Previous Page  30 / 38 Next Page
Information
Show Menu
Previous Page 30 / 38 Next Page
Page Background

J u n e 1 1 - 1 3 , 2 0 1 8 | D u b l i n , I r e l a n d

allied

academies

Page 54

CANCER STEM CELLS AND

ONCOLOGY RESEARCH

11

th

International Conference on

Journal of Medical Oncology and Therapeutics

|

Volume 3

Miaofen G Hu et al., J Med Oncl Ther 2018, Volume 3

CDK6-MEDIATED SUPPRESSION

OF CD25 IS REQUIRED FOR SELF-

RENEWAL OF LSCS

Miaofen G Hu

1

, Nilamani Jena

1, 2

, Alexander J Hu

1, 3

, Wei Li

1

,

Jamie K Hu

1, 4

and

Richard Van Etten

1,2

1

Tufts Medical Center, USA

2

University of California Irvine, USA

3

Tufts University School of Medicine, USA

4

Yale School of Medicine, USA

D

espite recent advances in chemotherapy, relapse is frequent, possibly

because the available therapies do not eradicate the cells that initiate

and sustain the disease

in vivo

, so-called leukemia stem cells (LSCs).

Cyclin-dependent kinase 6 (CDK6) regulates cell cycle progression and

modulates differentiation of certain cells. It is predominantly expressed

in hematopoietic cells and over-expressed in human T-ALL/LBL. To

clarify the role of CDK6 in cell cycle control and tumorigenesis, I have

generated mice with targeted mutations in Cdk6. These “knock-in” alleles

generate hyperactive or inactive kinase subunits that may better mimic

hyperactivation of CDK6 in tumor cells or model pharmaceutical inhibition

of CDK6, respectively. We have found that CDK6 is required for initiation

and maintenance of T-ALL leukemia and lymphomagenesis induced by

constitutively active Notch/Myr-AKT. Pharmacologic inhibition of CDK6

kinase induces CD25 expression, cell cycle arrest, and apoptosis inmouse

and human T-ALL. Ablation of Cd25 in a K43M background restores

Notch-induced T-leukemogenesis, with disease that is resistant to CDK6

inhibitors

in vivo

. Moreover, loss of Cd25 in a K43M background restore

the ability of LSCs to self-renew. These data support a model whereby

CDK6-mediated suppression of CD25 is required for initiation of T-ALL by

activated Notch1, and CD25 induction mediates the therapeutic response

to CDK6 inhibition in established T-ALL. These results both validate CDK6

as a molecular target for therapy

of this subset of T-ALL and

suggest that CD25 expression

could serve as a biomarker for

responsiveness of T-ALL to

CDK4/6 inhibitor therapy.

Figure. A working model of the role of

CDK6 in T-ALL. In response to stimuli,

increased expression of cyclin D1

and CDK6 leads to increased CDK6

activation, while Notch1 and AKT1

are also independently activated in

parallel. Notch1 further activates CDK6

via upregulation of CDK6 and/or by

Miaofen G Hu has completed her PhD from

Boston University School of Medicine and post-

doctoral studies fromHarvard University School

of Medicine. She has been Assistant Professor

at TUFTS medical Center since 2011. She has

published 24 papers in reputed journals. Her

most significant research accomplishments

thus far include creating a CDK6 mouse model,

discovering the role of CDK6 as a common me-

diator of Notch1 and AKT1 signaling pathways,

establishing the potential therapeutic role of

CDK6 in T cell malignance, revealing the func-

tion of CDK6 kinase activity in negatively reg-

ulating the conversion of fat-storing cells into

fat-burning cells.

mhu@tuftsmedicalcenter.org

BIOGRAPHY

increased cyclin D3 protein, while AKT1 activates CDK6

through the stabilization of cyclin D2. Once activated,

CDK6 can phosphorylate pRB, resulting in its inactivation.

CDK6, along with ERK and CDK1 can also phosphorylate

RUNX1 thereby promotingRUNX1proteolyticdegradation.

On a different molecular path, phosphorylation of FOXM1

by CDK6 stabilizes FOXM1, which in turn promotes

methylation of the GATA3 promoter, decreasing GATA3

expression and the subsequent recruitment to the

CD25 proximal promoter region. CD25 expression is

consequently reduced and T-ALL develops. Devoid of

CDK6 protein/kinase activity, pRB and RUNX1 remain

active, which suppress the tumorigenesis in CD25-

independent manner. Contrastingly, without CDK6, FOXM1

is in its inactive state, leading to CD25 upregulation.

Overall, T-ALL is suppressed by FOXM1 inactivity, and by

increasing RUNX1, pRB, GATA3, and CD25 expression or

activity.