lymphocytes/immunology , Transplantation, Homologous/adverse effects , Adenovirus Infections, Human/etiology , Adenovirus Infections, Human/immunology , Adolescent , Graft vs Host Disease/complications , Humans , Male , Sepsis/microbiology , Sepsis/mortality , Tissue Donors
2.
Am J Med Genet A ; 173(4): 1017-1037, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28168833

ABSTRACT

Heritable predisposition is an important cause of cancer in children and adolescents. Although a large number of cancer predisposition genes and their associated syndromes and malignancies have already been described, it appears likely that there are more pediatric cancer patients in whom heritable cancer predisposition syndromes have yet to be recognized. In a consensus meeting in the beginning of 2016, we convened experts in Human Genetics and Pediatric Hematology/Oncology to review the available data, to categorize the large amount of information, and to develop recommendations regarding when a cancer predisposition syndrome should be suspected in a young oncology patient. This review summarizes the current knowledge of cancer predisposition syndromes in pediatric oncology and provides essential information on clinical situations in which a childhood cancer predisposition syndrome should be suspected.


Subject(s)
Genetic Predisposition to Disease , Hematologic Neoplasms/diagnosis , Mutation , Neoplasm Proteins/genetics , Neoplasms/diagnosis , Adolescent , Child , Focus Groups/methods , Gene Expression , Genetic Counseling/ethics , Genetic Testing/methods , Genetics, Medical/history , Genetics, Medical/instrumentation , Genetics, Medical/methods , Hematologic Neoplasms/genetics , Hematologic Neoplasms/pathology , History, 21st Century , Humans , Neoplasms/genetics , Neoplasms/pathology , Societies, Medical/history , Syndrome
3.
J Clin Invest ; 133(12)2023 06 15.
Article in English | MEDLINE | ID: mdl-37159273

ABSTRACT

BACKGROUNDAdoptive transfer of EBV-specific T cells can restore specific immunity in immunocompromised patients with EBV-associated complications.METHODSWe provide results of a personalized T cell manufacturing program evaluating donor, patient, T cell product, and outcome data. Patient-tailored clinical-grade EBV-specific cytotoxic T lymphocyte (EBV-CTL) products from stem cell donors (SCDs), related third-party donors (TPDs), or unrelated TPDs from the allogeneic T cell donor registry (alloCELL) at Hannover Medical School were manufactured by immunomagnetic selection using a CliniMACS Plus or Prodigy device and the EBV PepTivators EBNA-1 and Select. Consecutive manufacturing processes were evaluated, and patient outcome and side effects were retrieved by retrospective chart analysis.RESULTSForty clinical-grade EBV-CTL products from SCDs, related TPDs, or unrelated TPDs were generated for 37 patients with refractory EBV infections or EBV-associated malignancies with and without a history of transplantation, within 5 days (median) after donor identification. Thirty-four patients received 1-14 EBV-CTL products (fresh and cryopreserved). EBV-CTL transfer led to a complete response in 20 of 29 patients who were evaluated for clinical response. No infusion-related toxicity was reported. EBV-specific T cells in patients' blood were detectable in 16 of 18 monitored patients (89%) after transfer, and their presence correlated with clinical response.CONCLUSIONPersonalized clinical-grade manufacture of EBV-CTL products via immunomagnetic selection from SCDs, related TPDs, or unrelated TPDs in a timely manner is feasible. Overall, EBV-CTLs were clinically effective and well tolerated. Our data suggest EBV-CTL transfer as a promising therapeutic approach for immunocompromised patients with refractory EBV-associated diseases beyond HSCT, as well as patients with preexisting organ dysfunction.TRIAL REGISTRATIONNot applicable.FUNDINGThis study was funded in part by the German Research Foundation (DFG, 158989968/SFB 900), the Deutsche Kinderkrebsstiftung (DKS 2013.09), Wilhelm-Sander-Stiftung (reference 2015.097.1), Ellen-Schmidt-Program of Hannover Medical School, and German Federal Ministry of Education and Research (reference 01EO0802).


Subject(s)
Epstein-Barr Virus Infections , Immunotherapy, Adoptive , Humans , Herpesvirus 4, Human , Immunotherapy, Adoptive/methods , Retrospective Studies , T-Lymphocytes, Cytotoxic , Unrelated Donors
4.
Eur J Haematol ; 88(5): 406-15, 2012 May.
Article in English | MEDLINE | ID: mdl-22309072

ABSTRACT

Heat shock protein 90 (HSP90) binds and stabilizes numerous proteins and kinases essential for myeloma cell survival and proliferation. We and others have recently demonstrated that inhibition of HSP90 by small molecular mass inhibitors induces cell death in multiple myeloma (MM). However, some of the HSP90 inhibitors involved in early clinical trials have shown limited antitumor activity and unfavorable toxicity profiles. Here, we analyzed the effects of the novel, orally bioavailable HSP90 inhibitor NVP-HSP990 on MM cell proliferation and survival. The inhibitor led to a significant reduction in myeloma cell viability and induced G2 cell cycle arrest, degradation of caspase-8 and caspase-3, and induction of apoptosis. Inhibition of the HSP90 ATPase activity was accompanied by the degradation of MM phospho-Akt and phospho-ERK1/2 and upregulation of Hsp70. Exposure of MM cells to a combination of NVP-HSP990 and either melphalan or histone deacetylase (HDAC) inhibitors caused synergistic inhibition of viability, increased induction of apoptosis, and was able to overcome the primary resistance of the cell line RPMI-8226 to HSP90 inhibition. Combined incubation with melphalan and NVP-HSP990 led to synergistically increased cleavage of caspase-2, caspase-9, and caspase-3. These data demonstrate promising activity for NVP-HSP990 as single agent or combination treatment in MM and provide a rationale for clinical trials.


Subject(s)
Apoptosis/drug effects , Caspases/metabolism , Cell Cycle/drug effects , Melphalan/pharmacology , Multiple Myeloma/pathology , Pyridones/pharmacology , Pyrimidines/pharmacology , Administration, Oral , Biological Availability , Enzyme Activation , Humans , Proteolysis , Pyridones/administration & dosage , Pyridones/pharmacokinetics , Pyrimidines/administration & dosage , Pyrimidines/pharmacokinetics
5.
Eur J Haematol ; 84(4): 337-44, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20028416

ABSTRACT

Heat shock protein 90 (HSP90) is a promising target for tumor therapy. The novel HSP90 inhibitor NVP-AUY922 has preclinical activity in multiple myeloma, however, little is known about effective combination partners to design clinical studies. Multiple myeloma cell lines, OPM-2, RPMI-8226, U-266, LP-1, MM1.S, and primary myeloma cells were exposed to NVP-AUY922 and one of the combination partners histone deacetylase inhibitor NVP-LBH589, suberoylanilide hydroxamic acid (SAHA), melphalan, or doxorubicin, either simultaneously or in sequential patterns. Effects on cell proliferation and apoptosis were determined. Synergistic effects were evaluated using the method of Chou and Talalay. Combined sequential incubation with NVP-AUY922 and SAHA showed that best synergistic effects were achieved with 24 h preincubation with SAHA followed by another 48 h of combination treatment. Combination of NVP-AUY922 with SAHA, NVP-LBH589, melphalan, or doxorubicin resulted in synergistic inhibition of viability, with strong synergy (combination index < 0.3) in the case of melphalan. Importantly, resistance of the RPMI-8226 cell line and relative resistance of some primary myeloma cells against NVP-AUY922 could be overcome by combination treatment. These data show impressive synergistic action of the novel HSP90 inhibitor NVP-AUY922 with melphalan, doxorubicin, NVP-LBH589, and SAHA in multiple myeloma and build the frame work for clinical trials.


Subject(s)
Doxorubicin/pharmacology , HSP90 Heat-Shock Proteins/antagonists & inhibitors , Histone Deacetylase Inhibitors/pharmacology , Isoxazoles/pharmacology , Melphalan/pharmacology , Multiple Myeloma/drug therapy , Resorcinols/pharmacology , Antibiotics, Antineoplastic , Antineoplastic Agents, Alkylating , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Doxorubicin/agonists , Drug Evaluation, Preclinical , Drug Synergism , HSP90 Heat-Shock Proteins/metabolism , Histone Deacetylase Inhibitors/agonists , Histone Deacetylases/metabolism , Humans , Isoxazoles/agonists , Isoxazoles/therapeutic use , Melphalan/agonists , Multiple Myeloma/metabolism , Resorcinols/agonists , Resorcinols/therapeutic use
6.
Exp Cell Res ; 315(14): 2471-8, 2009 Aug 15.
Article in English | MEDLINE | ID: mdl-19410573

ABSTRACT

Multiple myeloma is a malignancy of terminally differentiated plasma cells and is incurable in the majority of the patients. Thus, novel effective treatment regimens are urgently needed. In this study, we examined the effects of co-treatment with proteasome-inhibitor bortezomib and topoisomerase II inhibitor etoposide in multiple myeloma cells lines OPM-2, RPMI-S and NCI-H929. Using the median effect method of Chou and Talalay, we evaluated the combination indices (CI) for simultaneous and sequential treatment schedules. In the sequential treatment schedule, we found strong synergistic effects in all three cell lines, even at low single-agent cytotoxicity levels. When cells were treated simultaneously with both drugs, the synergy was present but less pronounced than in the sequential treatment schedule. The synergistic effects observed in the co-treatment schedules were accompanied by an inhibition of anti-apoptotic effects that were induced by etoposide alone. Namely, bortezomib abrogated both etoposide-induced NF-kappaB activation and etoposide-induced bcl-2 up-regulation. Our data suggest that combining etoposide with bortezomib might be useful for cancer treatment, as bortezomib potentially inhibits counter-regulatory mechanisms of tumor cells, which are induced by topoisomerase II inhibition and which may contribute to acquired chemoresistance.


Subject(s)
Antineoplastic Agents/therapeutic use , Boronic Acids/therapeutic use , Etoposide/therapeutic use , Multiple Myeloma/drug therapy , Protease Inhibitors/therapeutic use , Proteasome Inhibitors , Pyrazines/therapeutic use , Topoisomerase II Inhibitors , Antineoplastic Agents/pharmacology , Boronic Acids/pharmacology , Bortezomib , Cell Cycle/drug effects , Cell Cycle/physiology , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Cell Survival/physiology , DNA Topoisomerases, Type II/metabolism , Drug Synergism , Etoposide/pharmacology , Humans , Multiple Myeloma/enzymology , NF-kappa B/drug effects , NF-kappa B/metabolism , Protease Inhibitors/pharmacology , Proteasome Endopeptidase Complex/metabolism , Proto-Oncogene Proteins c-bcl-2/drug effects , Proto-Oncogene Proteins c-bcl-2/metabolism , Pyrazines/pharmacology
7.
Eur J Haematol ; 82(1): 31-8, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19067746

ABSTRACT

Lytic bone destruction is a hallmark of multiple myeloma (MM) and is because of an uncoupling of bone remodeling. Secretion of Dickkopf (DKK)-1 by myeloma cells is a major factor which causes inhibition of osteoblast precursors. In this study, the effect of different treatment regimens for MM on serum DKK-1 was evaluated and correlated with the response to treatment in 101 myeloma patients receiving bortezomib, thalidomide, lenalidomide, adriamycin and dexamethasone (AD) or high-dose chemotherapy (HDCT) followed by autologous stem cell transplantation (ASCT). At baseline, myeloma patients had increased serum DKK-1 as compared with patients with MGUS (mean 3786 pg/mL vs. 1993 pg/mL). There was no difference between previously untreated MM patients and patients at relapse. A significant decrease of DKK-1 after therapy was seen in the following groups: Bortezomib (4059 pg/mL vs. 1862 pg/mL, P = 0.016), lenalidomide (11837 pg/mL vs. 4374 pg/mL, P = 0.039), AD (1668 pg/mL vs. 1241 pg/mL, P = 0.016), and AD + HDCT + ASCT (2446 pg/mL vs. 1082 pg/mL, P = 0.001). Thalidomide led to a non-significant decrease in DKK-1 (1705 pg/mL vs. 1269 pg/mL, P = 0.081). Within all groups, a significant decrease of DKK-1 was only seen in responders (i.e. patients achieving complete remission or partial remission), but not in non-responders. We show for the first time that serum DKK-1 levels decrease in myeloma patients responding to treatment, irrespective of the regimen chosen. These data suggest that myeloma cells are the main source of circulating DKK-1 protein and provide a framework for clinical trials on anti-DKK-1 treatment in MM.


Subject(s)
Antineoplastic Agents/therapeutic use , Intercellular Signaling Peptides and Proteins/blood , Multiple Myeloma/blood , Multiple Myeloma/drug therapy , Adult , Aged , Aged, 80 and over , Female , Humans , Male , Middle Aged , Salvage Therapy
8.
Eur J Haematol ; 82(6): 440-9, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19220424

ABSTRACT

Proteasome inhibitors and histone deacetylase (HDAC) inhibitors are novel targeted therapies being evaluated in clinical trials for cutaneous T-cell lymphoma (CTCL). However, data in regard to tumor biology are limited with these agents. In the present study we analyzed the effects of the HDAC inhibitor suberoylanilide hydroxamic acid (SAHA) and the proteasome inhibitor bortezomib on human CTCL cells. Four CTCL cell lines (SeAx, Hut-78, MyLa, and HH) were exposed to bortezomib and/ or SAHA at different concentrations. Cell viability was quantified using the MTT assay. In addition, apoptosis and generation of reactive oxygen species were analyzed. Both agents potently inhibited cell viability and induced apoptosis. After 48 h of incubation, IC50 of bortezomib was noted at 8.3 nm, 7.9 nm, 6.3 nm, and 22.5 nm in SeAx, Hut-78, HH, and MyLa cells, respectively. For SAHA, the IC50 values were at 0.6 microm in SeAx cells, 0.75 microm in Hut-78 cells, 0.9 microm in HH cells, and 4.4 microm in MyLa cells. Importantly, combined treatment resulted in synergistic cytotoxic effects, as indicated by Combination indices values <1 using the median effect method of Chou and Talalay. We furthermore found that combined treatment with both agents lead to a decreased proteasome activity, an upregulation of the cell regulators p21 and p27 and increased expression of phosphorylated p38. In addition, we showed that SAHA reduced the vascular endothelial growth factor production of CTCL cells. Our results demonstrate that bortezomib and SAHA synergistically induce apoptosis in CTCL cells and thus provide a rationale for clinical trials of combined proteasome and histone deacetylase inhibition in the treatment of CTCL.


Subject(s)
Boronic Acids/pharmacology , Hydroxamic Acids/pharmacology , Lymphoma, T-Cell, Cutaneous/drug therapy , Pyrazines/pharmacology , Apoptosis , Boronic Acids/pharmacokinetics , Bortezomib , Cell Line, Tumor , Cell Survival , Dose-Response Relationship, Drug , Drug Synergism , Histone Deacetylase Inhibitors , Humans , Hydroxamic Acids/pharmacokinetics , Pharmacokinetics , Pyrazines/pharmacokinetics , Reactive Oxygen Species/analysis , Vorinostat
9.
Fam Cancer ; 16(1): 67-71, 2017 01.
Article in English | MEDLINE | ID: mdl-27573199

ABSTRACT

In a 14-year-old boy with polyposis and rectosigmoid carcinoma, we identified a novel POLE germline mutation, p.(Val411Leu), previously found as recurrent somatic mutation in 'ultramutated' sporadic cancers. This is the youngest reported cancer patient with polymerase proofreading-associated polyposis indicating that POLE mutation p.(Val411Leu) may confer a more severe phenotype than previously reported POLE and POLD1 germline mutations. The patient had multiple café-au-lait macules and a pilomatricoma mimicking the clinical phenotype of constitutional mismatch repair deficiency. We hypothesize that these skin features may be common to different types of constitutional DNA repair defects associated with polyposis and early-onset cancer.


Subject(s)
Brain Neoplasms/genetics , Colorectal Neoplasms/genetics , DNA Polymerase II/genetics , Germ-Line Mutation , Neoplastic Syndromes, Hereditary/genetics , Adolescent , Age of Onset , Cafe-au-Lait Spots/genetics , Hair Diseases/genetics , Humans , Male , Microsatellite Instability , Pilomatrixoma/genetics , Poly-ADP-Ribose Binding Proteins , Skin Neoplasms/genetics
10.
Expert Opin Investig Drugs ; 17(6): 879-95, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18491989

ABSTRACT

BACKGROUND: The ubiquitin-proteasome system has become a promising novel molecular target in cancer due to its critical role in cellular protein degradation, its interaction with cell cycle and apoptosis regulation and its unique mechanism of action. OBJECTIVE: This review focuses both on preclinical results and on data from clinical trials with proteasome inhibitors in cancer. METHODS: Results in hematological malignancies and solid tumors were included, and important data presented in abstract form were considered in this review. RESULTS/CONCLUSION: Bortezomib as first-in-class proteasome inhibitor has proven to be highly effective in some hematological malignancies, overcomes conventional chemoresistance, directly induces cell cycle arrest and apoptosis, and also targets the tumor microenvironment. It has been granted approval by the FDA for relapsed multiple myeloma, and recently for relapsed mantle cell lymphoma. Combination chemotherapy regimens have been developed providing high remission rates and remission quality in frontline treatment or in the relapsed setting in multiple myeloma. The combination of proteasome inhibition with novel targeted therapies is an emerging field in oncology. Moreover, novel proteasome inhibitors, such as NPI-0052 and carfilzomib, have been developed. This review summarizes our knowledge of the ubiquitin-proteasome system and recent data from cancer clinical trials.


Subject(s)
Antineoplastic Agents/therapeutic use , Neoplasm Proteins/antagonists & inhibitors , Neoplasms/drug therapy , Protease Inhibitors/therapeutic use , Proteasome Inhibitors , Antineoplastic Agents/adverse effects , Antineoplastic Agents/pharmacology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Boronic Acids/administration & dosage , Boronic Acids/adverse effects , Boronic Acids/pharmacology , Boronic Acids/therapeutic use , Bortezomib , Cell Line, Tumor/drug effects , Clinical Trials as Topic/statistics & numerical data , Combined Modality Therapy , Drug Design , Drug Screening Assays, Antitumor , Hematologic Neoplasms/drug therapy , Hematologic Neoplasms/metabolism , Hematologic Neoplasms/surgery , Hematopoietic Stem Cell Transplantation , Humans , Neoplasm Proteins/metabolism , Neoplasms/enzymology , Protease Inhibitors/administration & dosage , Protease Inhibitors/adverse effects , Protease Inhibitors/pharmacology , Protein Processing, Post-Translational/drug effects , Protein Processing, Post-Translational/physiology , Pyrazines/administration & dosage , Pyrazines/adverse effects , Pyrazines/pharmacology , Pyrazines/therapeutic use , Salvage Therapy , Ubiquitin/physiology , Ubiquitination
SELECTION OF CITATIONS
SEARCH DETAIL