Skip to main content
Erschienen in: Clinical Sarcoma Research 1/2015

Open Access 01.12.2015 | Research

Mesenchymal stromal cells of osteosarcoma patients do not show evidence of neoplastic changes during long-term culture

verfasst von: Emilie P Buddingh, S Eriaty N Ruslan, Christianne M A Reijnders, Karoly Szuhai, Marieke L Kuijjer, Helene Roelofs, Pancras C W Hogendoorn, R Maarten Egeler, Anne-Marie Cleton-Jansen, Arjan C Lankester

Erschienen in: Clinical Sarcoma Research | Ausgabe 1/2015

Abstract

Background

In vitro expanded mesenchymal stromal cells (MSCs) are increasingly used as experimental cellular therapy. However, there have been concerns regarding the safety of their use, particularly with regard to possible oncogenic transformation. MSCs are the hypothesized precursor cells of high-grade osteosarcoma, a tumor with often complex karyotypes occurring mainly in adolescents and young adults.

Methods

To determine if MSCs from osteosarcoma patients could be predisposed to malignant transformation we cultured MSCs of nine osteosarcoma patients and five healthy donors for an average of 649 days (range 601–679 days). Also, we compared MSCs derived from osteosarcoma patients at diagnosis and from healthy donors using genome wide gene expression profiling.

Results

Upon increasing passage, increasing frequencies of binucleate cells were detected, but no increase in proliferation suggestive of malignant transformation occurred in MSCs from either patients or donors. Hematopoietic cell specific Lyn substrate 1 (HLCS1) was differentially expressed (fold change 0.25, P value 0.0005) between MSCs of osteosarcoma patients (n = 14) and healthy donors (n = 9).

Conclusions

This study shows that although HCLS1 expression was downregulated in MSCs of osteosarcoma patients and binucleate cells were present in both patient and donor derived MSCs, there was no evidence of neoplastic changes to occur during long-term culture.
Hinweise

Electronic supplementary material

The online version of this article (doi:10.​1186/​s13569-015-0031-1) contains supplementary material, which is available to authorized users.
Anne-Marie Cleton-Jansen and Arjan C Lankester contributed equally to this work
Abkürzungen
ADM
adrenomedullin
EEF1A1
eukaryotic translation elongation factor 1 alpha 1
FBS
fetal bovine serum
FITC
fluorescein isothiocyanate
HCLS1
hematopoietic cell specific lyn substrate 1
MFI
mean fluorescence intensity
MSC
mesenchymal stromal cell
OS
osteosarcoma
PD
population doublings
PE
phycoerythrin
PerCP
peridinin chlorophyll protein
WASF4
WAS protein family member 4

Background

Mesenchymal stromal cells (MSCs) are increasingly used as experimental cellular therapy in a wide range of conditions, such as graft-versus-host disease in the context of allogeneic bone marrow transplantation, auto-immune diseases and for regenerative purposes in for example myocardial injury or cartilage defects [16]. However, since MSCs have to be expanded in vitro to achieve sufficient numbers, there have been concerns regarding the safety of their use, particularly with regard to possible oncogenic transformation [7]. Cultured murine MSCs readily transform and form sarcoma-like tumors in vivo [812]. Similarly, MSCs derived from rhesus macaques become polyploid and subsequently aneuploid during long-term culture [13]. In contrast, human MSCs appear resistant to spontaneous in vitro transformation [14]. Studies reporting that human MSCs undergo malignant transformation in vitro have been retracted because of cross-contamination issues [1520]. Despite the apparent difference between human and murine MSCs in their propensity to spontaneously transform in vitro, concerns remain. MSCs are hypothesized to be the precursor cells of high-grade osteosarcoma (OS) and a patient transplanted with bone marrow (containing hematopoietic stem cells and MSCs) from a sibling was diagnosed with OS originating from donor stem cells 17 years later [21]. This case demonstrates that donor-derived (pre-) cancerous MSCs can survive in a host and cause disease many years later. Another cause for concern is the observation that cultured MSCs can acquire chromosomal aberrations, although these do not seem to confer a selective growth advantage in vitro [22, 23].
High-grade osteosarcoma is a malignant primary bone tumor which often occurs at a relatively young age [24]. OS tumor cells are characterized by aneuploid karyotypes and gross chromosomal instability [25]. Such highly complex chromosomal rearrangements can occur as a result of a single catastrophic event, termed chromothripsis [26, 27]. However, this probably has to occur in a susceptible background, either as a genetic predisposition or acquired as a de novo event. In a murine model failed cytokinesis can lead to tetraploidy and subsequent tumorigenesis only in a p53 deficient host [28]. We previously showed loss of CDKN2A/p16 protein expression in tetraploid tumorigenic murine MSCs [9]. We hypothesized that normal MSCs from OS patients could be predisposed to malignant transformation, and performed long term in vitro culture and genome wide expression profiling of early passage MSCs from OS patients and healthy donors. Here we show that OS patient-derived MSCs do not transform in vitro, confirming previous reports in healthy individuals that spontaneous transformation of human MSCs in vitro is an extremely unlikely event.

Methods

Patients

Characteristics of OS patients and healthy stem cell donors can be found in Table 1. Bone marrow cells of OS patients were harvested under general anesthesia prior to start of the chemotherapeutic treatment. The site of MSC harvest (iliac crest) was different from the location of the primary tumor (metaphyseal ends of the long bones) in all cases. Healthy donors were either identical sibling donors for patients with malignant or benign disease requiring hematopoietic stem cell transplantation; or haploidentical donors for either hematopoietical stem cell transplantation or the therapeutic infusion of MSCs for steroid-resistant graft-versus-host disease. Written informed consent was obtained from all patients and donors prior to bone marrow harvesting. The study was approved by the Institutional Review Board on Medical Ethics [LUMC Medical Ethics Committee (CME), P06.152].
Table 1
Characteristics of osteosarcoma patients (OS) and healthy donors (HD) and overview of experiments
Culture
OS (UI)/HD
Sex
Age (years)
Histology
Location of the primary tumor
Included in experiment
Microarray
qPCR
Long-term culture
001OS
OS (352)
M
12
Osteoblastic
Distal femur
Y
Y
Y
002OS
OS (340)
F
13
Osteoblastic
Distal femur
Y
Y
Y
003OS
OS (376)
F
13
Telangiectatic
Distal femur
Y
Y
Y
004OS
OS (377)
F
14
Sclerosing
Proximal tibia
Y
Y
Y
005OS
OS (348)
F
15
Telangiectatic
Distal tibia
N
N
Y
006OS
OS (349)
M
8
Osteoblastic
Distal femur
Y
Y
Y
007OS
OS (350)
F
15
Osteoblastic
Distal femur
N
N
Y
008OS
OS (378)
F
9
Osteoblastic
Proximal humerus
Y
Y
Y
009OS
OS (382)
M
15
Chondroblastic
Proximal humerus
Y
Y
Y
010OS
OS (388)
M
14
Osteoblastic
Proximal humerus
N
Y
N
011OS
OS (391)
F
14
Osteoblastic
Proximal tibia
N
Y
N
012OS
OS (394)
F
5
Osteoblastic
Distal femur
N
Y
N
013OS
OS (393)
F
14
Osteoblastic
Distal femur
N
Y
N
014OS
OS (395)
M
10
Sclerosing
Distal femur
N
Y
N
015OS
OS (396)
M
13
Osteoblastic
Distal femur
N
Y
N
016OS
OS (402)
F
15
Sclerosing
Proximal tibia
N
Y
N
HB
HD (HB)
M
15
  
Y
Y
Y
HD3
HD (HD3)
F
27
  
Y
Y
Y
HD5
HD (HD5)
M
50
  
Y
Y
Y
MH
HD (MH)
M
15
  
Y
Y
Y
TD1
HD (TD1)
F
11
  
Y
Y
Y
TD2
HD (TD2)
F
43
  
N
Y
N
TD3
HD (TD3)
F
43
  
N
Y
N
TD4
HD (TD4)
F
25
  
N
Y
N
TD5
HD (TD5)
M
5
  
N
Y
N
UI unique identifier.

Mesenchymal stromal cell cultures

Bone marrow derived mononuclear cells were obtained from 5 to 15 mL of heparinized bone marrow aspirate by density gradient centrifugation on Ficoll. Cells were plated on non-coated 75 cm2 polystyrene flasks at a cell density of 160,000/cm2 in complete culture medium (LG-DMEM; Invitrogen, Paisley, UK) supplemented with penicillin/streptomycin (Invitrogen) and 10% fetal bovine serum (FBS; HyClone, Verviers, Belgium). We used a characterized and defined FBS batch preselected for its potential to support MSC expansion and continued to use this specific batch throughout the culture period. MSCs were plastic adherent and had spindle shaped morphology. Chondrogenic, adipogenic and osteoblastic differentiation were performed as described earlier [29]. Medium was refreshed twice a week and cells were replated when reaching 80–90% confluence at a density of 4,000/cm2. The first nine OS patient and first five healthy donor MSC samples that were obtained were cultured long-term, the subsequent samples were used for confirmatory mRNA expression analysis (Table 1). Morphology of the cells was recorded and population doublings per passage (PD) were calculated using the log ratio of the harvesting cell count (N) to the starting (baseline) count (X0), divided by the log of 2 (PD = [log (N/X0)]/log 2).

Flow cytometry

Expression of the cell surface markers CD73, CD90 and CD105 and absence of hematopoietic markers on MSCs was determined using flow cytometry, according to the statement by the International Society for Cellular Therapy [30]. Cells were detached using Trypsin/EDTA and washed in PBS/0.05% bovine serum albumin. Antibodies used were (FITC) conjugated anti-CD86-fluorescein isothiocyanate (FITC) (cat. no. 555657), anti-HLA-DR-FITC (cat. no. 347400), anti-CD31-phycoerythrin (PE) (cat no 555446), anti-CD34-PE (cat no 348057), anti-CD73-PE (cat no 550257), anti-CD90-PE (cat no 555596), anti-CD3-peridinin chlorophyll protein(PerCP)-Cy5.5 (cat no 332771), anti-CD45-PerCPCy5.5 (cat no 332784), all from BD (San Diego, CA, USA) and anti-CD105-PE (cat no SN6), from Ancell (Bayport, MN, USA). Flow cytometry was performed on a FACScalibur, and analyzed using Cellquest software (both Becton–Dickinson). Mean fluorescence intensity (MFI) ratio was calculated by determining the MFI of the specific staining relative to the MFI of the appropriate isotype control staining.

Analysis of binucleate cells

MSCs were grown on coverslips. Coverslips were washed with PBS and fixed in 4% PFA. Coverslips were incubated for 10 min in 1 μg/mL acridine orange (Sigma-Aldrich, Zwijndrecht, the Netherlands) to stain cytoplasm and 1 μg/mL wheat-germ agglutinin with an Alexa Fluor® 594 conjugate (Invitrogen) in PBS to stain cell membranes. Following washes in PBS, coverslips were mounted using Vectashield with DAPI (Vector laboratories, Burlingame, CA, USA) to stain nuclei. Images were acquired using a COHU 4910 series monochrome CCD camera (COHU, San Diego, CA, USA) attached to a DM fluorescence microscope (Leica, Wetzlar, Germany) and analyzed using ImageJ software with the Cell Counter plug-in (National Institute of Mental Health, Bethesda, MD, USA).

RNA isolation

RNA was isolated from frozen cell pellets of at least 1*10e6 undifferentiated MSCs at passage 2–5 using TRIzol reagent (Invitrogen). Cells were lysed in TRIzol, followed by phase separation in chloroform, precipitation using 2-propanolol and washing in 75% ethanol. RNA clean-up was performed using the QIAGEN Rneasy mini kit (Venlo, the Netherlands) with on-column DNAse treatment. RNA quality and concentration were measured using an Agilent 2100 Bioanalyzer (Santa Clara, CA, USA) and Nanodrop ND-1000 (Thermo Fisher Scientific, Waltham, MA, USA), respectively.

cDNA synthesis, cRNA amplification, and Illumina Human v2.0 Expression BeadChip hybridization

Gene expression profiling was performed using Human-6 v2 Expression BeadChips (San Diego, CA, USA) containing >48,000 transcript probes. Synthesis of cDNA, cRNA amplification, and hybridization of cRNA onto the Illumina Human v2.0 Expression BeadChips were performed as described previously [31, 32].

qPCR

cDNA synthesis for qPCR was performed as described earlier [32]. qPCR was performed using the iQ™ SYBR® Green Supermix (Bio-Rad, Hercules, CA, USA) according to the manufacture’s instructions, reverse transcriptase negative (RT−) and H2O controls were taken along for each sample. For normalization, three genes with stable expression in the microarray experiments were chosen (CPSF6, GPR108 and CAPNS1). Data were normalized by geometric mean expression levels of the three reference genes using geNorm (http://​medgen.​ugent.​be/​jvdesomp/​genorm/​). Primer sequences can be found in Additional file 1: Table S1. A standard curve was taken along for each primer set and used for quantification, PCR efficiencies ranged from 93 to 104%.

Karyotyping

Six early passage samples (three derived from OS patients, three derived from healthy donors) were subjected to a multicolor FISH based karyotyping test (COBRA-FISH) as described earlier [33]. From each sample at least 20 metaphase cells were recorded and analyzed.

Statistical analysis

Microarray data were normalized using the Cubic Spline normalization method with the Illumina BeadStudio Gene Expression Module. Microarray data are available at GEO using the accession no. GSE42572. Statistical analysis of microarray was performed using Significance Analysis for Microarrays, using a false discovery rate of 20% (SAM, http://​www-stat.​stanford.​edu/​~tibs/​SAM/​) [34]. Univariate statistical analyses were performed using GraphPad Prism software (version 5.01, La Jolla, CA, USA). Two-sided P values lower than 0.05 were determined to be significant.

Results

MSCs of OS patients and healthy donors do not differ in expression of cell surface markers or differentiation capacity

We tested the MSC cultures from OS patients and healthy donors for phenotypic markers and functionality. Samples from 16/16 patients and 9/9 controls were able to differentiate into chondrogenic (Additional file 2: Figure S1A), adipogenic (Additional file 2: Figures S1B and C) and osteoblastic lineages (Additional file 2: Figures S1D and E). Also, all MSC cultures expressed CD73, CD90 and CD105 and lacked expression of hematopoietic markers (Additional file 2: Figure S1F). Level of expression of CD73, CD90 and CD105 as determined by MFI-ratios (specific staining/isotype control) did not differ between MSC cultures derived from OS patients and healthy donors. Early passage samples from three OS patient derived MSCs and three healthy donor derived MSCs were karyotyped; no structural or numerical aberrations were observed in the analyzed samples (Additional file 3: Figure S2): MSC001: 46,XY (26 metaphases), MSC003: 46,XX (27 metaphases), MSC008:46,XX (22 metaphases), MSC-HB: 46,XY (21 metaphases), MSC-MH: 46,XY (20 metaphases), MSC-TD: 46,XX (21 metaphases). Late passage cells could not be karyotypically analyzed due to the low numbers of dividing cells.

Long-term in vitro culture of MSCs results in increased binucleation but not in malignant transformation

MSCs of nine OS patients and five healthy donors were long term cultured (average number of days in culture 649, range 601–679 days). From each individual sample, duplicate cultures were established. There were no significant differences between growth rate, cumulative population doublings (median cumulative population doublings OS patients 34 vs. healthy donors 39; P value Mann–Whitney U test 0.70), passage number at termination of culture (mean passage number OS patients 21 vs. healthy donors 23; P value Mann–Whitney U test 0.74) or time to growth arrest (median days to growth arrest OS patients 441 days vs. healthy donors 222 days; P value Mann–Whitney U test 0.15) between MSCs of OS patients and healthy donors. The cumulative population doublings are shown in Figure 1 (averages of the duplicate cultures are shown per sample). All cultures exhibited rapid exponential growth in the first few passages. Later, proliferation slowed down and eventually stopped, characteristic of cultured cells in crisis. At termination of the cultures, viable cells were present in all samples. Cultures were terminated at the end of the growth curves. At this point, in none of the cultures there was evidence of cells escaping the crisis by an increase in proliferation and corresponding morphological changes indicative for spontaneous in vitro malignant transformation.
Morphology of the cells was inspected with every passage. Low passage cells were spindle-shaped, higher passage cells were larger. Upon increasing passage number, increasing frequencies of binucleate cells were noted using phase contrast microscopy. To quantify binucleation, cells were grown on coverslips and cytoplasm, cell membranes and nuclei were stained. Mono- and binucleate cells were counted (Figure 2a–d). There was no difference in percentage of binucleate cells between MSCs derived from OS patients and healthy donors (Figure 2e).

HCLS1 mRNA expression is downregulated in OS patient derived MSCs

Using microarray gene expression analysis, five genes were found to be differentially expressed between early passage healthy donor derived MSCs (n = 5) and OS patient derived MSCs (n = 7), with a false discovery rate of 20%. Expression of HCLS1, ADM, EEF1A1, LOC644739 (or WASF4) and LOC441155 was lower in patient derived MSCs as compared to healthy donor derived MSCs. To perform simultaneous technical and biological validation of the microarray results, the original low passage MSC samples were recultured and the original series expanded to now include four additional healthy donor derived MSCs (total of n = 9) and seven additional OS patient derived MSCs (total of n = 14). Four of the differentially expressed genes were validated using qPCR: hematopoietic cell specific Lyn substrate 1 (HCLS1), adrenomedullin (ADM), eukaryotic translation elongation factor 1 alpha 1 (EEF1A1) and LOC644739 (or WAS protein family, member 4; WASF4). LOC441155 is a pseudogene for which we did not succeed in designing a sufficiently specific and efficient primer pair. In this second, newly cultured and expanded series, expression of HCLS1 was significantly lower in OS patient derived MSCs as compared to healthy donor derived MSCs (Figure 3, fold change 0.25, P value 0.0005). The other genes were not differentially expressed in this series, perhaps because this was a newly expanded batch of cells. We tried to determine if the observed difference in HCLS1 expression was also true at the protein level. Although HCLS1 protein could be clearly detected in the positive control cell line Jurkat, we were unable to reproducibly detect HCLS1 in MSCs. For five OS patients we had mRNA available from both bone-marrow derived MSCs and the primary tumor. In these samples, expression of HCLS1 was higher in tumor tissue as compared to MSCs, but there was no correlation between level of expression of HCLS1 in low passage MSCs and in the corresponding tumor tissue.

Discussion

In recent years, MSCs have gained increasing interest as a therapeutic modality for immune modulatory and regenerative purposes [14]. In order to achieve sufficient numbers of cells for therapeutic utility, MSCs need to be expanded in vitro prior to infusion. Since long-term culture of non-human MSCs can result in oncogenic transformation and the occurrence of sarcomas in the receiving hosts, there is ongoing debate regarding the safety of using cultured MSCs in clinical studies [35]. In the present study, we compared human MSCs derived from OS patients at diagnosis and from healthy donors to answer two questions. First, do human MSCs transform to a malignant phenotype when cultured in vitro, as has been shown for murine MSCs? Second, are MSCs derived from OS patients more likely to transform than healthy donor derived MSCs?
MSCs from patients and controls exhibited similar growth patterns during long-term in vitro culture. During this time, all cultures reached a plateau in proliferation characteristic of cells in crisis. In contrast to what we have previously observed in murine MSCs [9], we never observed escape of this crisis by a rapidly dividing cell population with changed morphology, even after almost 2 years of continuous culture. There were no phenotypical or functional differences between OS patient derived and healthy donor derived MSCs and early passage karyotypes were normal in both groups. Differentiation capacity and expression of specific cell surface markers was similar.
During prolonged culture, progressive shortening of telomeres occurs [36]. This can cause anaphase chromatin bridges, resulting in failed cytokinesis and consequently in binucleate cells [37]. In the context of loss of expression or function of tumor suppressor proteins and corresponding cell cycle checkpoints, tetraploidy and ultimately aneuploidy will occur [28]. Upon increasing passage, increasing frequencies of binucleate cells were noted; both in patient derived MSCs and healthy donor derived MSCs (Figure 2). However, since we did not observe an increase in proliferation in these cells despite the presence of increasingly high frequencies of binucleate cells, cell cycle checkpoints were probably functionally intact, both in MSCs derived from OS patients and from healthy donors.
In addition to the functional read-out of long-term culture, we also performed gene expression analysis on early passage cells. Expression of HCLS1 was downregulated in MSCs from OS patients as compared to MSCs of healthy donors. HCLS1 is primarily known for its role in the signaling cascade that follows B cell receptor activation. It is highly expressed in B-cell derived malignancies and is associated with a poor outcome [38, 39]. Further studies are needed to determine if HCLS1 might have a tumor suppressor function and if its loss of expression in OS patients derived MSCs has any relationship to in vivo tumorigenesis. Although we show no functional difference between healthy donor and OS patient derived MSCs, we do not advocate the use of MSCs from known cancer patients for clinical purposes. The relevance of the differential expression of HCLS1 remains unknown and there might be other undetected (pre-) malignant alterations.
Both on a transcriptional and on a functional level, OS patient and healthy donor derived MSCs were very similar. There are several possible explanations for the observed similarities between OS patient derived and donor derived MSCs. First, we obtained MSCs from the iliac crest, while all tumors in our series were located at the metaphyseal ends of the long bones. OS occurs at a time and place of active growth and perhaps this pro-proliferative microenvironment (with high expression of growth factors) is an essential prerequisite for oncogenic transformation of MSCs. Second, (pre-) oncogenic alterations may be present in only one or a few local mesenchymal tumor precursor cells and not in MSCs at a distant site such as the iliac crest. This could be due to somatic mosaicism, similar to what has been shown for the enchondromatosis syndromes Ollier disease and Maffucci syndrome [40]. Third, according to the ‘multiple hit hypothesis’, there may not have been sufficient ‘hits’ for in vitro transformation to occur, even if relevant predisposing alterations are present. Ionizing radiation is a well-known risk factor for developing osteosarcoma and low-dose radiation facilitates oncogenic transformation in telomerase-transduced immortalized MSCs [41, 42]. Perhaps one or two additional ‘hits’ (for example loss of cell cycle checkpoint control or radiation-induced DNA damage) would be enough for oncogenic transformation to occur.
Fourth, pre-malignant alterations may have been present in only a few cells, which would not lead to large enough differences in gene expression to be picked up by genome wide expression analysis. Novel techniques to study genetic alterations at a single cell level might be able to pick up these rare events [43, 44]. Finally, although there is compelling evidence to suggest MSCs are the precursor cells for human OS, this has not yet been unequivocally proven.

Conclusions

During long-term in vitro culture of human OS patient and healthy donor derived MSCs, there was no evidence for neoplastic changes to occur. We could not confirm our hypothesis that MSCs of OS patients might have a higher propensity to oncogenic transformation than healthy donor derived MSCs. In contrast to what has been reported for other species, under the tested conditions, human MSCs do not easily transform. Although we cannot exclude the occurrence of low frequencies of cells with genomic alterations, we did not see a selective growth advantage of aberrant cells nor did we observe karyotypic abnormalities in low passage cells. This data supports the currently held view that administration of low passage cultured healthy donor derived human MSCs for therapeutic purposes is unlikely to result in sarcomas in the host.

Authors’ contributions

EB participated in design of the study, harvested and cultured bone marrow derived MSCs and performed and analyzed flow cytometry, differentiation assays, binucleate cell assays and qPCR, and drafted the manuscript. SR cultured bone marrow derived MSCs and performed and analyzed flow cytometry, differentiation assays, binucleate cell assays and qPCR. CR cultured bone marrow derived MSCs and performed and analyzed flow cytometry and differentiation assays. KS performed and analyzed karyotypic analyses. MK performed and analyzed genome wide profiling studies. HR harvested, cultured and analyzed BM derived MSCs. PH and RE participated in design of the study and helped to draft the manuscript. AC and AL conceived of the study, participated in the design and coordination and helped to draft the manuscript. All authors read and approved the final manuscript.

Acknowledgements

The authors like to thank Ronald Duim for technical assistance, Marco Schilham for critical review, and Maarten Morsink for help in analysis of microarray data. Also, the authors would like to thank the medical and nursing staff of the departments of pediatrics and clinical oncology for help in obtaining informed consent. This work was financially supported by EuroBoNet, a European Commission granted Network of Excellence for studying the pathology and genetics of bone tumors (Grant Number LSHC-CT-2006-018814) and by the Netherlands Organization for Health Research and Development (ZonMw, Grant Number 92003-399 to EB).

Compliance with ethical guidelines

Competing interests The authors declare that they have no competing interests.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated.
Literatur
1.
Zurück zum Zitat Le Blanc K, Frassoni F, Ball L, Locatelli F, Roelofs H, Lewis I et al (2008) Mesenchymal stem cells for treatment of steroid-resistant, severe, acute graft-versus-host disease: a phase II study. Lancet 371:1579–1586PubMedCrossRef Le Blanc K, Frassoni F, Ball L, Locatelli F, Roelofs H, Lewis I et al (2008) Mesenchymal stem cells for treatment of steroid-resistant, severe, acute graft-versus-host disease: a phase II study. Lancet 371:1579–1586PubMedCrossRef
2.
Zurück zum Zitat Uccelli A, Prockop DJ (2010) Why should mesenchymal stem cells (MSCs) cure autoimmune diseases? Curr Opin Immunol 22:768–774PubMedCrossRef Uccelli A, Prockop DJ (2010) Why should mesenchymal stem cells (MSCs) cure autoimmune diseases? Curr Opin Immunol 22:768–774PubMedCrossRef
3.
Zurück zum Zitat Orlic D, Kajstura J, Chimenti S, Jakoniuk I, Anderson SM, Li B et al (2001) Bone marrow cells regenerate infarcted myocardium. Nature 410:701–705PubMedCrossRef Orlic D, Kajstura J, Chimenti S, Jakoniuk I, Anderson SM, Li B et al (2001) Bone marrow cells regenerate infarcted myocardium. Nature 410:701–705PubMedCrossRef
4.
Zurück zum Zitat Cipriani P, Carubbi F, Liakouli V, Marrelli A, Perricone C, Perricone R et al (2013) Stem cells in autoimmune diseases: implications for pathogenesis and future trends in therapy. Autoimmun Rev 12(7):709–716PubMedCrossRef Cipriani P, Carubbi F, Liakouli V, Marrelli A, Perricone C, Perricone R et al (2013) Stem cells in autoimmune diseases: implications for pathogenesis and future trends in therapy. Autoimmun Rev 12(7):709–716PubMedCrossRef
5.
Zurück zum Zitat Ball LM, Bernardo ME, Roelofs H, Lankester A, Cometa A, Egeler RM et al (2007) Cotransplantation of ex vivo-expanded mesenchymal stem cells accelerates lymphocyte recovery and may reduce the risk of graft failure in haploidentical hematopoietic stem-cell transplantation. Blood 110:2764–2767PubMedCrossRef Ball LM, Bernardo ME, Roelofs H, Lankester A, Cometa A, Egeler RM et al (2007) Cotransplantation of ex vivo-expanded mesenchymal stem cells accelerates lymphocyte recovery and may reduce the risk of graft failure in haploidentical hematopoietic stem-cell transplantation. Blood 110:2764–2767PubMedCrossRef
6.
Zurück zum Zitat Wakitani S, Imoto K, Yamamoto T, Saito M, Murata N, Yoneda M (2002) Human autologous culture expanded bone marrow mesenchymal cell transplantation for repair of cartilage defects in osteoarthritic knees. Osteoarthritis Cartilage 10:199–206PubMedCrossRef Wakitani S, Imoto K, Yamamoto T, Saito M, Murata N, Yoneda M (2002) Human autologous culture expanded bone marrow mesenchymal cell transplantation for repair of cartilage defects in osteoarthritic knees. Osteoarthritis Cartilage 10:199–206PubMedCrossRef
7.
Zurück zum Zitat Ross AL, Leder DE, Weiss J, Izakovic J, Grichnik JM (2011) Genomic instability in cultured stem cells: associated risks and underlying mechanisms. Regen Med 6:653–662PubMedCrossRef Ross AL, Leder DE, Weiss J, Izakovic J, Grichnik JM (2011) Genomic instability in cultured stem cells: associated risks and underlying mechanisms. Regen Med 6:653–662PubMedCrossRef
8.
Zurück zum Zitat Miura M, Miura Y, Padilla-Nash HM, Molinolo AA, Fu BJ, Patel V et al (2006) Accumulated chromosomal instability in murine bone marrow mesenchymal stem cells leads to malignant transformation. Stem Cells 24:1095–1103PubMedCrossRef Miura M, Miura Y, Padilla-Nash HM, Molinolo AA, Fu BJ, Patel V et al (2006) Accumulated chromosomal instability in murine bone marrow mesenchymal stem cells leads to malignant transformation. Stem Cells 24:1095–1103PubMedCrossRef
9.
Zurück zum Zitat Mohseny AB, Szuhai K, Romeo S, Buddingh EP, Briaire-de Bruijn I, de Jong D et al (2009) Osteosarcoma originates from mesenchymal stem cells in consequence of aneuploidization and genomic loss of Cdkn2. J Pathol 219:294–305PubMedCrossRef Mohseny AB, Szuhai K, Romeo S, Buddingh EP, Briaire-de Bruijn I, de Jong D et al (2009) Osteosarcoma originates from mesenchymal stem cells in consequence of aneuploidization and genomic loss of Cdkn2. J Pathol 219:294–305PubMedCrossRef
10.
Zurück zum Zitat Tolar J, Nauta AJ, Osborn MJ, Mortari AP, McElmurry RT, Bell S et al (2007) Sarcoma derived from cultured mesenchymal stem cells. Stem Cells 25:371–379PubMedCrossRef Tolar J, Nauta AJ, Osborn MJ, Mortari AP, McElmurry RT, Bell S et al (2007) Sarcoma derived from cultured mesenchymal stem cells. Stem Cells 25:371–379PubMedCrossRef
11.
Zurück zum Zitat Jeong JO, Han JW, Kim JM, Cho HJ, Park C, Lee N et al (2011) Malignant tumor formation after transplantation of short-term cultured bone marrow mesenchymal stem cells in experimental myocardial infarction and diabetic neuropathy. Circ Res 108:1340–1347PubMedCentralPubMedCrossRef Jeong JO, Han JW, Kim JM, Cho HJ, Park C, Lee N et al (2011) Malignant tumor formation after transplantation of short-term cultured bone marrow mesenchymal stem cells in experimental myocardial infarction and diabetic neuropathy. Circ Res 108:1340–1347PubMedCentralPubMedCrossRef
12.
Zurück zum Zitat Josse C, Schoemans R, Niessen NA, Delgaudine M, Hellin AC, Herens C et al (2010) Systematic chromosomal aberrations found in murine bone marrow-derived mesenchymal stem cells. Stem Cells Dev 19:1167–1173PubMedCrossRef Josse C, Schoemans R, Niessen NA, Delgaudine M, Hellin AC, Herens C et al (2010) Systematic chromosomal aberrations found in murine bone marrow-derived mesenchymal stem cells. Stem Cells Dev 19:1167–1173PubMedCrossRef
13.
Zurück zum Zitat Izadpanah R, Kaushal D, Kriedt C, Tsien F, Patel B, Dufour J et al (2008) Long-term in vitro expansion alters the biology of adult mesenchymal stem cells. Cancer Res 68:4229–4238PubMedCentralPubMedCrossRef Izadpanah R, Kaushal D, Kriedt C, Tsien F, Patel B, Dufour J et al (2008) Long-term in vitro expansion alters the biology of adult mesenchymal stem cells. Cancer Res 68:4229–4238PubMedCentralPubMedCrossRef
14.
Zurück zum Zitat Bernardo ME, Zaffaroni N, Novara F, Cometa AM, Avanzini MA, Moretta A et al (2007) Human bone marrow-derived mesenchymal stem cells do not undergo transformation after long-term In vitro culture and do not exhibit telomere maintenance mechanisms. Cancer Res 67:9142–9149PubMedCrossRef Bernardo ME, Zaffaroni N, Novara F, Cometa AM, Avanzini MA, Moretta A et al (2007) Human bone marrow-derived mesenchymal stem cells do not undergo transformation after long-term In vitro culture and do not exhibit telomere maintenance mechanisms. Cancer Res 67:9142–9149PubMedCrossRef
15.
Zurück zum Zitat Rubio D, Garcia-Castro J, Martin MC, de la Fuente R, Cigudosa JC, Lloyd AC et al (2005) Spontaneous human adult stem cell transformation. Cancer Res 65:3035–3039PubMed Rubio D, Garcia-Castro J, Martin MC, de la Fuente R, Cigudosa JC, Lloyd AC et al (2005) Spontaneous human adult stem cell transformation. Cancer Res 65:3035–3039PubMed
16.
Zurück zum Zitat Rubio D, Garcia S, De la Cueva T, Paz MF, Lloyd AC, Bernad A et al (2008) Human mesenchymal stem cell transformation is associated with a mesenchymal-epithelial transition. Exp Cell Res 314:691–698PubMedCrossRef Rubio D, Garcia S, De la Cueva T, Paz MF, Lloyd AC, Bernad A et al (2008) Human mesenchymal stem cell transformation is associated with a mesenchymal-epithelial transition. Exp Cell Res 314:691–698PubMedCrossRef
17.
Zurück zum Zitat Rubio D, Garcia S, Paz MF, De la CT, Lopez-Fernandez LA, Lloyd AC et al (2008) Molecular characterization of spontaneous mesenchymal stem cell transformation. PLoS One 3:e1398PubMedCentralPubMedCrossRef Rubio D, Garcia S, Paz MF, De la CT, Lopez-Fernandez LA, Lloyd AC et al (2008) Molecular characterization of spontaneous mesenchymal stem cell transformation. PLoS One 3:e1398PubMedCentralPubMedCrossRef
18.
Zurück zum Zitat Rosland GV, Svendsen A, Torsvik A, Sobala E, McCormack E, Immervoll H et al (2009) Long-term cultures of bone marrow-derived human mesenchymal stem cells frequently undergo spontaneous malignant transformation. Cancer Res 69:5331–5339PubMedCrossRef Rosland GV, Svendsen A, Torsvik A, Sobala E, McCormack E, Immervoll H et al (2009) Long-term cultures of bone marrow-derived human mesenchymal stem cells frequently undergo spontaneous malignant transformation. Cancer Res 69:5331–5339PubMedCrossRef
19.
Zurück zum Zitat Torsvik A, Rosland GV, Svendsen A, Molven A, Immervoll H, McCormack E et al (2010) Spontaneous malignant transformation of human mesenchymal stem cells reflects cross-contamination: putting the research field on track-letter. Cancer Res 70:6393–6396PubMedCrossRef Torsvik A, Rosland GV, Svendsen A, Molven A, Immervoll H, McCormack E et al (2010) Spontaneous malignant transformation of human mesenchymal stem cells reflects cross-contamination: putting the research field on track-letter. Cancer Res 70:6393–6396PubMedCrossRef
20.
Zurück zum Zitat de la Fuente R, Bernad A, Garcia-Castro J, Martin MC, Cigudosa JC (2010) Retraction: spontaneous human adult stem cell transformation. Cancer Res 70:6682PubMedCrossRef de la Fuente R, Bernad A, Garcia-Castro J, Martin MC, Cigudosa JC (2010) Retraction: spontaneous human adult stem cell transformation. Cancer Res 70:6682PubMedCrossRef
21.
Zurück zum Zitat Berger M, Muraro M, Fagioli F, Ferrari S (2008) Osteosarcoma derived from donor stem cells carrying the Norrie’s disease gene. N Engl J Med 359:2502–2504PubMedCrossRef Berger M, Muraro M, Fagioli F, Ferrari S (2008) Osteosarcoma derived from donor stem cells carrying the Norrie’s disease gene. N Engl J Med 359:2502–2504PubMedCrossRef
22.
Zurück zum Zitat Ben-David U, Mayshar Y, Benvenisty N (2011) Large-scale analysis reveals acquisition of lineage-specific chromosomal aberrations in human adult stem cells. Cell Stem Cell 9:97–102PubMedCrossRef Ben-David U, Mayshar Y, Benvenisty N (2011) Large-scale analysis reveals acquisition of lineage-specific chromosomal aberrations in human adult stem cells. Cell Stem Cell 9:97–102PubMedCrossRef
23.
Zurück zum Zitat Tarte K, Gaillard J, Lataillade JJ, Fouillard L, Becker M, Mossafa H et al (2010) Clinical-grade production of human mesenchymal stromal cells: occurrence of aneuploidy without transformation. Blood 115:1549–1553PubMedCrossRef Tarte K, Gaillard J, Lataillade JJ, Fouillard L, Becker M, Mossafa H et al (2010) Clinical-grade production of human mesenchymal stromal cells: occurrence of aneuploidy without transformation. Blood 115:1549–1553PubMedCrossRef
24.
Zurück zum Zitat Fletcher CDM, Bridge JA, Hogendoorn PCW, Mertens F (2013) WHO classification of tumours of soft tissue and bone. International Agency for Research on Cancer, Lyon Fletcher CDM, Bridge JA, Hogendoorn PCW, Mertens F (2013) WHO classification of tumours of soft tissue and bone. International Agency for Research on Cancer, Lyon
25.
Zurück zum Zitat Szuhai K, Cleton-Jansen AM, Hogendoorn PC, Bovee JV (2012) Molecular pathology and its diagnostic use in bone tumors. Cancer Genet 205:193–204PubMedCrossRef Szuhai K, Cleton-Jansen AM, Hogendoorn PC, Bovee JV (2012) Molecular pathology and its diagnostic use in bone tumors. Cancer Genet 205:193–204PubMedCrossRef
26.
Zurück zum Zitat Stephens PJ, Greenman CD, Fu B, Yang F, Bignell GR, Mudie LJ et al (2011) Massive genomic rearrangement acquired in a single catastrophic event during cancer development. Cell 144:27–40PubMedCentralPubMedCrossRef Stephens PJ, Greenman CD, Fu B, Yang F, Bignell GR, Mudie LJ et al (2011) Massive genomic rearrangement acquired in a single catastrophic event during cancer development. Cell 144:27–40PubMedCentralPubMedCrossRef
27.
Zurück zum Zitat Holland AJ, Cleveland DW (2012) Chromoanagenesis and cancer: mechanisms and consequences of localized, complex chromosomal rearrangements. Nat Med 18:1630–1638PubMedCentralPubMedCrossRef Holland AJ, Cleveland DW (2012) Chromoanagenesis and cancer: mechanisms and consequences of localized, complex chromosomal rearrangements. Nat Med 18:1630–1638PubMedCentralPubMedCrossRef
28.
Zurück zum Zitat Fujiwara T, Bandi M, Nitta M, Ivanova EV, Bronson RT, Pellman D (2005) Cytokinesis failure generating tetraploids promotes tumorigenesis in p53-null cells. Nature 437:1043–1047PubMedCrossRef Fujiwara T, Bandi M, Nitta M, Ivanova EV, Bronson RT, Pellman D (2005) Cytokinesis failure generating tetraploids promotes tumorigenesis in p53-null cells. Nature 437:1043–1047PubMedCrossRef
29.
Zurück zum Zitat Bernardo ME, Emons JAM, Karperien M, Nauta AJ, Willemze R, Roelofs H et al (2007) Human mesenchymal stem cells derived from bone marrow display a better chondrogenic differentiation compared with other sources. Connect Tissue Res 48:132–140PubMedCrossRef Bernardo ME, Emons JAM, Karperien M, Nauta AJ, Willemze R, Roelofs H et al (2007) Human mesenchymal stem cells derived from bone marrow display a better chondrogenic differentiation compared with other sources. Connect Tissue Res 48:132–140PubMedCrossRef
30.
Zurück zum Zitat Dominici M, Le Blanc K, Mueller I, Slaper-Cortenbach I, Marini FC, Krause DS et al (2006) Minimal criteria for defining multipotent mesenchymal stromal cells. The International Society for Cellular Therapy position statement. Cytotherapy 8:315–317PubMedCrossRef Dominici M, Le Blanc K, Mueller I, Slaper-Cortenbach I, Marini FC, Krause DS et al (2006) Minimal criteria for defining multipotent mesenchymal stromal cells. The International Society for Cellular Therapy position statement. Cytotherapy 8:315–317PubMedCrossRef
31.
Zurück zum Zitat Hallor KH, Staaf J, Bovee JVMG, Hogendoorn PCW, Cleton-Jansen AM, Knuutila S et al (2009) Genomic profiling of chondrosarcoma: chromosomal patterns in central and peripheral tumors. Clin Cancer Res 15:2685–2694PubMedCrossRef Hallor KH, Staaf J, Bovee JVMG, Hogendoorn PCW, Cleton-Jansen AM, Knuutila S et al (2009) Genomic profiling of chondrosarcoma: chromosomal patterns in central and peripheral tumors. Clin Cancer Res 15:2685–2694PubMedCrossRef
32.
Zurück zum Zitat Buddingh EP, Kuijjer ML, Duim RA, Burger H, Agelopoulos K, Myklebost O et al (2011) Tumor-infiltrating macrophages are associated with metastasis suppression in high-grade osteosarcoma: a rationale for treatment with macrophage activating agents. Clin Cancer Res 17:2110–2119PubMedCrossRef Buddingh EP, Kuijjer ML, Duim RA, Burger H, Agelopoulos K, Myklebost O et al (2011) Tumor-infiltrating macrophages are associated with metastasis suppression in high-grade osteosarcoma: a rationale for treatment with macrophage activating agents. Clin Cancer Res 17:2110–2119PubMedCrossRef
33.
Zurück zum Zitat Szuhai K, Tanke HJ (2006) COBRA: combined binary ratio labeling of nucleicacid probes for multi-color fluorescence in situ hybridization karyotyping. Nat Protoc 1:264–275PubMedCrossRef Szuhai K, Tanke HJ (2006) COBRA: combined binary ratio labeling of nucleicacid probes for multi-color fluorescence in situ hybridization karyotyping. Nat Protoc 1:264–275PubMedCrossRef
34.
Zurück zum Zitat Tusher VG, Tibshirani R, Chu G (2001) Significance analysis of microarrays applied to the ionizing radiation response. Proc Natl Acad Sci USA 98:5116–5121PubMedCentralPubMedCrossRef Tusher VG, Tibshirani R, Chu G (2001) Significance analysis of microarrays applied to the ionizing radiation response. Proc Natl Acad Sci USA 98:5116–5121PubMedCentralPubMedCrossRef
35.
Zurück zum Zitat Prockop DJ, Keating A (2012) Relearning the lessons of genomic stability of human cells during expansion in culture: implications for clinical research. Stem Cells 30:1051–1052PubMedCrossRef Prockop DJ, Keating A (2012) Relearning the lessons of genomic stability of human cells during expansion in culture: implications for clinical research. Stem Cells 30:1051–1052PubMedCrossRef
36.
Zurück zum Zitat Kim J, Kang JW, Park JH, Choi Y, Choi KS, Park KD et al (2009) Biological characterization of long-term cultured human mesenchymal stem cells. Arch Pharm Res 32:117–126PubMedCrossRef Kim J, Kang JW, Park JH, Choi Y, Choi KS, Park KD et al (2009) Biological characterization of long-term cultured human mesenchymal stem cells. Arch Pharm Res 32:117–126PubMedCrossRef
37.
Zurück zum Zitat Pampalona J, Frias C, Genesca A, Tusell L (2012) Progressive telomere dysfunction causes cytokinesis failure and leads to the accumulation of polyploid cells. PLoS Genet 8:e1002679PubMedCentralPubMedCrossRef Pampalona J, Frias C, Genesca A, Tusell L (2012) Progressive telomere dysfunction causes cytokinesis failure and leads to the accumulation of polyploid cells. PLoS Genet 8:e1002679PubMedCentralPubMedCrossRef
38.
Zurück zum Zitat Scielzo C, Ghia P, Conti A, Bachi A, Guida G, Geuna M et al (2005) HS1 protein is differentially expressed in chronic lymphocytic leukemia patient subsets with good or poor prognoses. J Clin Invest 115:1644–1650PubMedCentralPubMedCrossRef Scielzo C, Ghia P, Conti A, Bachi A, Guida G, Geuna M et al (2005) HS1 protein is differentially expressed in chronic lymphocytic leukemia patient subsets with good or poor prognoses. J Clin Invest 115:1644–1650PubMedCentralPubMedCrossRef
39.
Zurück zum Zitat Frezzato F, Gattazzo C, Martini V, Trimarco V, Teramo A, Carraro S et al (2012) HS1, a Lyn kinase substrate, is abnormally expressed in B-chronic lymphocytic leukemia and correlates with response to fludarabine-based regimen. PLoS One 7:e39902PubMedCentralPubMedCrossRef Frezzato F, Gattazzo C, Martini V, Trimarco V, Teramo A, Carraro S et al (2012) HS1, a Lyn kinase substrate, is abnormally expressed in B-chronic lymphocytic leukemia and correlates with response to fludarabine-based regimen. PLoS One 7:e39902PubMedCentralPubMedCrossRef
40.
Zurück zum Zitat Pansuriya TC, van ER, d’Adamo P, van Ruler MA, Kuijjer ML, Oosting J et al (2011) Somatic mosaic IDH1 and IDH2 mutations are associated with enchondroma and spindle cell hemangioma in Ollier disease and Maffucci syndrome. Nat Genet 43:1256–1261PubMedCentralPubMedCrossRef Pansuriya TC, van ER, d’Adamo P, van Ruler MA, Kuijjer ML, Oosting J et al (2011) Somatic mosaic IDH1 and IDH2 mutations are associated with enchondroma and spindle cell hemangioma in Ollier disease and Maffucci syndrome. Nat Genet 43:1256–1261PubMedCentralPubMedCrossRef
41.
Zurück zum Zitat Christensen R, Alsner J, Brandt SF, Dagnaes-Hansen F, Kolvraa S, Serakinci N (2008) Transformation of human mesenchymal stem cells in radiation carcinogenesis: long-term effect of ionizing radiation. Regen Med 3:849–861PubMedCrossRef Christensen R, Alsner J, Brandt SF, Dagnaes-Hansen F, Kolvraa S, Serakinci N (2008) Transformation of human mesenchymal stem cells in radiation carcinogenesis: long-term effect of ionizing radiation. Regen Med 3:849–861PubMedCrossRef
42.
Zurück zum Zitat Ottaviani G, Jaffe N (2009) The etiology of osteosarcoma. Cancer Treat Res 152:15–32PubMed Ottaviani G, Jaffe N (2009) The etiology of osteosarcoma. Cancer Treat Res 152:15–32PubMed
43.
Zurück zum Zitat Navin N, Kendall J, Troge J, Andrews P, Rodgers L, McIndoo J et al (2011) Tumour evolution inferred by single-cell sequencing. Nature 472:90–94PubMedCrossRef Navin N, Kendall J, Troge J, Andrews P, Rodgers L, McIndoo J et al (2011) Tumour evolution inferred by single-cell sequencing. Nature 472:90–94PubMedCrossRef
44.
Zurück zum Zitat Zamani EM, Dimitriadou E, Mateiu L, Melotte C, Van der Aa N, Kumar P et al (2015) Concurrent whole-genome haplotyping and copy-number profiling of single cells. Am J Hum Genet 96(6):894–912CrossRef Zamani EM, Dimitriadou E, Mateiu L, Melotte C, Van der Aa N, Kumar P et al (2015) Concurrent whole-genome haplotyping and copy-number profiling of single cells. Am J Hum Genet 96(6):894–912CrossRef
Metadaten
Titel
Mesenchymal stromal cells of osteosarcoma patients do not show evidence of neoplastic changes during long-term culture
verfasst von
Emilie P Buddingh
S Eriaty N Ruslan
Christianne M A Reijnders
Karoly Szuhai
Marieke L Kuijjer
Helene Roelofs
Pancras C W Hogendoorn
R Maarten Egeler
Anne-Marie Cleton-Jansen
Arjan C Lankester
Publikationsdatum
01.12.2015
Verlag
BioMed Central
Erschienen in
Clinical Sarcoma Research / Ausgabe 1/2015
Elektronische ISSN: 2045-3329
DOI
https://doi.org/10.1186/s13569-015-0031-1

Weitere Artikel der Ausgabe 1/2015

Clinical Sarcoma Research 1/2015 Zur Ausgabe

Umsetzung der POMGAT-Leitlinie läuft

03.05.2024 DCK 2024 Kongressbericht

Seit November 2023 gibt es evidenzbasierte Empfehlungen zum perioperativen Management bei gastrointestinalen Tumoren (POMGAT) auf S3-Niveau. Vieles wird schon entsprechend der Empfehlungen durchgeführt. Wo es im Alltag noch hapert, zeigt eine Umfrage in einem Klinikverbund.

CUP-Syndrom: Künstliche Intelligenz kann Primärtumor finden

30.04.2024 Künstliche Intelligenz Nachrichten

Krebserkrankungen unbekannten Ursprungs (CUP) sind eine diagnostische Herausforderung. KI-Systeme können Pathologen dabei unterstützen, zytologische Bilder zu interpretieren, um den Primärtumor zu lokalisieren.

Sind Frauen die fähigeren Ärzte?

30.04.2024 Gendermedizin Nachrichten

Patienten, die von Ärztinnen behandelt werden, dürfen offenbar auf bessere Therapieergebnisse hoffen als Patienten von Ärzten. Besonders gilt das offenbar für weibliche Kranke, wie eine Studie zeigt.

Adjuvante Immuntherapie verlängert Leben bei RCC

25.04.2024 Nierenkarzinom Nachrichten

Nun gibt es auch Resultate zum Gesamtüberleben: Eine adjuvante Pembrolizumab-Therapie konnte in einer Phase-3-Studie das Leben von Menschen mit Nierenzellkarzinom deutlich verlängern. Die Sterberate war im Vergleich zu Placebo um 38% geringer.

Update Onkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.