Skip to main content
Erschienen in: Journal of Hematology & Oncology 1/2020

Open Access 01.12.2020 | Letter to the Editor

Discovery of proangiogenic CD44+mesenchymal cancer stem cells in an acute myeloid leukemia patient’s bone marrow

verfasst von: Huynh Cao, Jeffrey Xiao, Mark E. Reeves, Kimberly Payne, Chien Shing Chen, David J. Baylink, Guido Marcucci, Yi Xu

Erschienen in: Journal of Hematology & Oncology | Ausgabe 1/2020

Abstract

Here, we report a unique acute myeloid leukemia (AML) bone marrow-derived mesenchymal stem cell (MSC) with both mesenchymal and endothelial potential, which we have named Mesenchymal Cancer Stem Cells (MCSCs). These MCSCs are CD90-CD13-CD44+ and differ from MSCs in isolation, expansion, differentiation, immunophenotype, and cytokine release profile. Furthermore, blocking CD44 inhibited the proliferation and cluster formation of early MCSCs with lower ICAM-1 protein levels. Similar CD90-CD44+ cancer stem cells have been reported in both gastric and breast cancers, which grew in floating spheres in vitro and exhibited mesenchymal features and high metastatic/tumorigenic capabilities in vivo. Our novel discovery provides the first evidence that certain AMLs may be comprised of both hematopoietic and stromal malignant cells. Targeting MCSCs and their cytokine release has potential as a novel therapeutic approach in AML.
Begleitmaterial
Hinweise
Huynh Cao and Jeffrey Xiao are co-first authors.

Supplementary information

Supplementary information including Materials and Methods (Additional file 2) accompanies this paper at https://​doi.​org/​10.​1186/​s13045-020-00899-x.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Abkürzungen
AML
Acute myeloid leukemia
MSC
Mesenchymal stem cell
MCSCs
Mesenchymal cancer stem cells
BM
Bone marrow
HSCs
Hematopoietic stem cells
FACS
Flow cytometry
P
Passages
To the Editor
Acute myeloid leukemia (AML) most commonly occurs in older adults [1] and has the lowest survival rate among all types of leukemia [2]. Mechanistically, AML is a hematopoietic cancer that involves a complex interplay among different types of bone marrow (BM) cells, cytokines, growth factors, and neural modulation—all of which contribute to the survival and growth of malignant Hematopoietic Stem Cells (HSCs) [3, 4]. Currently, most AML therapies have been developed to target malignant HSCs because of their significant role in initiating uncontrolled clonal proliferation and transforming the naïve BM niche to allow disease progression and treatment resistance [5]. However, the overall survival of relapsed or refractory AML patients has remained dismal for the past three decades [6]. Therefore, more research on the mechanisms of AML etiology and relapse are urgently needed to further the development of effective treatments.
A new concept of niche-induced oncogenesis has been proposed that is based on the observation that artificial genetic perturbation of mesenchymal stem cells (MSCs), and their progenies leads to leukemogenesis of myelodysplasia and AML in transgenic mice [7, 8]. In this study from an AML patient BM, for the first time, we discovered previously unrecognized CD90-CD13-CD44+ MSCs with tumorigenic proliferation and proangiogenic properties ex vivo.
The AML bone marrow donor is a 32-year-old female with a past medical history of diabetes and hypothyroidism. She was admitted to the emergency department with intermittent chest pain, shortness of breath, and gum bleeding, and diagnosed with AML with myelomonocytic differentiation. Peripheral blood smears showed leukocytosis with ~ 52% blasts/equivalents and marked thrombocytopenia. FISH showed normal cytogenetics. An AML panel was positive for mutations of DNMT3A, NPM1, and NRAS without FLT3-ITD.
During ex vivo isolation of MSCs from this patient BM, we observed two different cell populations. The early plastic-attached cells were CD90+CD13+CD44+ MSCs (Fig. 1(a1, a3, d1 upper panels)). The late attached cells in the non-adherent floating cells of MSC cultures became adherent after 7 or more days (Fig. 1(a2)). We named them mesenchymal cancer stem cells (MCSCs) because of their capabilities of multiple lineage differentiation (Supplementary Figure 1), rapid cell proliferation and self-renewal (Fig. 1b, c, Supplementary Figure 2), and release of high amounts of cytokines and growth factors reported to be essential for angiogenesis and blasts’ growth. At early passages (P), there were about 20% CD90-CD13-CD44+ cells within large-sized strong CD44+ MCSCs (Fig. 1d lower panels). MCSCs grew in plastic attachment patterns; however, the number of viable floating cells increased with passage (Fig. 1(A4)). After P7, most MCSCs were CD90-CD13-CD44+, expanded in floating cluster conditions (Fig. 1g, i), and generated tube-like vessel structures (Fig. 1j) by expressing endothelial cell biomarkers like VE-Cadherin/CD144 (Fig. 1k). To understand the interaction between MCSCs and AML blasts, we plated MCSCs or MSCs with GFP-MOLM-14 (AML blasts) together. After 24 h of 1:1 co-cultures, GFP-MOLM-14 were found in the middle of floating clusters and surrounded by MCSCs (Fig. 1m), in contrast to their position on top of MSCs when cultured with MSCs only (Fig. 1l). These data are consistent with a previous report that AML blasts integrate into vascular niches to progress and metastasize [9].
Finally, we performed a proteome assay of supernatants from MSC and MCSC cultures. The release of key angiogenic cytokines like VEGF and growth factors such as FGF basic and PDGF were significantly increased in the P7 MCSC culture (Fig. 2b). To evaluate therapeutic potential, we performed blocking experiments with anti-CD44 monoclonal antibodies, which stopped tumorigenic proliferation of P5 MCSCs, but not P7 MCSCs (Supplementary Figure 3). It is possible that the large amount of ICAM-1 (53-fold increase in P7 MCSCs versus P5 MCSCs, Supplementary Figure 3E) compensated the loss of CD44 as previously reported in CD44 null mice [10].
In summary, we provide novel evidence of MCSCs as the cellular origin of angiogenesis and cytokine release in an AML patient’s BM. More research with additional patients is needed to discover if this phenomenon is unique to this one AML patient or has broader ramifications for AML as a whole. Our data suggest that future therapeutic strategies should also be developed to target MCSCs and their cytokine release to achieve stringent complete remission and prevent AML progression and relapse. The existence of MCSCs might be applicable to other cancers as well.

Supplementary information

Supplementary information including Materials and Methods (Additional file 2) accompanies this paper at https://​doi.​org/​10.​1186/​s13045-020-00899-x.

Acknowledgements

We thank Drs. Chung-Tsen Hsueh and Saied Mirshahidi for helpful comments.
This research was approved by the Loma Linda University Institutional Review Board. The sample was provided upon written informed consent.
Not applicable

Competing interests

The authors have filed for a patent (62/947,118).
Open AccessThis article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by/​4.​0/​. 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 in a credit line to the data.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Unsere Produktempfehlungen

e.Med Interdisziplinär

Kombi-Abonnement

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

e.Med Innere Medizin

Kombi-Abonnement

Mit e.Med Innere Medizin erhalten Sie Zugang zu CME-Fortbildungen des Fachgebietes Innere Medizin, den Premium-Inhalten der internistischen Fachzeitschriften, inklusive einer gedruckten internistischen Zeitschrift Ihrer Wahl.

Anhänge

Supplementary information

Literatur
2.
Zurück zum Zitat Medeiros BC, et al. Big data analysis of treatment patterns and outcomes among elderly acute myeloid leukemia patients in the United States. Ann Hematol. 2015;94(7):1127–38.CrossRef Medeiros BC, et al. Big data analysis of treatment patterns and outcomes among elderly acute myeloid leukemia patients in the United States. Ann Hematol. 2015;94(7):1127–38.CrossRef
3.
Zurück zum Zitat Bruserud O, et al. Subclassification of patients with acute myelogenous leukemia based on chemokine responsiveness and constitutive chemokine release by their leukemic cells. Haematologica. 2007;92(3):332–41.CrossRef Bruserud O, et al. Subclassification of patients with acute myelogenous leukemia based on chemokine responsiveness and constitutive chemokine release by their leukemic cells. Haematologica. 2007;92(3):332–41.CrossRef
4.
Zurück zum Zitat Kittang AO, et al. The chemokine network in acute myelogenous leukemia: molecular mechanisms involved in leukemogenesis and therapeutic implications. Curr Top Microbiol Immunol. 2010;341:149–72.PubMed Kittang AO, et al. The chemokine network in acute myelogenous leukemia: molecular mechanisms involved in leukemogenesis and therapeutic implications. Curr Top Microbiol Immunol. 2010;341:149–72.PubMed
5.
Zurück zum Zitat Kumar B, et al. Acute myeloid leukemia transforms the bone marrow niche into a leukemia-permissive microenvironment through exosome secretion. Leukemia. 2018;32(3):575–87.CrossRef Kumar B, et al. Acute myeloid leukemia transforms the bone marrow niche into a leukemia-permissive microenvironment through exosome secretion. Leukemia. 2018;32(3):575–87.CrossRef
6.
Zurück zum Zitat Ganzel C, et al. Very poor long-term survival in past and more recent studies for relapsed AML patients: The ECOG-ACRIN experience. Am J Hematol. 2018;93:1074–81. Ganzel C, et al. Very poor long-term survival in past and more recent studies for relapsed AML patients: The ECOG-ACRIN experience. Am J Hematol. 2018;93:1074–81.
7.
Zurück zum Zitat Kode A, et al. Leukaemogenesis induced by an activating beta-catenin mutation in osteoblasts. Nature. 2014;506(7487):240–4.CrossRef Kode A, et al. Leukaemogenesis induced by an activating beta-catenin mutation in osteoblasts. Nature. 2014;506(7487):240–4.CrossRef
8.
Zurück zum Zitat Raaijmakers MH, et al. Bone progenitor dysfunction induces myelodysplasia and secondary leukaemia. Nature. 2010;464(7290):852–7.CrossRef Raaijmakers MH, et al. Bone progenitor dysfunction induces myelodysplasia and secondary leukaemia. Nature. 2010;464(7290):852–7.CrossRef
9.
Zurück zum Zitat Cogle CR, et al. Functional integration of acute myeloid leukemia into the vascular niche. Leukemia. 2014;28(10):1978–87.CrossRef Cogle CR, et al. Functional integration of acute myeloid leukemia into the vascular niche. Leukemia. 2014;28(10):1978–87.CrossRef
10.
Zurück zum Zitat Olaku V, et al. c-Met recruits ICAM-1 as a coreceptor to compensate for the loss of CD44 in Cd44 null mice. Mol Biol Cell. 2011;22(15):2777–86.CrossRef Olaku V, et al. c-Met recruits ICAM-1 as a coreceptor to compensate for the loss of CD44 in Cd44 null mice. Mol Biol Cell. 2011;22(15):2777–86.CrossRef
Metadaten
Titel
Discovery of proangiogenic CD44+mesenchymal cancer stem cells in an acute myeloid leukemia patient’s bone marrow
verfasst von
Huynh Cao
Jeffrey Xiao
Mark E. Reeves
Kimberly Payne
Chien Shing Chen
David J. Baylink
Guido Marcucci
Yi Xu
Publikationsdatum
01.12.2020
Verlag
BioMed Central
Erschienen in
Journal of Hematology & Oncology / Ausgabe 1/2020
Elektronische ISSN: 1756-8722
DOI
https://doi.org/10.1186/s13045-020-00899-x

Weitere Artikel der Ausgabe 1/2020

Journal of Hematology & Oncology 1/2020 Zur Ausgabe

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.

Alectinib verbessert krankheitsfreies Überleben bei ALK-positivem NSCLC

25.04.2024 NSCLC Nachrichten

Das Risiko für Rezidiv oder Tod von Patienten und Patientinnen mit reseziertem ALK-positivem NSCLC ist unter einer adjuvanten Therapie mit dem Tyrosinkinase-Inhibitor Alectinib signifikant geringer als unter platinbasierter Chemotherapie.

Bei Senioren mit Prostatakarzinom auf Anämie achten!

24.04.2024 DGIM 2024 Nachrichten

Patienten, die zur Behandlung ihres Prostatakarzinoms eine Androgendeprivationstherapie erhalten, entwickeln nicht selten eine Anämie. Wer ältere Patienten internistisch mitbetreut, sollte auf diese Nebenwirkung achten.

ICI-Therapie in der Schwangerschaft wird gut toleriert

Müssen sich Schwangere einer Krebstherapie unterziehen, rufen Immuncheckpointinhibitoren offenbar nicht mehr unerwünschte Wirkungen hervor als andere Mittel gegen Krebs.

Update Onkologie

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