Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 74
Filter
1.
N Engl J Med ; 386(1): 11-23, 2022 01 06.
Article in English | MEDLINE | ID: mdl-34986284

ABSTRACT

BACKGROUND: A single-group, phase 1-2 study indicated that eltrombopag improved the efficacy of standard immunosuppressive therapy that entailed horse antithymocyte globulin (ATG) plus cyclosporine in patients with severe aplastic anemia. METHODS: In this prospective, investigator-led, open-label, multicenter, randomized, phase 3 trial, we compared the efficacy and safety of horse ATG plus cyclosporine with or without eltrombopag as front-line therapy in previously untreated patients with severe aplastic anemia. The primary end point was a hematologic complete response at 3 months. RESULTS: Patients were assigned to receive immunosuppressive therapy (Group A, 101 patients) or immunosuppressive therapy plus eltrombopag (Group B, 96 patients). The percentage of patients who had a complete response at 3 months was 10% in Group A and 22% in Group B (odds ratio, 3.2; 95% confidence interval [CI], 1.3 to 7.8; P = 0.01). At 6 months, the overall response rate (the percentage of patients who had a complete or partial response) was 41% in Group A and 68% in Group B. The median times to the first response were 8.8 months (Group A) and 3.0 months (Group B). The incidence of severe adverse events was similar in the two groups. With a median follow-up of 24 months, a karyotypic abnormality that was classified as myelodysplastic syndrome developed in 1 patient (Group A) and 2 patients (Group B); event-free survival was 34% and 46%, respectively. Somatic mutations were detected in 29% (Group A) and 31% (Group Β) of the patients at baseline; these percentages increased to 66% and 55%, respectively, at 6 months, without affecting the hematologic response and 2-year outcome. CONCLUSIONS: The addition of eltrombopag to standard immunosuppressive therapy improved the rate, rapidity, and strength of hematologic response among previously untreated patients with severe aplastic anemia, without additional toxic effects. (Funded by Novartis and others; RACE ClinicalTrials.gov number, NCT02099747; EudraCT number, 2014-000363-40.).


Subject(s)
Anemia, Aplastic/therapy , Antilymphocyte Serum/therapeutic use , Benzoates/therapeutic use , Cyclosporine/therapeutic use , Hydrazines/therapeutic use , Immunosuppression Therapy , Immunosuppressive Agents/therapeutic use , Pyrazoles/therapeutic use , Adolescent , Adult , Aged , Aged, 80 and over , Anemia, Aplastic/drug therapy , Anemia, Aplastic/genetics , Antilymphocyte Serum/adverse effects , Benzoates/adverse effects , Cyclosporine/adverse effects , Drug Therapy, Combination , Female , Humans , Hydrazines/adverse effects , Immunosuppressive Agents/adverse effects , Male , Middle Aged , Progression-Free Survival , Prospective Studies , Pyrazoles/adverse effects , Receptors, Thrombopoietin/agonists , Remission Induction , Young Adult
2.
Br J Haematol ; 191(5): 764-774, 2020 12.
Article in English | MEDLINE | ID: mdl-32583458

ABSTRACT

Primary objective of this non-interventional, post-authorisation safety study was to provide real-world safety data [incidence of adverse drug reactions (ADRs)/serious adverse events (SAEs)] on adult patients with myelofibrosis exposed/or not exposed to ruxolitinib. Key secondary objectives included the incidence/outcome of events of special interest (bleeding events, serious/opportunistic infections, second primary malignancies, and deaths). Overall, 462 patients were included [prevalent users = 260, new users = 32, non-exposed = 170 (inclusive of ruxolitinib-switch, n = 57)]. The exposure-adjusted incidence rates (per 100 patient-years) of ADRs (19·3 vs. 19·6) and SAEs (25·2 vs. 25·0) were comparable amongst new-users versus prevalent-users cohorts, respectively; most frequent ADRs across all cohorts included thrombocytopenia, anaemia, epistaxis, urinary tract infection, and herpes zoster. Anaemia, pneumonia, general physical health deterioration, sepsis, and death were the most frequent SAEs across all cohorts. Incidence rates of bleeding events (21·6) and serious/opportunistic infections (34·5) were higher in ruxolitinib-switch cohort versus other cohorts. The incidence rate of second primary malignancies was higher in the prevalent-users cohort (10·1) versus other cohorts. The observed safety profile of ruxolitinib in the present study along with the safety findings from the COMFORT/JUMP/EXPAND studies support the use of ruxolitinib for long-term treatment of patients with myelofibrosis.


Subject(s)
Primary Myelofibrosis/drug therapy , Pyrazoles/administration & dosage , Pyrazoles/adverse effects , Adult , Aged , Aged, 80 and over , Female , Humans , Male , Middle Aged , Nitriles , Prospective Studies , Pyrimidines
3.
Haematologica ; 104(1): 147-154, 2019 01.
Article in English | MEDLINE | ID: mdl-30115656

ABSTRACT

Lenalidomide has been proven to be effective but with a distinct and difficult to manage toxicity profile in the context of chronic lymphocytic leukemia, potentially hampering combination treatment with this drug. We conducted a phase 1-2 study to evaluate the efficacy and safety of six cycles of chlorambucil (7 mg/m2 daily), rituximab (375 mg/m2 cycle 1 and 500 mg/m2 cycles 2-6) and individually-dosed lenalidomide (escalated from 2.5 mg to 10 mg) (induction-I) in first-line treatment of patients with chronic lymphocytic leukemia unfit for treatment with fludarabine, cyclophosphamide and rituximab. This was followed by 6 months of 10 mg lenalidomide monotherapy (induction-II). Of 53 evaluable patients in phase 2 of the study, 47 (89%) completed induction-I and 36 (68%) completed induction-II. In an intention-to-treat analysis, the overall response rate was 83%. The median progression-free survival was 49 months, after a median follow-up time of 27 months. The 2- and 3-year progression-free survival rates were 58% and 54%, respectively. The corresponding rates for overall survival were 98% and 95%. No tumor lysis syndrome was observed, while tumor flair reaction occurred in five patients (9%, 1 grade 3). The most common hematologic toxicity was grade 3-4 neutropenia, which occurred in 73% of the patients. In conclusion, addition of lenalidomide to a chemotherapy backbone followed by a fixed duration of lenalidomide monotherapy resulted in high remission rates and progression-free survival rates, which seem comparable to those observed with novel drug combinations including novel CD20 monoclonal antibodies or kinase inhibitors. Although lenalidomide-specific toxicity remains a concern, an individualized dose-escalation schedule is feasible and results in an acceptable toxicity profile. EuraCT number: 2010-022294-34.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy , Leukemia, Lymphocytic, Chronic, B-Cell/mortality , Adolescent , Adult , Chlorambucil/administration & dosage , Disease-Free Survival , Feasibility Studies , Female , Humans , Lenalidomide/administration & dosage , Leukemia, Lymphocytic, Chronic, B-Cell/metabolism , Leukemia, Lymphocytic, Chronic, B-Cell/pathology , Male , Middle Aged , Rituximab/administration & dosage , Survival Rate , Vidarabine/administration & dosage , Vidarabine/analogs & derivatives
4.
Br J Haematol ; 183(1): 60-67, 2018 10.
Article in English | MEDLINE | ID: mdl-30080247

ABSTRACT

This single-centre retrospective observational study analysed the efficacy of retreatment with immunomodulatory agents (IMiDs) and proteasome inhibitors (PIs) after treatment with daratumumab monotherapy in patients with relapsed and/or refractory multiple myeloma (RRMM). In total 55 patients were treated with daratumumab monotherapy between 2010 and 2017. From this group 29 (53%) IMiD-refractory patients were retreated with an IMiD after daratumumab and 6 (11%) PI-refractory patients were retreated with a PI-based regimen. For the IMiD-refractory patients the overall response rate (ORR) was 52% (15/29 patients, partial response or better) upon IMiD retreatment, whereas the ORR to PI retreatment was 67% (4/6 patients) in the PI-refractory group. The immunomodulatory effects of daratumumab may play a role in these high response rates in previously refractory patients. Due to the >6 month-long persistence of daratumumab in the plasma the subsequent therapies can effectively be considered as combination therapy. Furthermore, the excellent tolerability of daratumumab treatment may enable patients to recover from prior lines of treatment and receive full dosing of subsequent therapies. In conclusion, a high proportion of RRMM patients benefitted from retreatment with IMiDs and PIs after daratumumab treatment. These retreatment options should therefore be explored in RRMM patients progressing on daratumumab monotherapy.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Immunologic Factors/therapeutic use , Multiple Myeloma/drug therapy , Proteasome Inhibitors/therapeutic use , Salvage Therapy/methods , Cohort Studies , Female , Humans , Male , Multiple Myeloma/mortality , Remission Induction , Retreatment/methods , Retrospective Studies , Survival Analysis , Treatment Outcome
5.
Blood ; 128(4): 497-503, 2016 07 28.
Article in English | MEDLINE | ID: mdl-27226434

ABSTRACT

Patients with relapsed and/or refractory multiple myeloma (RRMM) have poor prognosis. The STRATUS study assessed safety and efficacy of pomalidomide plus low-dose dexamethasone in the largest cohort to date of patients with RRMM. Patients who failed treatment with bortezomib and lenalidomide and had adequate prior alkylator therapy were eligible. Pomalidomide 4 mg was given on days 1-21 of 28-day cycles with low-dose dexamethasone 40 mg (20 mg for patients aged >75 years) on days 1, 8, 15, and 22 until progressive disease or unacceptable toxicity. Safety was the primary end point; secondary end points included overall response rate (ORR), duration of response (DOR), progression-free survival (PFS), and overall survival (OS). Among 682 patients enrolled, median age was 66 years, and median time since diagnosis was 5.3 years. Median number of prior regimens was 5. Most patients were refractory to both lenalidomide and bortezomib (80.2%). Median follow-up was 16.8 months; median duration of treatment was 4.9 months. Most frequent grade 3/4 treatment-emergent adverse events were hematologic (neutropenia [49.7%], anemia [33.0%], and thrombocytopenia [24.1%]). Most common grade 3/4 nonhematologic toxicities were pneumonia (10.9%) and fatigue (5.9%). Grade 3/4 venous thromboembolism and peripheral neuropathy were rare (1.6% each). The ORR was 32.6%, and the median DOR was 7.4 months. Median PFS and OS were 4.6 months and 11.9 months, respectively. We present the largest trial to date evaluating pomalidomide plus low-dose dexamethasone in patients with RRMM, further confirming that this regimen offers clinically meaningful benefit and is generally well tolerated. www.Clinicaltrials.gov identifier NCT01712789.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Multiple Myeloma/drug therapy , Adult , Aged , Aged, 80 and over , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Dexamethasone/administration & dosage , Dexamethasone/adverse effects , Female , Follow-Up Studies , Humans , Male , Middle Aged , Thalidomide/administration & dosage , Thalidomide/adverse effects , Thalidomide/analogs & derivatives
6.
Blood ; 128(19): 2297-2306, 2016 11 10.
Article in English | MEDLINE | ID: mdl-27647864

ABSTRACT

The prognosis of multiple myeloma (MM) patients who become refractory to lenalidomide and bortezomib is very poor, indicating the need for new therapeutic strategies for these patients. Next to the development of new drugs, the strategy of combining agents with synergistic activity may also result in clinical benefit for patients with advanced myeloma. We have previously shown in a retrospective analysis that lenalidomide combined with continuous low-dose cyclophosphamide and prednisone (REP) had remarkable activity in heavily pretreated, lenalidomide-refractory MM patients. To evaluate this combination prospectively, we initiated a phase 1/2 study to determine the optimal dose and to assess its efficacy and safety in lenalidomide-refractory MM patients. The maximum tolerated dose (MTD) was defined as 25 mg lenalidomide (days 1-21/28 days), combined with continuous cyclophosphamide (50 mg/d) and prednisone (20 mg/d). At the MTD (n = 67 patients), the overall response rate was 67%, and at least minimal response was achieved in 83% of the patients. Median progression-free survival and overall survival were 12.1 and 29.0 months, respectively. Similar results were achieved in the subset of patients with lenalidomide- and bortezomib-refractory disease as well as in patients with high-risk cytogenetic abnormalities, defined as t(4;14), t(14;16), del(17p), and/or ampl(1q) as assessed by fluorescence in situ hybridization. Neutropenia (22%) and thrombocytopenia (22%) were the most common grade 3-4 hematologic adverse events. Infections (21%) were the most common grade 3-5 nonhematologic adverse events. In conclusion, the addition of continuous low-dose oral cyclophosphamide to lenalidomide and prednisone offers a new therapeutic perspective for multidrug refractory MM patients. This trial was registered at www.clinicaltrials.gov as #NCT01352338.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Cyclophosphamide/therapeutic use , Drug Resistance, Neoplasm , Multiple Myeloma/drug therapy , Prednisone/therapeutic use , Thalidomide/analogs & derivatives , Adult , Aged , Aged, 80 and over , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Cyclophosphamide/adverse effects , Disease-Free Survival , Dose-Response Relationship, Drug , Female , Humans , Lenalidomide , Male , Maximum Tolerated Dose , Middle Aged , Prednisone/adverse effects , Prognosis , Thalidomide/adverse effects , Thalidomide/therapeutic use , Treatment Outcome
7.
Eur J Haematol ; 99(3): 199-206, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28504846

ABSTRACT

OBJECTIVES: Heavily pretreated patients with relapsed and refractory multiple myeloma are susceptible to treatment-related adverse events (AEs). Managing AEs are important to ensure patients continue therapy long enough to receive the best clinical benefit. Data from the MM-002, MM-003, and MM-010 trials were pooled to further characterize the safety profile of pomalidomide plus low-dose dexamethasone and AE management. METHODS: This analysis included 1088 patients who received ≥ 2 prior therapies, including lenalidomide and bortezomib, and progressed ≤ 60 days of last therapy. Patients received 28-day cycles of pomalidomide 4 mg/day on days 1-21 and low-dose dexamethasone 40 mg (20 mg if aged > 75 years) weekly until disease progression or unacceptable toxicity. Thromboprophylaxis was required. RESULTS: The most common grade 3/4 AEs were neutropenia (56.2%), anemia (32.3%), and thrombocytopenia (25.8%), which occurred within the first few cycles of treatment. Grade 3/4 infections occurred in 33.7% patients, of whom 13.9% had pneumonia, and 40.3% had neutropenia. Pomalidomide dose reductions or interruptions were reported in 24.2% and 66.0% of patients, respectively. AEs were managed by dose modifications and/or supportive care. CONCLUSIONS: Pomalidomide plus low-dose dexamethasone showed an acceptable safety profile, and AEs were well managed according to study protocols and established guidelines.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/adverse effects , Drug-Related Side Effects and Adverse Reactions/diagnosis , Drug-Related Side Effects and Adverse Reactions/therapy , Multiple Myeloma/drug therapy , Multiple Myeloma/pathology , Adult , Aged , Aged, 80 and over , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Clinical Trials as Topic , Combined Modality Therapy , Dexamethasone/administration & dosage , Disease Management , Drug Resistance, Neoplasm , Humans , Middle Aged , Multicenter Studies as Topic , Neoplasm Recurrence, Local , Thalidomide/administration & dosage , Thalidomide/analogs & derivatives , Time Factors
8.
Biol Blood Marrow Transplant ; 22(10): 1883-1887, 2016 10.
Article in English | MEDLINE | ID: mdl-27470287

ABSTRACT

Cytomegalovirus (CMV) serostatus of donor and recipient are frequently used in algorithms of donor selection, whereas the impact of CMV reactivation on transplantation-related mortality, leukemia control, and overall survival (OS) remains controversial. Therefore, we retrospectively studied the impact of latent or active CMV infections on the outcome and occurrence of graft-versus-host disease (GVHD) after reduced-intensity conditioning (RIC) allogeneic hematopoietic stem cell transplantation (SCT) in 294 patients during the period from 2004 to 2010. CMV viral load was routinely monitored in plasma using a quantitative PCR. Preemptive antiviral therapy was initiated when the viral load in plasma exceeded a predefined threshold. In a proportional hazards model, a seropositive recipient was significantly associated with increased occurrence of acute GVHD. However the CMV serostatus of both recipient and donor and the presence of active CMV infection was not associated with the occurrence of relapses, chronic GVHD, or OS. We conclude that in the presence of viral load monitoring and preemptive treatment, latent or active CMV infection does not substantially affect the OS after T cell-replete RIC allogeneic SCT.


Subject(s)
Cytomegalovirus Infections/prevention & control , Cytomegalovirus/physiology , Hematopoietic Stem Cell Transplantation/adverse effects , Virus Activation , Adolescent , Adult , Aged , Antiviral Agents/therapeutic use , Cytomegalovirus Infections/drug therapy , Graft vs Host Disease/etiology , Hematopoietic Stem Cell Transplantation/mortality , Humans , Middle Aged , Premedication/methods , Retrospective Studies , Survival Analysis , Transplantation Conditioning/methods , Transplantation, Homologous/adverse effects , Transplantation, Homologous/mortality , Young Adult
9.
Blood ; 123(25): 3873-86; quiz 4005, 2014 Jun 19.
Article in English | MEDLINE | ID: mdl-24665134

ABSTRACT

During recent years, our understanding of the pathogenesis of inherited microcytic anemias has gained from the identification of several genes and proteins involved in systemic and cellular iron metabolism and heme syntheses. Numerous case reports illustrate that the implementation of these novel molecular discoveries in clinical practice has increased our understanding of the presentation, diagnosis, and management of these diseases. Integration of these insights into daily clinical practice will reduce delays in establishing a proper diagnosis, invasive and/or costly diagnostic tests, and unnecessary or even detrimental treatments. To assist the clinician, we developed evidence-based multidisciplinary guidelines on the management of rare microcytic anemias due to genetic disorders of iron metabolism and heme synthesis. These genetic disorders may present at all ages, and therefore these guidelines are relevant for pediatricians as well as clinicians who treat adults. This article summarizes these clinical practice guidelines and includes background on pathogenesis, conclusions, and recommendations and a diagnostic flowchart to facilitate using these guidelines in the clinical setting.


Subject(s)
Anemia, Hypochromic/diagnosis , Anemia, Hypochromic/therapy , Heme/biosynthesis , Iron/metabolism , Practice Guidelines as Topic , Anemia, Hypochromic/genetics , Anemia, Iron-Deficiency/diagnosis , Anemia, Iron-Deficiency/genetics , Anemia, Iron-Deficiency/therapy , Anemia, Sideroblastic/diagnosis , Anemia, Sideroblastic/genetics , Anemia, Sideroblastic/therapy , Evidence-Based Medicine , Genetic Predisposition to Disease/genetics , Humans , Mutation
10.
Eur J Haematol ; 97(5): 479-488, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27028304

ABSTRACT

Allogeneic stem cell transplantation (allo-SCT) has the potential to induce long-term remission in multiple myeloma (MM), but the role of allo-SCT in MM is controversial due to the high rate of treatment-related mortality (TRM). However, although proteasome inhibitors and immunomodulatory drugs have improved the outcome of patients with MM, high-risk patients still have a very poor prognosis. This indicates the need for new treatment strategies and identification of patients who might benefit from allo-SCT. We therefore analyzed the outcome of one hundred and forty-seven patients with MM who received an allo-SCT at our institution (58 in first line, 89 in relapsed/refractory setting) after a median follow-up of 88.8 months. For the first-line setting, median progression-free survival (PFS) and overall survival (OS) were remarkably good, with a CR rate of 48.3%, median PFS of 30.2 months, and 10-yr OS of 51%. We found no difference in outcome for patients with high-risk metaphase cytogenetics or FISH del(13q14), but efficacy in current standard high-risk patients could not be determined. The outcome in the relapsed/refractory setting was poor, especially in the subgroup of patients relapsing within 18 months after auto-SCT. Therefore, if applied at all in these patients, improvement of allo-SCT is needed, focusing on reduction of TRM and more effective immunotherapy.


Subject(s)
Hematopoietic Stem Cell Transplantation , Multiple Myeloma/diagnosis , Multiple Myeloma/therapy , Adult , Aged , Combined Modality Therapy , Disease Progression , Disease-Free Survival , Female , Follow-Up Studies , Graft vs Host Disease/etiology , Hematopoietic Stem Cell Transplantation/adverse effects , Humans , Male , Middle Aged , Multiple Myeloma/mortality , Prognosis , Recurrence , Retreatment , Transplantation, Homologous , Treatment Outcome
12.
Haematologica ; 100(5): 677-82, 2015 May.
Article in English | MEDLINE | ID: mdl-25661441

ABSTRACT

In a prospective multicenter phase II study, we evaluated the effect of three courses of vincristine, doxorubicin and dexamethasone followed by high-dose melphalan and autologous stem cell transplantation on an intention-to-treat basis. Sixty-nine newly diagnosed patients with amyloid light chain amyloidosis were included between November 2000 and January 2006: 37 men and 32 women with a median age of 56 years, including 46% of patients with cardiac and 22% of patients with involvement of 3 or 4 organs. Initial results presented in 2008 showed a 4-year overall survival rate of 62% among all the patients, while the 4-year survival rate after transplantation was 78%. Here we report the long-term follow-up data after a median follow up of 115 months of the patients still alive. Median survival of all patients was 96 months from registration and for the transplanted patients ten years from the date of transplantation. Twelve (12%) patients died during induction therapy with vincristine, doxorubicin and dexamethasone, including 8 patients (12%) due to treatment-related mortality. Two patients died within one month following high-dose melphalan. We conclude that vincristine, doxorubicin and dexamethasone should not be applied as induction therapy for intensification in amyloid light chain amyloidosis. However, a 2-step approach consisting of a non-intensive less toxic induction therapy followed by high-dose melphalan and autologous stem cell transplantation may result in extended survival in newly diagnosed patients with amyloid light chain amyloidosis (clinicaltrials.gov identifier: 01207094).


Subject(s)
Amyloidosis/therapy , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Hematopoietic Stem Cell Transplantation , Melphalan/administration & dosage , Adult , Aged , Amyloidosis/diagnosis , Amyloidosis/etiology , Amyloidosis/mortality , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Combined Modality Therapy , Dexamethasone/administration & dosage , Doxorubicin/administration & dosage , Female , Follow-Up Studies , Hematopoietic Stem Cell Transplantation/adverse effects , Humans , Kaplan-Meier Estimate , Male , Middle Aged , Remission Induction , Transplantation, Autologous , Treatment Outcome , Vincristine/administration & dosage
13.
Blood ; 119(26): 6219-25; quiz 6399, 2012 Jun 28.
Article in English | MEDLINE | ID: mdl-22442350

ABSTRACT

To prospectively evaluate allogeneic stem cell transplantation (allo-SCT) for myeloma as part of first-line therapy, a donor versus no-donor analysis was performed of patients treated in the HOVON-50 study, a study that was originally designed to examine thalidomide combined with intensive therapy. Two hundred sixty patients having received an autologous-SCT fulfilled the criteria to be included, 138 patients without an HLA-identical sibling donor and 122 patients with a donor. After a median follow-up of 77 months, complete remission, progression-free survival (PFS), and overall survival were not significantly different between the 2 groups. PFS at 6 years was 28% for patients with a donor versus 22% for patients without a donor (P = .19) and overall survival at 6 years from high-dose melphalan was 55%, irrespective of having a donor (P = .68). Cumulative incidence of nonrelapse mortality at 6 years after autologous-SCT was 16% in the donor group versus 3% in the no-donor group (P < .001). However, PFS was significantly prolonged in the 99 patients who actually proceeded to allo-SCT compared with the 115 patients who continued maintenance or received a second high-dose melphalan, but the difference did not translate into a prolonged survival benefit. These results do not support a general application of allo-SCT in all myeloma patients as part of first-line therapy.


Subject(s)
Hematopoietic Stem Cell Transplantation/methods , Multiple Myeloma/therapy , Tissue Donors , Adult , Aged , Algorithms , Female , Hematopoietic Stem Cell Transplantation/adverse effects , Hematopoietic Stem Cell Transplantation/statistics & numerical data , Histocompatibility Testing , Humans , Male , Middle Aged , Multiple Myeloma/diagnosis , Multiple Myeloma/immunology , Neoadjuvant Therapy , Netherlands , Siblings , Transplantation Conditioning/methods , Transplantation, Homologous , Treatment Outcome
14.
Blood ; 120(3): e9-e16, 2012 Jul 19.
Article in English | MEDLINE | ID: mdl-22653974

ABSTRACT

Interactions within the hematopoietic niche in the BM microenvironment are essential for maintenance of the stem cell pool. In addition, this niche is thought to serve as a sanctuary site for malignant progenitors during chemotherapy. Therapy resistance induced by interactions with the BM microenvironment is a major drawback in the treatment of hematologic malignancies and bone-metastasizing solid tumors. To date, studying these interactions was hampered by the lack of adequate in vivo models that simulate the human situation. In the present study, we describe a unique human-mouse hybrid model that allows engraftment and outgrowth of normal and malignant hematopoietic progenitors by implementing a technology for generating a human bone environment. Using luciferase gene marking of patient-derived multiple myeloma cells and bioluminescent imaging, we were able to follow pMM cells outgrowth and to visualize the effect of treatment. Therapeutic interventions in this model resulted in equivalent drug responses as observed in the corresponding patients. This novel human-mouse hybrid model creates unprecedented opportunities to investigate species-specific microenvironmental influences on normal and malignant hematopoietic development, and to develop and personalize cancer treatment strategies.


Subject(s)
Hematopoietic Stem Cells/cytology , Multiple Myeloma/immunology , Multiple Myeloma/pathology , Stem Cell Niche/immunology , Transplantation Chimera/immunology , Tumor Microenvironment/immunology , Animals , DNA-Binding Proteins/genetics , Disease Models, Animal , Ear Ossicles/cytology , Hematopoietic Stem Cell Transplantation/methods , Humans , Immunologic Deficiency Syndromes/genetics , Immunologic Deficiency Syndromes/immunology , Mice , Mice, Mutant Strains , Neoplasm Transplantation , Osteolysis/immunology , Tissue Scaffolds , Transplantation, Heterologous
15.
Am J Hematol ; 89(3): 315-9, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24166784

ABSTRACT

X-linked sideroblastic anemia (XLSA) is the most common form of congenital sideroblastic anemia. In affected males, it is uniformly associated with partial loss-of-function missense mutations in the erythroid-specific heme biosynthesis protein 5-aminolevulinate synthase 2 (ALAS2). Here, we report five families with XLSA owing to mutations in a GATA transcription factor binding site located in a transcriptional enhancer element in intron 1 of the ALAS2 gene. As such, this study defines a new class of mutations that should be evaluated in patients undergoing genetic testing for a suspected diagnosis of XLSA.


Subject(s)
5-Aminolevulinate Synthetase/genetics , Anemia, Sideroblastic/genetics , Enhancer Elements, Genetic/genetics , GATA Transcription Factors/metabolism , Genetic Diseases, X-Linked/genetics , Introns/genetics , Mutation , Adult , Aged , Anemia, Sideroblastic/blood , Binding Sites , Europe/ethnology , Female , Genetic Diseases, X-Linked/blood , Genotype , Humans , Male , Middle Aged , Pedigree , Young Adult
16.
Virchows Arch ; 484(5): 837-845, 2024 May.
Article in English | MEDLINE | ID: mdl-38602559

ABSTRACT

The classical BCR::ABL1-negative myeloproliferative neoplasms (MPN) form a group of bone marrow (BM) diseases with the potential to progress to acute myeloid leukemia or develop marrow fibrosis and subsequent BM failure. The mechanism by which BM fibrosis develops and the factors that drive stromal activation and fibrosis are not well understood. Cellular Communication Network 2 (CCN2), also known as CTGF (Connective Tissue Growth Factor), is a profibrotic matricellular protein functioning as an important driver and biomarker of fibrosis in a wide range of diseases outside the marrow. CCN2 can promote fibrosis directly or by acting as a factor downstream of TGF-ß, the latter already known to contribute to myelofibrosis in MPN.To study the possible involvement of CCN2 in BM fibrosis in MPN, we assessed CCN2 protein expression by immunohistochemistry in 75 BM biopsies (55 × MPN and 20 × normal controls). We found variable expression of CCN2 in megakaryocytes with significant overexpression in a subgroup of 7 (13%) MPN cases; 4 of them (3 × essential thrombocytemia and 1 × prefibrotic primary myelofibrosis) showed no fibrosis (MF-0), 2 (1 × post-polycythemic myelofibrosis and 1 × primary myelofibrosis) showed moderate fibrosis (MF-2), and 1 (primary myelofibrosis) severe fibrosis (MF-3). Remarkably, CCN2 expression did not correlate with fibrosis or other disease parameters such as platelet count or thrombovascular events, neither in this subgroup nor in the whole study group. This suggests that in BM of MPN patients other, CCN2-independent pathways (such as noncanonical TGF-ß signaling) may be more important for the development of fibrosis.


Subject(s)
Connective Tissue Growth Factor , Myeloproliferative Disorders , Primary Myelofibrosis , Signal Transduction , Transforming Growth Factor beta , Humans , Connective Tissue Growth Factor/metabolism , Transforming Growth Factor beta/metabolism , Primary Myelofibrosis/pathology , Primary Myelofibrosis/metabolism , Middle Aged , Male , Female , Aged , Myeloproliferative Disorders/pathology , Myeloproliferative Disorders/metabolism , Adult , Bone Marrow/pathology , Bone Marrow/metabolism , Aged, 80 and over , Immunohistochemistry , Fibrosis/pathology
17.
EMBO Mol Med ; 16(10): 2560-2582, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39198715

ABSTRACT

Dyskeratosis congenita (DC) is a rare inherited bone marrow failure syndrome, caused by genetic mutations that principally affect telomere biology. Approximately 35% of cases remain uncharacterised at the genetic level. To explore the genetic landscape, we conducted genetic studies on a large collection of clinically diagnosed cases of DC as well as cases exhibiting features resembling DC, referred to as 'DC-like' (DCL). This led us to identify several novel pathogenic variants within known genetic loci and in the novel X-linked gene, POLA1. In addition, we have also identified several novel variants in POT1 and ZCCHC8 in multiple cases from different families expanding the allelic series of DC and DCL phenotypes. Functional characterisation of novel POLA1 and POT1 variants, revealed pathogenic effects on protein-protein interactions with primase, CTC1-STN1-TEN1 (CST) and shelterin subunit complexes, that are critical for telomere maintenance. ZCCHC8 variants demonstrated ZCCHC8 deficiency and signs of pervasive transcription, triggering inflammation in patients' blood. In conclusion, our studies expand the current genetic architecture and broaden our understanding of disease mechanisms underlying DC and DCL disorders.


Subject(s)
Dyskeratosis Congenita , Telomere-Binding Proteins , Telomere , Dyskeratosis Congenita/genetics , Humans , Telomere-Binding Proteins/genetics , Telomere-Binding Proteins/metabolism , Telomere/genetics , Telomere/metabolism , Male , Shelterin Complex , Female , Mutation , Child
19.
Br J Haematol ; 162(1): 50-61, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23617231

ABSTRACT

This systematic review of studies compared magnetic resonance imaging (MRI), (18) F-fluorodeoxyglucose positron emission tomography (FDG-PET), FDG-PET with computerized tomography (PET-CT) and CT with whole body X-Ray (WBXR) or (whole body) CT in order to provide evidence-based diagnostic guidelines in multiple myeloma bone disease. A comprehensive search of 3 bibliographic databases was performed; methodological quality was assessed using Quality Assessment of Diagnostic Accuracy Studies (QUADAS) criteria (score 1-14). Data from 32 directly comparative studies were extracted. The mean QUADAS score was 7·1 (3-11), with quality hampered mainly by a poor description of selection and execution criteria. All index tests had a higher detection rate when compared to WBXR, with up to 80% more lesions detected by the newer imaging techniques; MRI (1·12-1·82) CT (1·04-1·33), PET (1·00-1·58) and PET-CT (1·27-1·45). However, the modern imaging techniques detected fewer lesions in the skull and ribs. In a direct comparison CT and MRI performed equally with respect to detection rate and sensitivity. This systematic review supports the International Myeloma Working Group guidelines, which recommend that WBCT can replace WBXR. In our opinion, the equal performance of MRI also indicates that it is a valuable alternative. As lesions of the skull and ribs are underdiagnosed by modern imaging techniques we advise additional X-rays of these regions. The consequences of this approach are discussed.


Subject(s)
Bone Diseases/diagnosis , Bone Diseases/etiology , Diagnostic Imaging , Multiple Myeloma/complications , Diagnostic Imaging/methods , Humans , Magnetic Resonance Imaging , Multiple Myeloma/diagnosis , Positron-Emission Tomography , Prognosis , Reproducibility of Results , Sensitivity and Specificity , Tomography, X-Ray Computed
20.
Cancer Immunol Immunother ; 62(8): 1381-92, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23728352

ABSTRACT

The introduction of autologous stem cell transplantation (SCT) and novel drugs has improved overall survival in multiple myeloma (MM) patients. However, minimal residual disease (MRD) remains and most patients eventually relapse. Myeloma plasma cells express tumor-associated antigens (TAA), which are interesting targets for immunotherapy. In this phase 1 study, we investigated the safety and immunological effects of TAA-mRNA-loaded dendritic cell (DC) vaccination for treatment for MRD in MM after SCT. Mature monocyte-derived DCs were pulsed with keyhole limpet hemocyanin (KLH) and electroporated with MAGE3, Survivin or B-cell maturation antigen (BCMA) mRNA. Twelve patients were vaccinated three times with intravenous (5-22 × 10(6) DCs) and intradermal vaccines (4-11 × 10(6) DCs), at biweekly intervals. Immunological responses were monitored in blood and delayed-type hypersensitivity (DTH) biopsies. All patients developed strong anti-KLH T-cell responses, but not KLH antibodies. In 2 patients, vaccine-specific T cells were detected in DTH biopsies. In one patient, we found MAGE3-specific CD4(+) and CD8(+) T cells, and CD3(+) T cells reactive against BCMA and Survivin. In the other patient, we detected low numbers of MAGE3 and BCMA-reactive CD8(+) T cells. Vaccination was well tolerated with limited toxicity. These findings illustrate that TAA-mRNA-electroporated mature DCs are capable of inducing TAA-T-cell responses in MM patients after SCT.


Subject(s)
Cancer Vaccines/immunology , Dendritic Cells/immunology , Multiple Myeloma/immunology , RNA, Messenger/immunology , Aged , Antigens, Neoplasm/genetics , B-Cell Maturation Antigen/genetics , CD3 Complex/immunology , CD3 Complex/metabolism , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Combined Modality Therapy , Dendritic Cells/cytology , Dendritic Cells/transplantation , Female , Hemocyanins/immunology , Humans , Immunotherapy, Adoptive/methods , Inhibitor of Apoptosis Proteins/genetics , Male , Middle Aged , Monitoring, Immunologic , Multiple Myeloma/surgery , Multiple Myeloma/therapy , Neoplasm Proteins/genetics , Neoplasm, Residual/immunology , Neoplasm, Residual/surgery , Neoplasm, Residual/therapy , RNA, Messenger/genetics , Stem Cell Transplantation/methods , Survivin , Transplantation, Autologous , Treatment Outcome , Vaccination/methods
SELECTION OF CITATIONS
SEARCH DETAIL