• No results found

The interplay between microenvironmental signaling and novel targeted drugs in CLL - Chapter 3: Dual TORK/DNA-PK inhibition blocks critical signalling pathways in chronic lymphocytic leukemia

N/A
N/A
Protected

Academic year: 2021

Share "The interplay between microenvironmental signaling and novel targeted drugs in CLL - Chapter 3: Dual TORK/DNA-PK inhibition blocks critical signalling pathways in chronic lymphocytic leukemia"

Copied!
32
0
0

Bezig met laden.... (Bekijk nu de volledige tekst)

Hele tekst

(1)

UvA-DARE is a service provided by the library of the University of Amsterdam (https://dare.uva.nl)

UvA-DARE (Digital Academic Repository)

The interplay between microenvironmental signaling and novel targeted drugs in

CLL

Thijssen, R.

Publication date

2016

Document Version

Final published version

Link to publication

Citation for published version (APA):

Thijssen, R. (2016). The interplay between microenvironmental signaling and novel targeted

drugs in CLL.

General rights

It is not permitted to download or to forward/distribute the text or part of it without the consent of the author(s)

and/or copyright holder(s), other than for strictly personal, individual use, unless the work is under an open

content license (like Creative Commons).

Disclaimer/Complaints regulations

If you believe that digital publication of certain material infringes any of your rights or (privacy) interests, please

let the Library know, stating your reasons. In case of a legitimate complaint, the Library will make the material

inaccessible and/or remove it from the website. Please Ask the Library: https://uba.uva.nl/en/contact, or a letter

to: Library of the University of Amsterdam, Secretariat, Singel 425, 1012 WP Amsterdam, The Netherlands. You

will be contacted as soon as possible.

(2)

DUAL TORK/DNA-PK INHIBITION BLOCKS

CRITICAL SIGNALLING PATHWAYS

IN CHRONIC LYMPHOCYTIC LEUKEMIA

3

(3)

3

TORK/DNA-PK INHIBITION IN CLL

ABSTRACT

Inhibition of B cell receptor (BCR) signaling pathways in chronic lymphocytic leukemia (CLL) provides significant clinical benefit to patients, mainly by blocking adhesion of CLL cells in the lymph node (LN) microenvironment. The currently applied inhibitors ibrutinib and idelalisib have limited capacity however to induce cell death as monotherapy and are unlikely to eradicate the disease. Acquired resistance to therapy in CLL is often caused by mutations in the response network being targeted, both for DNA damage or BCR signaling pathways. Thus, drugs with dual targeting capacity could offer improved therapeutic value. Here, the potency of CC-115, a novel inhibitor of mammalian target of rapamycin kinase (TORK) and DNA-dependent protein kinase (DNA-PK), was evaluated in primary CLL cells in vitro and in CLL patients. Combined TORK and DNA-PK inhibition in vitro resulted in caspase-dependent cell killing irrespective of p53, ATM, NOTCH1 or SF3B1 status. Proliferation induced by CD40+IL-21 stimulation was completely blocked by CC-115, and CD40-mediated resistance to fludarabine and venetoclax could be reverted by CC-115. BCR mediated signaling was inhibited by CC-115, also in CLL samples obtained from patients with acquired resistance to idelalisib treatment. Clinical efficacy of CC-115 was demonstrated in eight patients with relapsed/refractory CLL/SLL harboring ATM deletions/ mutations; all but one patient had a decrease in lymphadenopathy, resulting in one IWCLL partial response (PR) and three PRs with lymphocytosis. The clinical trial was registered at www.ClinicalTrials.gov as NCT01353625.

In conclusion, these preclinical results, along with early promising clinical activity, suggest that CC-115 may be developed further for treatment of CLL.

Rachel Thijssen1,2, Johanna ter Burg1,2, Brett Garrick3, Gregor G.W. van Bochove1,2,

Jennifer R. Brown4, Stacey M. Fernandes4, María Solé Rodríguez5, Jean-Marie Michot6,

Michael Hallek7, Barbara Eichhorst7, Hans Christian Reinhardt7, Johanna Bendell8,

Ingrid A.M. Derks1, Roel J.W. van Kampen9, Kristen Hege3, Marie José Kersten2,10,

Torsten Trowe3, Ellen H. Filvaroff3, Eric Eldering1,10*, Arnon P. Kater2,10*

Departments of Experimental Immunology1 and Hematology2, Academic Medical Center, University

of Amsterdam, Amsterdam, The Netherlands.

3Celgene, San Francisco, CA, USA.

4Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA. 5Department of Hematology, Hospital Universitario Virgen del Rocío, Seville, Spain. 6Department of Hematology, Institut Gustave Roussy, Villejuif, France.

7Department of Hematology, University Hospital, Cologne, Germany. 8Sarah Cannon Research Institute/Tennessee Oncology, Nashville, TN, USA. 9Orbis Medisch Centrum, Sittard, The Netherlands.

10Lymphoma and Myeloma Center Amsterdam, LYMMCARE, The Netherlands.

* Shared senior authorship

(4)

3

TORK/DNA-PK INHIBITION IN CLL

INTRODUCTION

Chronic lymphocytic leukemia (CLL) cells highly depend on both B cell receptor (BCR)-mediated signaling as well as on stimuli received from the tumor microenvironment for their survival and proliferation1-6. The importance of the microenvironment is substantiated

by the recent success of novel drugs that target kinases involved in B cell receptor (BCR) signaling. Treatment with the Bruton tyrosine kinase (BTK) inhibitor ibrutinib or the phosphatidylinositol 3 kinase-

δ

(PI3K

δ

) inhibitor idelalisib abolishes chemotaxis towards stroma and BCR-controlled integrin-mediated cell adhesion. This results in rapid reduction of lymph node size and is followed by prolonged lymphocytosis7,8. Such

prolonged lymphocytosis during kinase-inhibitor treatment appears to pose no clinical disadvantage9,10. However, prolonged lymphocytosis could enhance the chance of

accumulating resistance-inducing mutations. Indeed, acquired resistance to ibrutinib was reported in patients due to mutations in BTK or in downstream kinases11,12. Drugs that

target both BCR-signaling, as well as critical survival pathways could provide an improved therapeutic strategy for CLL.

From this perspective, there is increased interest in compounds that target other kinases in the PI3K family13-15. The PI3K-related protein kinase (PIKK) family includes

mammalian target of rapamycin kinase (TORK), ataxia telangiectasia mutated (ATM), ataxia telangiectasia and Rad3 related (ATR) and DNA-dependent protein kinase (DNA-PK). TORK is the main downstream kinase of the PI3K/AKT pathway, and exists in two protein complexes, mTORC1 and mTORC216. mTORC1 (Raptor) is activated by AKT and leads to

phosphorylation of downstream effectors, which include 4EBP1 and S612, 17, while mTORC2

(Rictor) phosphorylates AKT at the Serine residue 473, leading to AKT activation16,18. In

healthy B cells, mTORC1 and mTORC2 are both critical for proliferation and differentiation through distinct mechanisms19-22. While inhibition of mTORC1 by rapamycin results

in suppression of B cell growth without induction of cell death19, deletion of mTORC2

affects cell viability23. Complete deletion of the TORK kinase (TORK) gene in mouse B

cells resulted in impaired germinal center formation24. Inhibition of mTORC1 by rapamycin

in CLL cells results in increased fludarabine sensitivity25 and inhibition of CpG-induced

proliferation of CLL cells26.

ATM, ATR and DNA-PK are critical regulators of the DNA damage repair (DDR) pathway. DNA-PK is required for the repair of DNA double-strand breaks (DSBs) through the process of non-homologous end joining (NHEJ)27. NHEJ is active throughout the cell

cycle, whereas homologous recombination mediated by ATM/ATR is active in late S phase and in G228-30. As peripheral blood CLL cells are in cell cycle arrest31, it is likely that DNA

repair in CLL cells predominantly depends on NHEJ.

In this study, the potency of a dual TORK and DNA-PK inhibitor (CC-115) was analysed in primary CLL samples of different prognostic subgroups with respect to induction of cytotoxicity, and blocking of CD40-mediated chemo-resistance and proliferation. Furthermore, clinical efficacy of CC-115 was tested in one small lymphocytic lymphoma (SLL) patient and seven CLL patients.

(5)

3

TORK/DNA-PK INHIBITION IN CLL

MATERIALS AND METHODS

Patient material

After written informed consent, patient material was obtained during diagnostic or follow-up procedures at the Department of Hematology of the Academic Medical Center Amsterdam and affiliated hospitals. This study was approved by the AMC Ethical Review Board and the ethical board of the Dana Farber Medical Center, Boston, MA, USA and written informed consent was obtained in accordance with the Declaration of Helsinki. Blood mononuclear cells of patients with CLL (Supplemental table 1), obtained after Ficoll density gradient centrifugation (Pharmacia Biotech, Roosendaal, The Netherlands) were frozen and stored as previously described32. Expression of CD5 and CD19 (both Beckton

Dickinson (BD) Biosciences, San Jose, CA, USA) on leukemic cells was assessed by flow cytometry (FACScanto; BD Biosciences). CLL samples included in this study contained 81-99% CD5+/CD19+ cells.

Peripheral blood mononuclear cells (PBMCs) were obtained from healthy blood donors derived buffy coats, aged between 18 and 64 years, from Sanquin Blood Supply, Amsterdam, Netherlands. PBMCs were isolated and frozen and stored in liquid nitrogen until use.

FISH and Gene mutational analyses

Deletions at the 11q22-q23 (ATM), 17p13 (TP53) and 13q14 loci and trisomy of chromosome 12 were detected by FISH by using locus-specific probes (Abott Vysis Inc). DNA was extracted by using the QiAamp DNA Blood Mini kit (Invitrogen) according to the manufacturer’s instructions. TP53 mutational analysis was either performed by a 454-based next generation sequencing (NGS) approach (Junior 454 platform, Roche, Penzberg, Germany) or using Sanger sequencing (exons 4-10)33. Primer sequences are

provided in Supplemental table 3. Mutation analysis of ATM (exons 1-62) was performed by Sanger sequencing as described previously34,35.

Reagents

CC-115, CC-214-1 [CC-214] and CC-292 were obtained from Celgene (Summit, NJ, USA). The PI3K

δ

inhibitor CAL-101/idelalisib and the DNA-PK inhibitor NU7441 were from Selleckchem (Housten, TX, USA). Fludarabine (F-Ara-A), chlorambucil, bendamustine and N-acetylcysteine (NAC) were purchased from Sigma Chemical Co. (St. Louis, MO, USA). The pan-caspase inhibitor Q-VD was purchased from R&D systems (Minneapolis, USA). ABT-199 was purchased from Active Biochem (Bonn, Germany).

Cell culture and detection of apoptosis

Freshly isolated CLL cells were treated with different concentrations of CC-115 for 30 minutes. Subsequently the cells were exposed to 5 Gy

γ

-radiation or treated with 10µg/ ml bleomycin (EMD Millipore, Billerica, MA, USA) and incubated for 30 minutes and cell lysates were made.

For the apoptosis assays, PBMCs from healthy donors or CLL patients were thawed and incubated with different concentrations of drugs for the indicated 48 hours. Samples

(6)

3

TORK/DNA-PK INHIBITION IN CLL

culture. Where indicated, CLL cells were co-cultured in the presence/absence of 20μM of the pan-caspase inhibitor Q-VD or 5mM N-acetyl-L-cysteine (NAC). Viability was measured by DiOC6/PI staining as previously described32. Specific apoptosis was defined as

( ([% cell death in treated cells] – [% cell death in medium control]) / [% viable cells medium control]) x 100.

To mimic the anti-apoptotic properties of microenvironmental stimulated CLL cells, CLL cells were stimulated by coculture with NIH3T3 fibroblasts stably transfected with human CD40L (3T40L) or negative control plasmid (3T3) as described32 and co-cultured in

the presence/absence of drugs at 1μM or the indicated concentrations.

Western blot analysis

Cells were lysed in RIPA sample buffer and sonificated32. Samples were separated by

3-8% TA gel (Thermo Fisher Scientific, Grand Island, NY, USA), 4-12% bris-tris protein gel (Thermo Fisher Scientific) or 13% SDS-PAGE gel electrophoresis. Membranes were probed with the following antibodies: anti-DNA-PK, pHSP90

α

, pS6, p4EBP1, pEIF4E, Mcl-1 (Cell Signaling, Boston, MA, USA), pDNA-PK, ATM, HSP90

α

,

γ

H2AX (Abcam, Cambridge, UK), Bcl-XL (BD Biosciences), Bim (Stressmarq, Victoria, Canada), pATM, vinculin (Sigma), actin (Santa Cruz Biotechnology, Dallas, TX, USA), and anti-A1/Bfl-1 was a kind gift of Prof. Dr. J. Borst (The Netherlands Cancer Institute, Amsterdam, The Netherlands). Odyssey Imager (Li-Cor Biosciences) was used as a detection method according to the manufacturer’s protocol.

γ

H2AX FACS staining

CLL cells were thawed and incubated for 30 minutes either with or without 1µM CC-115 or NU7441. Subsequently the cells were exposed to 5 Gy

γ

-radiation and incubated for 2 hours. CLL cells were fixed and incubated with CD5-PE (eBioscience), CD19-APC (BD biosiences) and intracellular staining was performed for isotype-AF488 or

γ

H2AX-AF488 (Cell Signaling) for 30 min. Expression of

γ

H2AX was determined within the CD5+CD19+

CLL cells using the FACSCalibur flow cytometer and CellQuest software was used for data acquisition. Data were analyzed with FlowJo software.

Proliferation assay

CLL cells (1.0x107/mL) were labelled with 0,5μM CFSE (Molecular Probes, Life Technologies, Bleiswijk, The Netherlands) as described before36. Cells were cultured on 3T40L cells,

in absence or presence of recombinant human IL-21 (25ng/ml, Gibco, Invitrogen, Life Technologies), with or without 1 μM of drugs. After 4 days, proliferation was assessed in a FACS Canto (BD Biosciences) and analyzed with FlowJo software (TreeStar, Ashland, OR).

Activation of healthy PBMC

PBMCs from healthy donors were stimulated with

α

CD3 (1xE, ascites) and

α

CD28 (15E8; 5µg/ml) for 3 days. PBMCs were resuspended in PBS, containing 0.5%(w/v) BSA and 0.01% sodium azide. PBMCs were incubated with saturating concentrations of

(7)

CD19-PerCP-3

TORK/DNA-PK INHIBITION IN CLL

Cy5.5, CD20-APC-H7, IgD-PE, CD27-APC, CD38-PE-Cy7, CD3-AF700, CD4-PE-Cy7, CD8- PerCP-Cy5.5, CD38-PE, CD25-APC (BD-biosciences). Flow cytometry measurements were performed on a FACSCanto using FACSDiva Software (BD Biosciences).

Clinical Study

Eight patients with refractory/relapsed CLL/SLL (small lymphocytic lymphoma) were enrolled into the dose expansion part of a larger phase 1A/1B multicenter study of CC-115 entitled a Phase 1A/1B, multicenter open label, dose-finding study to assess the safety, tolerability, pharmacokinetics and preliminary efficacy of the dual DNA-PK and TOR kinase inhibitor CC-115, administered orally to subjects with advanced solid tumors and hematologic malignancies. The study was registered at www.ClinicalTrials.gov as NCT01353625. SLL/ CLL patients with 11q22 (ATM) deletions were eligible. The second, non-deleted ATM allele was sequenced to determine heterozygosity at this locus. The study was approved by relevant Ethical Review Boards and regulatory authorities. Written informed consent was given and the study was performed according to Good Clinical Practice. The clinical trial was sponsored by Celgene. Patients had received at least one prior line of systemic therapy and symptomatic progression or another indication for treatment was required. Patients received the earlier established recommended dose of 10mg b.i.d of CC-115 continuously in 28 day cycles, and no other concomitant anti-leukaemia treatment. Steroids were only allowed in physiological doses or for treatment of toxicities. The efficacy endpoint was response rate assessed by CT scan and laboratory parameters at 8, 16 and 24 weeks and then every 12 weeks thereafter using the International Workshop on Chronic Lymphocytic Leukemia (IWCLL) 2008 guidelines. Patients meeting the IWCLL criteria for response, but with persistent lymphocytosis were categorised as relevant response with lymphocytosis. Three patients are ongoing having received 13, 15 and 23 cycles of therapy.

Statistics

The one-way ANOVA was used to analyze differences between groups. * p<0.05; ** p<0.01; *** p<0.001.

RESULTS

CC-115 is a dual DNA-PK and TORK inhibitor

Upon induction of DNA double stranded breaks, DNA-PK is recruited to the site of DNA damage and activated, which leads to phosphorylation of downstream targets H2AX and the heat shock protein 90

α

(HSP90

α

)37-40. Inhibition of DNA-PK and the immediate

DNA damage response by CC-115 was measured upon irradiation or treatment with the chemotherapeutic agent bleomycin, a potent inducer of DNA strand breaks, in primary CLL cells. Phosphorylation of DNA-PK, ATM, HSP90

α

and H2AX was reduced to a variable degree (Figure 1A-B). CC-115 showed dose-dependent repression of Ser-2056 phosphorylation on DNA-PK at clinically achievable doses (0.1 to 0.35µM), and reduced baseline and bleomycin-induced Ser-1981 phosphorylation on ATM, Thr-5/7 phosphorylation on HSP90

α

and

γ

H2AX. These effects were comparable in wildtype, as

(8)

3

TORK/DNA-PK INHIBITION IN CLL

Figure 1. CC-115 inhibits the DNA damage repair pathway and TORK in CLL cells. A-B. Freshly isolated CLL cells were incubated with the indicated concentration CC-115 for 30 minutes and irradiated (5Gy) and lysates were made after 30 minutes. Protein lysates were probed for pDNA-PK (S2056), DNA-PK, pATM (S1981), ATM, pHSP90

α

(T5/7), HSP90

α

, yH2AX and vinculin and cofilin for loading control. A. Blots from two representative CLL samples are shown of five analyzed (Supplemental table 1 CLL pt#1, 2A, 3A, 4A, 5). B. Densitometric analysis of pDNA-PK, pATM, pHSP90

α

and yH2AX are shown. ***p<0.001 (one-way ANOVA). C. CLL cells of ATM mutated patients (n=4, pt#12-15) were treated with or without 1 µM NU7441 or CC-115 followed by irradiation (5 Gy) and yH2AX expression was measured at 2 hours using flowcytometry. *p<0.05, **p<0.01 (one-way ANOVA). D. CLL cells were incubated with the indicated concentration CC-115 for 30 minutes and lysates were made after 30 minutes. Protein lysates were probed for pS6 (S240/244) and S6. Blot from pt#85 and densitometric analysis are shown (n=3, pt#85, 87-88). Bars represent the mean ± SEM, ***p<0.001 (one-way ANOVA). E. CLL cells were cultured in the presence or absence of 1 µM CC-115, CC-214, CC-292 or idelalisib for 1 hour. Protein lysates were probed for pS6 (S240/244) and actin for loading control. Blots from three representative CLL samples are shown (Supplemental table 1 CLL pt#19-21). F. CLL cells (pt#20) pretreated with 1 µM idelalisib, CC-115, CC-214 and CC-292 were stimulated with

α

IgM for 20 min. Protein lysates were probed for pAKT(S473), pS6 (S240/244), p4EBP1 (T37/46), pERK (T202/204) and actin as loading control.

A C con t rol con t rol C C-1 15 N U74 41 0 1 0 0 2 0 0 3 0 0 4 0 0 5 G y ** * M FI γ-H 2 A X 0 0 0.35 1 3.5 0 0 0.35 1 3.5 CC-115 [µM] 5 Gy 5 Gy pDNA-PK (S2056) DNA-PK pATM (S1981) ATM Vinculin pHSP90α (T5/7) HSP90α γH2AX (S139) Cofilin CLL pt#1 CLL pt#2A 0 0 0.35 1 3.5 0 50 100 CC-115 [µM] 5 Gy ****** pD NA-PK/ D NA-PK 0 0 0.35 1 3.5 0 50 100 CC-115 [µM] 5 Gy *** pA TM /A TM 0 0 0.35 1 3.5 0 50 100 CC-115 [µM] 5 Gy ****** *** pH SP 90 α/ H SP9 0α 0 0 0.35 1 3.5 0 50 100 CC-115 [µM] 5 Gy ****** γH 2AX/ co fil in B CLL #19 CLL #21 Actin pS6 Actin pS6 Actin pS6 Control CC-115 CC-214 CC-292 Ide D p -S6 /S6 0 0 .3 5 1 3 .5 0 5 0 1 0 0 *** ****** C C -1 1 5 [µ M ] E CLL #20 pS6 S6 Actin p4EBP1 (T37/46) pS6 (S240/244) αIgM idela CC-115 CC-214 CC-292 pAKT (S473) pERK (T202/Y204) F

(9)

3

TORK/DNA-PK INHIBITION IN CLL

well as ATM/11q mutated CLL cells (Supplemental Figure 1A-B). Dependence of ATM/11q mutated CLL cells on DNA-PK activity with respect to DNA repair was demonstrated by measuring

γ

H2AX by a flow cytometry-based assay as described before33. Treatment

with the DNA-PK inhibitor NU7441 (1µM) or CC-115 (1µM) inhibited irradiation-induced

γ

H2AX levels in ATM/11q mutated CLL cells (Figure 1C).

Next, TORK inhibitory activity of CC-115 was determined. Constitutive phosphorylation of S6, a marker for mTORC1 activity, was detected in all CLL samples and this was inhibited by both CC-115 at low doses (Figure 1D) and the specific TORK inhibitor CC-214 at 1µM (Figure 1E). Inhibitors of kinases more upstream in the BCR pathway, the BTK inhibitor CC-292 or the PI3K

δ

inhibitor idelalisib, did not decrease levels of pS6 (S240/244) (Figure 1E). In BCR-stimulated CLL cells, phosphorylation of the mTORC2 target, AKT (S473), and the mTORC1 targets, S6 and 4EBP1, were blocked completely by CC-115 and CC-214, while phosphorylation of ERK was not affected by the TORK inhibitors (Figure 1F). These data demonstrate robust phosphorylation of DNA-PK both in ATM-mutant and wildtype CLL cells upon DNA damage and constitutive activation of TORK. Both TORK and DNAP-PK were inhibited by CC-115 in these cultured primary CLL cells.

CC-115 induces caspase-dependent cell death in resting CLL cells

To determine whether combined inhibition of TORK and DNA-PK induces cell death, we compared CC-115 to more specific inhibitors (Figure 2A). In order to correct for variability in viability between thawed primary CLL samples, specific apoptosis was calculated (Figure 2B). CC-214 (TORKi), CC-292 (BTKi), idelalisib (PI3K

δ

i) and NU7441 (DNAPKi) induced modest cell death (IC50 > 10µM and maximum induction of apoptosis at 10µM of 30-40%) (Figure 2B), while CC-115 induced cell death with an IC50 of 0.51µM (Figure 2B). Cell death was due to on-target inhibition of TORK and DNA-PK, since the combination of the TORK inhibitor CC-214 and the DNA-PK inhibitor NU7441 resulted in cell death comparable to CC-115 (Figure 2C). CC-115 induced cell death in clinically relevant prognostic CLL subgroups. All subgroups, including those with high risk features were equally sensitive to ≥ 1µM CC-115. CLL cells harbouring a TP53- and SF3B1 mutations appeared to be less sensitive to CC-115 at lower concentrations, but this was not statistically significant in this smaller patient cohort (TP53, n=6; SF3B1 n=4) (Figure 2D).

The pan-caspase inhibitor Q-VD completely blocked CC-115 induced cytotoxicity, demonstrating that CC-115-induced cell death is caspase-dependent (Figure 2E). A variety of chemotherapeutic drugs induce apoptosis in CLL cells by the generation of reactive oxygen species (ROS), as we have shown for platinum-based compounds41. Co-treatment

with NAC, a ROS inhibitor, was able to rescue CCCP-induced cell death (data not shown) but not CC-115-induced cell death (Figure 2E).

Next, we assessed whether inhibition of the DNA repair pathway by CC-115 enhanced the sensitivity to the DNA damage-inducing agents chlorambucil and bendamustine. Combination of low dose of the DNA-PK/TORK inhibitor with chlorambucil (6.25 µM) or bendamustine (6.25 µM) had a significant additive effect on the induced apoptosis (Figure 2F). Thus, combined inhibition of DNA-PK and TORK results in caspase-dependent cell death, irrespective of the p53 status and enhances sensitivity to chemotherapy.

(10)

3

TORK/DNA-PK INHIBITION IN CLL

A

B

Figure 2

CC-11 5 CC-21 4 NU74 41 CC-21 4 + N U744 1 0 20 40 60 80 100 ****** ns Sp eci fic ap op to si s ( % ) 0 0.01 0.1 1 10 0 20 40 60 80 100 Medium 5 mM NAC 20 µM QVD CC-115 [µM] Sp eci fic ap op to si s ( % )

C

E

Chlor ambu cil CC-11 5 Chlor ambu cil + CC-11 5 Bend amus tin CC-11 5 Bend amus tin + CC-11 5 0 20 40 60 80 100 ** * * Sp eci fic ap op to si s ( % )

D

0 0.01 0.1 1 10 0 20 40 60 80 100 wildtype ATM mutated p53 mutated SF3B1 mutated NOTCH1 mutated CC-115 [µM] Spec ifi c apopt os is (% ) 0 0.001 0.01 0.1 1 10 0 20 40 60 80 100 CC-115 CC-214 CC-292 Idelalisib NU7441 ** ** *** Drug concentration [µM] Sp eci fic ap op to si s ( % ) 0 0.001 0.01 0.1 1 10 0 20 40 60 80 100 CC-115 CC-214 CC-292 Idelalisib NU7441 Drug concentration [µM] Via bilit y ( % )

F

Figure 2. CC-115 induces apoptosis in CLL cells from distinct prognostic groups. A-B. CLL cells were incubated with 0.001-10 μM CC-115, CC-214, CC-292, idelalisib or NU7441 for 48 hours. Viability was assessed by DiOC6/PI staining (A) and specific apoptosis was calculated (material and methods) (B). Results are shown as mean ± SEM. (n=23, pt#6, 7, 8A, 21-31, 32A,B, 33A,B, 34A, 35A, 36-38). C. CLL cells were incubated with 1 µM CC-115, CC-214, NU7441 or CC-214 + NU7441 for 48 hours. Results are shown as mean ± SEM (n=11 pt#8B, 23, 39-46). D. CLL cells of patients of distinct prognostic CLL groups; wildtype (n=23, pt#6, 7, 8A, 21-31, 32A,B, 33A,B, 34A, 35A, 36-38), ATM mutated (n=10, pt#2C, 47-55), p53 mutated (n=6, pt#56-61), SF3B1 mutated (n=4, pt#62-65) and NOTCH1 mutated (n=3, pt#66-68) were incubated withCC-115 for 48 hours. Results are shown as mean ± SEM. E. CLL cells were cultured with 20 µM Q-VD or 5 mM NAC and with increasing concentrations of CC-115 for 48 hours. Results are shown as mean ± SEM (n=3, pt#21, 33B, 31). F. CLL cells were cultured with 6.25 µM chlorambucil, 6.25 µM bendamustine, 0.1 µM CC-115 or the combination of 6.25 µM chlorambucil/bendamustine and 0.1 µM CC-115 for 48 hours. Results are shown as mean ± SEM (n=5, pt#2C, 19B, 29, 37, 69). *p<0.05, **p<0.01, ***p<0.001 (one-way ANOVA).

CC-115 reverts CD40-mediated resistance to chemotherapy or venetoclax

In the lymph node microenvironment, CLL cells receive prosurvival signals from surrounding cells 1,2,6,43. This can largely be mimicked by in vitro CD40 stimulation6,43, which resulted

in increased mTORC1 markers (pS6, peIF4E and p4EBP1), which was inhibited by CC-115 (Figure 3A and Supplemental Figure 2). CC-115 and the TORK inhibitor CC-214 inhibited CD40-mediated activation of CLL cells, as measured by induction of immune accessory molecules, such as death receptor (CD95) and adhesion receptors (CD54, CD58, CD44)43,44

(Supplemental Figure 3). In contrast, BTK- and PI3K

δ

inhibitors modestly inhibited CD40-induced upregulation of CD58 and did not affect upregulation of other immune accessory molecules (Supplemental Figure 3). CD40 stimulation inhibits spontaneous cell death, as

(11)

3

TORK/DNA-PK INHIBITION IN CLL

previously reported2, 45. Of the four inhibitors tested, only CC-115 reverted CD40-induced

survival (Figure 3B). This was also observed by combining the TORK and DNA-PK inhibitor (Supplemental Figure 4). CD40 stimulation induces resistance to cytotoxic agents, including fludarabine and the specific Bcl-2 inhibitor ABT-199 (venetoclax)43,46. Inhibitors

of BTK and PI3K

δ

had no impact on CD40-induced chemoresistance. The TORK inhibitor CC-214 partly and the dual TORK/DNA-PK inhibitor CC-115 completely abolished CD40-induced fludarabine resistance (Figure 3C). Moreover, CC-115 also reduced the extensive ABT-199 resistance conferred by CD40 stimulation (Figure 3D).

CLL cells originating from lymph nodes show an altered expression of anti- and pro-apoptotic proteins including increased expression of Bcl-XL , Bfl-1 and Mcl-1 and downregulation of Bim32,46,47. CD40-mediated decreased expression of Bim was

abolished by co-culture with CC-115 (Figure 3E). In accordance, CC-115 treatment significantly reduced induction of expression Mcl-1, Bfl-1 and Bcl-XL upon CD40 stimulation (Figure 3F-I).

Thus, CD40-mediated expression of immune accessory molecules and drug resistance could be reverted by CC-115, which correlated with suppressed induction of the anti-apoptotic proteins Bcl-XL, Bfl-1 and Mcl-1 and repression of the CD40-mediated reduction of Bim expression.

CC-115 blocks downstream signaling pathways and induces cell death in

idelalisib-resistant CLL cells

Targeting PI3K

δ

downstream of the BCR by idelalisib showed significant clinical activity in CLL patients42, however a proportion of CLL patients develop resistance to idelalisib. We

assessed CC-115 activity in vitro in CLL cells from patients who had become resistant to idelalisib treatment. Following BCR triggering, phosphorylation of ERK was inhibited by idelalisib in CLL samples obtained from responding patients, but this no longer occurred following acquired idelalisib resistance (Figure 4A). CC-115 inhibited both BCR mediated phosphorylation of S6 as well as CD40 mediated S6 and AKT (S473) phosphorylation (Figure 4A-B). Both idelalisib responsive and resistant samples were sensitive to CC-115 induced cell death (Figure 4C.)

CC-115 blocks proliferation of CLL cells

Activated T cells and follicular helper T cells present in the CLL LN express membrane-bound CD40L and can secrete cytokines, such as IL-21. We have shown previously that in

vitro, combination of CD40+IL-21 signals induces proliferation36. This BCR-independent

proliferation was inhibited partially by CC-292, idelalisib or NU7441 and fully by CC-115 or CC-214 (figures 5A-B).

Effects of CC-115 on B- and T- cells

We next studied the impact of the different kinase inhibitors on cell death and function of healthy B- and T cells. In healthy B cells, CC-115 induced cell death with an IC50 of 0.93µM (Figure 6A), while the TORK, BTK, PI3K

δ

and DNA-PK inhibitors were not cytotoxic at doses up to 10µM. None of the kinase inhibitors induced cell death in T cells from healthy

(12)

3

TORK/DNA-PK INHIBITION IN CLL

Figure 3

0 1.56 6.25 25 0 20 40 60 80 100 3T3 CD40L CD40L+CC-115 CD40L+CC-214 CD40L+CC-292 CD40L+Idelalisib Fludarabine [µM] Spec ifi c apopt os is (% )

C

E

Bim Actin CD40L 3T3 CC-115 CLL #75 CLL #74 CD40L 3T3 CC-115 Bcl-XL Bfl-1 Actin 3T3 CD40L CC-115 CC-214 CC-292 Idelalisib Mcl-1 3T3 Mediu m CC-11 5 CC-21 4 CC-29 2 Idelal isib 0.0 0.5 1.0 1.5 CD40L * Mc l-1 /β -a ct in 3T3 Mediu m CC-11 5 CC-21 4 CC-29 2 Idelal isib 0.0 0.5 1.0 1.5 CD40L * Bc l-XL /β -a ct in 3T3 Mediu m CC-11 5 CC-21 4 CC-29 2 Idelal isib 0.0 0.5 1.0 1.5 CD40L **** B fl-1 /β -a ct in

F

G

H

I

0 0.001 0.01 0.1 1 10 0 20 40 60 80 100 Control CD40L CD40L + CC115 ABT-199 [µM] Sp eci fic ap op to si s ( % )

D

3T3 mediu m CC-11 5 CC-21 4 CC-29 2 Idelal isib 0 20 40 60 CD40L ** Sp eci fic ap op to si s ( % ) Actin peIF4E (S209) CLL pt#69 p4EBP1 (T37/46) pS6 (S240/244) CLL pt#37 3T3 CD40L CC-115 3T3 CD40L CC-115

B

A

Figure 3. CC-115 reverts CD40-induced chemoresistance. A-I. CLL cells were cultured on 3T3 fibroblasts or CD40L-expressing fibroblasts (CD40L) in the presence or absence of 1 µM of CC-115, CC-214, CC-292 or idelalisib for 3 days. A. Protein lysates were probed for pS6 (S240/244), peIF4E (S209), p4EBP1 (T37/46) and actin as loading control. Blots from two representative CLL samples are shown (pt#70, 37). B. Survival was analyzed by DiOC6 staining. Results are shown as mean ± SEM. *p<0.05, **p<0.01 (one-way ANOVA) (n=10 pt#6, 25, 30, 33B, 34A, 43B, 72-74, 75). C. After 3 days, fludarabine sensitivity assay was performed. Apoptosis was assessed by DiOC6/PI staining and specific apoptosis are shown as mean ± SEM (n=9, pt# 6, 25, 30, 33B, 34A, 43B, 72-74). D. After 3 days, ABT-199 sensitivity assay was performed. Results are shown as mean ± SEM (n=3, pt#33B, 35B, 45). E. Protein lysates were probed for Bim and actin as loading control. Blots of two representative CLL samples are shown of 7 analyzed. F. Protein lysates were probed for Mcl-1, Bcl-XL, Bfl-1 and actin

as loading control. Blot from one representative CLL sample is shown of 9 analyzed. G-I. Densitometric analysis of Mcl-1/actin (G), Bcl-XL/actin (H), Bfl-1/actin (I) level is shown. Bars represent the mean ±

(13)

3

TORK/DNA-PK INHIBITION IN CLL

Figure 4

A

- Idelalisib-resistant pERK (T202/Y204) αIgM idela CC-115 αIgM idela CC-115 pS6 (S240/244) Actin

B

C

- Idelalisib-resistant pERK (T202/Y204) CD40L idela CC-115 CD40L idela CC-115 pS6 (S240/244) Actin pAKT (S473) 0 0 .1 1 1 0 0 2 0 4 0 6 0 8 0 1 0 0 S pe c if ic a popt os is ( % ) P t 1 -P t 1 id e la lis ib -re s is ta n t P t 2 -P t 2 id e la lis ib -re s is ta n t C C -1 1 5 [µ M ]

Figure 4. A. Idelalisib-resistant CLL cells are sensitive to CC-115 treatment. A. CLL cells obtained from a patient clinically sensitive to idelalisib (-) and CLL cells obtained after the patient became resistant to idelalisib (idelalisib-resistant) were pretreated with 1 µM CC-115 or 1 µM idelalisib and stimulated with

α

IgM for 20 min. Protein lysates were probed for pS6 (S240/244), pERK (T202/204) and actin as loading control. B. CLL cells obtained from a patient clinically sensitive to idelalisib (-) and CLL cells obtained after the patient became resistant to idelalisib (idelalisib-resistant) were cultured on CD40L-expressing fibroblast in the presence or absence of 1 µM CC-115 or idelalisib for 24 hours. Protein lysates were probed for pAKT (S473), pERK (T202/204), pS6 (S240/244) and actin as loading control. C. CLL cells from 2 patients that became resistant to idelalisib (idelalisib-resistant) and samples that response to idelalisib treatment (-) were incubated with 0. 1-10 μM CC-115 for 48 hours. Viability was assessed by DiOC6/PI staining and specific apoptosis was calculated.

donors or T cells from CLL patients (Figure 6B, Supplemental Figure 4A). Furthermore, none of the kinase inhibitors significantly altered CD3/CD28-induced upregulation of the activation markers CD25 and CD38 (Supplemental Figure 4B-C). CC-115 and NU7441 completely blocked the proliferation of CD4+ and CD8+ T cells (Figure 6C). Inhibitors of

(14)

3

TORK/DNA-PK INHIBITION IN CLL

Early clinical effect of CC-115 treatment

Seven CLL patients and one SLL patient were enrolled in a larger Phase I clinical study, including 110 additional patients with solid tumors. Median age was 56 years (Supplemental Table 2). Median number of prior therapies was two (range, 1- 8) with all patients receiving prior chemotherapy with alkylating agents and/or fludarabine, anti-CD20 (rituximab) or anti-CD52 (alemtuzumab) therapy. All patients had a del(11q) clone and thus were devoid of at least one copy of ATM. Sequencing the second allele showed that three patients had mutations, which may be deleterious to ATM function, consistent with the possibility of bi-allelic ATM loss (Supplemental Table 2). Three patients are still ongoing and the median number of cycles was 13.5 (range 1-23). All but one patient had a decrease in lymphadenopathy, with four subjects having decreases of ≥ 50% (Figure 7A). One of these patients, who received 21 days of therapy, had a > 50% decrease in lymphadenopathy but had Richter’s syndrome diagnosed simultaneously. Three patients experienced a ≥ 50% decrease in lymphocytes (Figure 7B). Overall, there was one partial response according to iwCLL criteria, and three partial responses with lymphocytosis.

Analysis of one full set of pharmacodynamics blood samples from 1 of 7 patients treated with CC-115 showed > 50% inhibition of DNA-PK in circulating CLL cells. (Figure 7C-D).

Figure 5

A

B

CD40 L Mediu m CC-11 5 CC-21 4 CC-29 2 Idelal isib NU74 41 0.0 0.2 0.4 0.6 0.8 ****** ** CD40L + IL-21 D ivi si on i ndex CLL pt#19B CLL pt#35B CD40L CD40L + IL-21 CD40L + IL-21 + CC-115

Figure 5. CC-115 and mTOR inhibitors block proliferation of CLL cells. A. CFSE labelled CLL cells were cultured on fibroblast expressing CD40L with (black line) or without (grey) IL-21 and co-treated with 1 µM CC-115 (thick black line). After 4 days, CFSE was measured by FACS. Results are shown as representative histograms for 2 patients. B. Division index was calculated with FlowJo program. Results are shown as mean ± SEM (n=11, pt# 2C, 19B, 20, 35B, 71, 78-82). *p<0.05, **p<0.01, ***p<0.001 (one-way ANOVA).

(15)

3

TORK/DNA-PK INHIBITION IN CLL

Figure 6

T cells 0 0.01 0.1 1 10 0 20 40 60 80 100 CC-115 CC-214 CC-292 Idelalisib NU7441 Drug concentration [µM] Sp eci fic ap op to si s ( % ) B cells 0 0.01 0.1 1 10 0 20 40 60 80 100 CC-115 CC-214 CC-292 Idelalsib NU7441 Drug concentration [µM] Sp eci fic ap op to si s ( % )

A

Contr ol CC-11 5 CC-21 4 CC-29 2 Idelal isib NU74 41 Contr ol CC-11 5 CC-21 4 CC-29 2 Idelal isib NU74 41 0.0 0.5 1.0 1.5 CD8+ CD4+ * *** ** *** *** ** *** D iv is io n in de x

B

C

Figure 6. CC-115 inhibits proliferation of healthy T cells. A-B. PBMCs from healthy donors were incubated with 0.001-10 μM CC-115, CC-214, CC-292, idelalisib or NU7441 for 48 hours. Specific apoptosis was analyzed in CD19+ B cells (A) or in CD3+ T cells (B). Results are shown as mean ± SEM, n=3, ns (one-way ANOVA). C. PBMCs from healthy donors were stimulated with

α

CD3/

α

CD28 in the presence or absence of 1 μM CC-115, CC-214, CC-292, idelalisib or NU7441 for 72 hours (n=3). CFSE was measured by FACS in CD8+ T or CD4+ T cells. Bars represent the mean ± SEM, ns (one-way ANOVA).

DISCUSSION

DNA damaging chemotherapeutic agents cause the formation of toxic DNA double-strand breaks (DSBs) leading to cell cycle arrest and cell death. Cells have multiple mechanisms for repairing DSBs, which include homologous recombination (HR) and NHEJ48. HR, an

(16)

3

TORK/DNA-PK INHIBITION IN CLL

Figure 7

A

B

C

D

Baseline Day 15 treatment

CLL #2011007 - + - + - + - + [Bleo 10 µg/ml] Control 1.5 hr Control 1.5 hr CC-115 CC-115 pDNA-PK (S2056) DNA-PK p DNA-P K/ DNA-P K - + - + - + - + 0 2 0 4 0 6 0 8 0 1 0 0 B le o C o n tro l C C -1 1 5 C o n tro l C C -1 1 5 b a se lin e d a y 1 5 -1 2 0 -8 0 -4 0 0 4 0 8 0 1 2 0 201 100 5 001 103 4 401 100 2 401 100 1 201 100 7 301 101 9 201 100 4 % b est ch an g e fo r S P D f or t a rge t le s ions 2 01 10 03* C ha n g e f ro m ba s e li ne i n LC ( % ) 0 1 0 0 2 0 0 3 0 0 4 0 0 5 0 0 6 0 0 7 0 0 -2 0 0 0 2 0 0 4 0 0 6 0 0 8 0 0 4 0 1 1 0 0 1 2 0 1 1 0 0 4 2 0 1 1 0 0 5 4 0 1 1 0 0 2 3 0 1 1 0 1 9 4 0 1 1 0 0 3 2 0 1 1 0 0 7 0 0 1 1 0 3 4 d a y s

Figure 7. CC-115 decreases lymphadenopathy in CLL patients. A. Best percentage change in sum of product of diameters (SPD) for target lesions for patients with single ATM deletion (black) and those with likely or potentially deleterious mutation in remaining ATM allele (grey). *Patient had Richter’s diagnosed at the same time B. Changes in lymphocyte counts of patients with single ATM deletion (black) and those with likely or potentially deleterious mutation in remaining ATM allele (grey) (Supplemental Table 2) during CC-115 treatment over time. C. Western Blot analysis of phospho- and total DNA-PK in PBMCs from CC-115-ST-001 patient 2011007. PBMCs in plasma were collected at Screening, Cycle 1 Day 1/baseline (pre and 1.5 hr post CC-115 dose), and Cycle 1 Day 15 (pre and 1.5 hr post CC-115 dose). PBMCs in plasma were incubated with or without 10µg/ml bleomycin for 60 minutes . Total and phospho-DNA-PK were detected by Western Blot analysis of protein lysates. D. Quantification of the western blots in C.

as such, functions primarily in late S-phase and G2. NHEJ, although error prone, is able to function at all stages of the cell cycle and is thought to be the main pathway for DSB repair28. As such, it has been postulated that targeting the molecular machinery driving

the DNA damage response (DDR), particularly NHEJ and DSB repair, with small molecule inhibitors, will effectively enhance the efficacy of current cancer treatments that generate DNA damage49. Indeed, DNA-PK inhibition using small molecule inhibitors showed

promising preclinical results in both solid malignancies as well as in lymphoma and CLL50-52.

In fact, a previous study showed that DNA-PK activity is highly increased in resistant CLL cells53 and high concentrations of the DNA-PK inhibitor NU7026 or NU7441 could restore

irradiation-induced or chlorambucil-induced apoptosis sensitivity52-55. Clinical development

of DNA-PK inhibitors has been hampered so far due to poor pharmacokinetics49.

CLL cells harboring a 17p or 11q deletion have been reported to be protected from chemotherapy by DNA-PK overexpression54,56-58, and inhibition of DNA-PK restores

(17)

3

TORK/DNA-PK INHIBITION IN CLL

sensitivity to chemotherapeutic drugs in these CLL cells52-54. Inhibitors of DNA-PK might

therefore be of clinical interest in CLL, especially in patients with high risk disease. Indeed, our findings confirm that treatment with a DNA-PK inhibitor, CC-115 or NU7441, inhibited the DDR pathway in ATM mutated CLL cells, as demonstrated by the inhibition of irradiation-induced

γ

H2AX. We found high expression of pDNA-PK and pHSP90

α

in irradiated CLL cells, which was blocked by clinically achievable doses of CC-115. Pharmacodynamic analysis of blood samples after CC-115 treatment showed inhibition of pDNA-PK in PBMCs. Inhibition of the DDR pathway by CC-115 resulted in increased sensitivity for chlorambucil and bendamustine in CLL cells. Treatment with the mTORC1 inhibitor everolimus has been reported to decrease lymphadenopathy due to mobilization of CLL cells in some patients59. Treatment with the TORK inhibitor CC-214 blocked

proliferation and partially inhibited CD40-induced drug resistance, with minimal direct cytotoxicity. In contrast, combined inhibition of TORK and DNA-PK by CC-115 induced caspase-dependent cell death in CLL cells. As CC-115-induced cytotoxicity was observed in p53, ATM, SF3B1 or NOTCH1-deficient samples, this points to potential clinical activity of CC-115 in CLL, irrespective of mutation/prognostic status. In accordance, murine studies with MYC-driven lymphomas revealed that combined inhibition of TORK and DNA-PK results in strong induction of p53-independent cell death, and in tumor regression and prolonged survival13.

CC-115 treatment diminished CD40-mediated suppression of Bim protein level. Dual PI3K/TORK inhibition results in decreased microenvironment-induced chemoresistance by increased levels of Bim60. Likewise, rapamycin-induced apoptosis has been reported to be

mediated by Bim61. Combined with increased levels of Bim, the levels of the anti-apoptotic

proteins Bcl-XL, Mcl-1 and Bfl-1 following CD40 activation were lower in CC-115 treated cells, which might contribute to decreased resistance to fludarabine and venetoclax.

Despite the significant clinical activity of inhibitors of BTK or PI3K

δ

, they induce little/no direct cell-death as evidenced by prolonged presence of leukemia cells under treatment9-11. The in vitro activities of CC-115 regarding cytotoxicity, reversal of

CD40-induced drug resistance and inhibition of non-BCR mediated proliferation suggest a favorable profile to prevent emergence or outgrowth of resistant clones. We demonstrated that CC-115 induces cytotoxicity and reduces downstream BCR and CD40L signaling as evidenced by inhibition of pS6, also including in CLL cells obtained from idelalisib-resistant patients. These results provide a rationale for clinical testing of CC-115 in patients resistant to idelalisib, who currently have very poor prognosis.

CC-115 did not induce cytotoxicity in healthy T cells. Activated T cells upregulate the PI3K/mTOR pathway which leads to activation of metabolism, proliferation and survival 16,62. Accordingly, activated T cells treated with CC-115 showed inhibition of

proliferation, which could play an adverse role in protection against infections in vivo, especially upon long term treatment. On the other hand, it has been reported that PI3K

δ

inhibition reduces regulatory T cell (Treg)-mediated suppression of cancer immune surveillance63.

Preliminary clinical testing of CC-115, in patients harboring ATM alterations revealed reduction in LN sizes in almost all patients, while effects on lymphocytosis were more variable. Interestingly, although preclinical data suggest cytotoxic effects of CC-115 to

(18)

3

TORK/DNA-PK INHIBITION IN CLL

effects on lymphocytosis in patients with bi-allelic ATM deletions/mutations, as compared to monoallelic deletions. This observation suggests increased dependency on DNA-PK for DNA damage repair in cells with bi-allelic ATM deletions and is in line with in vivo studies using an ATM-defective Eµ:Myc-driven lymphoma model64. However, patients with

a mono-allelic ATM alteration also showed a decrease in lymphadenopathy. Moreover, this clinical study included only patients with an ATM mutation and it would therefore be interesting to clinically evaluate CC-115 in patients with various prognostic subgroups.

Taken together, our study reveals that dual TORK/DNA-PK inhibition by CC-115 induces direct cytotoxicity and can block signaling pathways that are important for CLL survival, chemo-resistance and proliferation in the LN microenvironment. Preliminary data indeed indicate clinical activity of CC-115. Further clinical evaluation, especially combination therapy with agents that induce DNA damage including fludarabine or combination with venetoclax, seems warranted. Clinical results suggest that CC-115 can be useful for the treatment of CLL.

ACKNOWLEDGEMENTS

We thank the CLL patients for their blood donations and Marjolein Spiering and Dieuwertje Luijks for database management and CLL cell phenotyping. APK is a recipient of a Dutch Cancer Society Clinical Fellowship. CR received funding through the German Research Foundation (KFO-286-RP2).

(19)

3

TORK/DNA-PK INHIBITION IN CLL

REFERENCES

1. Ghia P, Strola G, Granziero L, et al. Chronic lymphocytic leukemia B cells are endowed with the capacity to attract CD4+, CD40L+ T cells by producing CCL22. Eur J

Immunol. 2002;32(5):1403-1413.

2. Burger JA, Gandhi V. The lymphatic tissue microenvironments in chronic lymphocytic leukemia: in vitro models and the significance of CD40-CD154 interactions. Blood. 2009;114(12):2560-2561; author reply 2561-2562.

3. Kurtova AV, Balakrishnan K, Chen R, et al. Diverse marrow stromal cells protect CLL cells from spontaneous and drug-induced apoptosis: development of a reliable and reproducible system to assess stromal cell adhesion-mediated drug resistance. Blood. 2009;114(20):4441-4450. 4. Panayiotidis P, Jones D, Ganeshaguru K, Foroni L, Hoffbrand AV. Human bone marrow stromal

cells prevent apoptosis and support the survival of chronic lymphocytic leukaemia cells in vitro.

Br J Haematol. 1996;92(1):97-103.

5. Burger JA, Tsukada N, Burger M, Zvaifler NJ, Dell’Aquila M, Kipps TJ. Blood-derived nurse-like cells protect chronic lymphocytic leukemia B cells from spontaneous apoptosis through stromal cell-derived factor-1. Blood. 2000;96(8):2655-2663.

6. Smit LA, Hallaert DY, Spijker R, et al. Differential Noxa/Mcl-1 balance in peripheral versus lymph node chronic lymphocytic leukemia cells correlates with survival capacity.

Blood. 2007;109(4):1660-1668.

7. Hoellenriegel J, Meadows SA, Sivina M, et al. The phosphoinositide 3’-kinase delta inhibitor, CAL-101, inhibits B-cell receptor signaling and chemokine networks in chronic lymphocytic leukemia. Blood. 2011;118(13):3603-3612.

8. Burger JA. Bruton’s tyrosine kinase (BTK) inhibitors in clinical trials. Curr Hematol Malig

Rep. 2014;9(1):44-49.

9. Herman SE, Niemann CU, Farooqui M, et al. Ibrutinib-induced lymphocytosis in patients with chronic lymphocytic leukemia: correlative analyses from a phase II study.

Leukemia. 2014;28(11):2188-2196.

10. Furman RR, Sharman JP, Coutre SE, et al. Idelalisib and rituximab in relapsed chronic lymphocytic leukemia. N Engl J Med. 2014;370(11):997-1007.

11. Woyach JA, Furman RR, Liu TM, et al. Resistance mechanisms for the Bruton’s tyrosine kinase inhibitor ibrutinib. N Engl J Med. 2014;370(24):2286-2294.

12. Cheng S, Guo A, Lu P, Ma J, Coleman M, Wang YL. Functional characterization of BTK(C481S) mutation that confers ibrutinib resistance: exploration of alternative kinase inhibitors.

Leukemia. 2015;29(4):895-900.

13. Shortt J, Martin BP, Newbold A, et al. Combined inhibition of PI3K-related DNA damage response kinases and mTORC1 induces apoptosis in MYC-driven B-cell lymphomas.

Blood. 2013;121(15):2964-2974.

14. Blunt MD, Carter MJ, Larrayoz M, et al. The PI3K/mTOR inhibitor PF-04691502 induces apoptosis and inhibits microenvironmental signaling in CLL and the Emicro-TCL1 mouse model.

Blood. 2015;125(26):4032-4041.

15. Knittel G, Liedgens P, Reinhardt HC. Targeting ATM-deficient CLL through interference with DNA repair pathways. Front Genet. 2015;6:207.

16. Laplante M, Sabatini DM. mTOR signaling in growth control and disease. Cell. 2012;149(2):274-293. 17. Hara K, Yonezawa K, Weng QP, Kozlowski MT, Belham C, Avruch J. Amino acid sufficiency and

mTOR regulate p70 S6 kinase and eIF-4E BP1 through a common effector mechanism. J Biol

(20)

3

TORK/DNA-PK INHIBITION IN CLL

the rictor-mTOR complex. Science. 2005;307(5712):1098-1101.

19. Wicker LS, Boltz RC, Jr., Matt V, Nichols EA, Peterson LB, Sigal NH. Suppression of B cell activation by cyclosporin A, FK506 and rapamycin. Eur J Immunol. 1990;20(10):2277-2283.

20. Kay JE, Kromwel L, Doe SE, Denyer M. Inhibition of T and B lymphocyte proliferation by rapamycin. Immunology. 1991;72(4):544-549.

21. Aagaard-Tillery KM, Jelinek DF. Inhibition of human B lymphocyte cell cycle progression and differentiation by rapamycin. Cell Immunol. 1994;156(2):493-507.

22. Decker T, Hipp S, Ringshausen I, et al. Rapamycin-induced G1 arrest in cycling B-CLL cells is associated with reduced expression of cyclin D3, cyclin E, cyclin A, and survivin. Blood. 2003;101(1):278-285. 23. Lee K, Heffington L, Jellusova J, et al. Requirement for Rictor in homeostasis and function of

mature B lymphoid cells. Blood. 2013;122(14):2369-2379.

24. Zhang S, Pruitt M, Tran D, et al. B cell-specific deficiencies in mTOR limit humoral immune responses. J Immunol. 2013;191(4):1692-1703.

25. Sharma A, Janocha AJ, Hill BT, Smith MR, Erzurum SC, Almasan A. Targeting mTORC1-mediated metabolic addiction overcomes fludarabine resistance in malignant B cells. Mol Cancer

Res. 2014;12(9):1205-1215.

26. Ringshausen I, Peschel C, Decker T. Mammalian target of rapamycin (mTOR) inhibition in chronic lymphocytic B-cell leukemia: a new therapeutic option. Leuk Lymphoma. 2005;46(1):11-19. 27. Hill R, Lee PW. The DNA-dependent protein kinase (DNA-PK): More than just a case of making

ends meet? Cell Cycle. 2010;9(17):3460-3469.

28. Mao Z, Bozzella M, Seluanov A, Gorbunova V. DNA repair by nonhomologous end joining and homologous recombination during cell cycle in human cells. Cell cycle

(Georgetown, Tex). 2008;7(18):2902-2906.

29. Dietlein F, Reinhardt HC. Molecular pathways: exploiting tumor-specific molecular defects in DNA repair pathways for precision cancer therapy. Clin Cancer Res. 2014;20(23):5882-5887. 30. Dietlein F, Thelen L, Reinhardt HC. Cancer-specific defects in DNA repair pathways as targets for

personalized therapeutic approaches. Trends Genet. 2014;30(8):326-339.

31. Vrhovac R, Delmer A, Tang R, Marie JP, Zittoun R, Ajchenbaum-Cymbalista F. Prognostic significance of the cell cycle inhibitor p27Kip1 in chronic B-cell lymphocytic leukemia. Blood. 1998;91(12):4694-4700. 32. Hallaert DY, Jaspers A, van Noesel CJ, van Oers MH, Kater AP, Eldering E. c-Abl kinase inhibitors

overcome CD40-mediated drug resistance in CLL: implications for therapeutic targeting of chemoresistant niches. Blood. 2008;112(13):5141-5149.

33. Te Raa GD, Derks IA, Navrkalova V, et al. The impact of SF3B1 mutations in CLL on the DNA-damage response. Leukemia. 2015;29(5):1133-1142.

34. Austen B, Powell JE, Alvi A, et al. Mutations in the ATM gene lead to impaired overall and treatment-free survival that is independent of IGVH mutation status in patients with B-CLL.

Blood. 2005;106(9):3175-3182.

35. Skowronska A, Parker A, Ahmed G, et al. Biallelic ATM inactivation significantly reduces survival in patients treated on the United Kingdom Leukemia Research Fund Chronic Lymphocytic Leukemia 4 trial. J Clin Oncol. 2012;30(36):4524-4532.

36. Pascutti MF, Jak M, Tromp JM, et al. IL-21 and CD40L signals from autologous T cells can induce antigen-independent proliferation of CLL cells. Blood. 2013;122(17):3010-3019.

37. Stiff T, O’Driscoll M, Rief N, Iwabuchi K, Lobrich M, Jeggo PA. ATM and DNA-PK function redundantly to phosphorylate H2AX after exposure to ionizing radiation. Cancer Res. 2004;64(7):2390-2396.

(21)

3

TORK/DNA-PK INHIBITION IN CLL

38. Wang H, Wang M, Wang H, Bocker W, Iliakis G. Complex H2AX phosphorylation patterns by multiple kinases including ATM and DNA-PK in human cells exposed to ionizing radiation and treated with kinase inhibitors. J Cell Physiol. 2005;202(2):492-502.

39. Quanz M, Herbette A, Sayarath M, et al. Heat shock protein 90alpha (Hsp90alpha) is phosphorylated in response to DNA damage and accumulates in repair foci. J Biol Chem. 2012;287(12):8803-8815. 40. Reinhardt HC, Yaffe MB. Phospho-Ser/Thr-binding domains: navigating the cell cycle and DNA

damage response. Nat Rev Mol Cell Biol. 2013;14(9):563-580.

41. Tonino SH, van Laar J, van Oers MH, Wang JY, Eldering E, Kater AP. ROS-mediated upregulation of Noxa overcomes chemoresistance in chronic lymphocytic leukemia. Oncogene. 2011;30(6):701-713. 42. Herman SE, Gordon AL, Wagner AJ, et al. Phosphatidylinositol 3-kinase-delta inhibitor CAL-101

shows promising preclinical activity in chronic lymphocytic leukemia by antagonizing intrinsic and extrinsic cellular survival signals. Blood. 2010;116(12):2078-2088.

43. Kater AP, Evers LM, Remmerswaal EB, et al. CD40 stimulation of B-cell chronic lymphocytic leukaemia cells enhances the anti-apoptotic profile, but also Bid expression and cells remain susceptible to autologous cytotoxic T-lymphocyte attack. Br J Haematol. 2004;127(4):404-415. 44. Girbl T, Hinterseer E, Grossinger EM, et al. CD40-mediated activation of chronic lymphocytic

leukemia cells promotes their CD44-dependent adhesion to hyaluronan and restricts CCL21-induced motility. Cancer Res. 2013;73(2):561-570.

45. Thijssen R, Ter Burg J, van Bochove GG, et al. The pan phosphoinositide 3-kinase/mammalian target of rapamycin inhibitor SAR245409 (voxtalisib/XL765) blocks survival, adhesion and proliferation of primary chronic lymphocytic leukemia cells. Leukemia. 2015.

46. Thijssen R, Slinger E, Weller K, et al. Resistance to ABT-199 induced by microenvironmental signals in chronic lymphocytic leukemia can be counteracted by CD20 antibodies or kinase inhibitors. Haematologica. 2015;100(8):e302-306.

47. Tromp JM, Tonino SH, Elias JA, et al. Dichotomy in NF-kappaB signaling and chemoresistance in immunoglobulin variable heavy-chain-mutated versus unmutated CLL cells upon CD40/TLR9 triggering. Oncogene. 2010;29(36):5071-5082.

48. Ciccia A, Elledge SJ. The DNA damage response: making it safe to play with knives. Mol

Cell. 2010;40(2):179-204.

49. Davidson D, Amrein L, Panasci L, Aloyz R. Small Molecules, Inhibitors of DNA-PK, Targeting DNA Repair, and Beyond. Front Pharmacol. 2013;4:5.

50. Willmore E, de Caux S, Sunter NJ, et al. A novel DNA-dependent protein kinase inhibitor, NU7026, potentiates the cytotoxicity of topoisomerase II poisons used in the treatment of leukemia. Blood. 2004;103(12):4659-4665.

51. Zhao Y, Thomas HD, Batey MA, et al. Preclinical evaluation of a potent novel DNA-dependent protein kinase inhibitor NU7441. Cancer Res. 2006;66(10):5354-5362.

52. Amrein L, Loignon M, Goulet AC, et al. Chlorambucil cytotoxicity in malignant B lymphocytes is synergistically increased by 2-(morpholin-4-yl)-benzo[h]chomen-4-one (NU7026)-mediated inhibition of DNA double-strand break repair via inhibition of DNA-dependent protein kinase. J

Pharmacol Exp Ther. 2007;321(3):848-855.

53. Deriano L, Guipaud O, Merle-Beral H, et al. Human chronic lymphocytic leukemia B cells can escape DNA damage-induced apoptosis through the nonhomologous end-joining DNA repair pathway. Blood. 2005;105(12):4776-4783.

54. Willmore E, Elliott SL, Mainou-Fowler T, et al. DNA-dependent protein kinase is a therapeutic target and an indicator of poor prognosis in B-cell chronic lymphocytic leukemia. Clin Cancer

(22)

3

TORK/DNA-PK INHIBITION IN CLL

and the nonhomologous end-joining repair pathways in chronic lymphocytic leukemia therapy.

Leuk Res. 2011;35(8):1080-1086.

56. Muller C, Salles B. Regulation of DNA-dependent protein kinase activity in leukemic cells.

Oncogene. 1997;15(19):2343-2348.

57. Eriksson A, Lewensoh R, Larsson R, Nilsson A. DNA-dependent protein kinase in leukaemia cells and correlation with drug sensitivity. Anticancer Res. 2002;22(3):1787-1793.

58. Muller C, Christodoulopoulos G, Salles B, Panasci L. DNA-Dependent protein kinase activity correlates with clinical and in vitro sensitivity of chronic lymphocytic leukemia lymphocytes to nitrogen mustards. Blood. 1998;92(7):2213-2219.

59. Zent CS, LaPlant BR, Johnston PB, et al. The treatment of recurrent/refractory chronic lymphocytic leukemia/small lymphocytic lymphoma (CLL) with everolimus results in clinical responses and mobilization of CLL cells into the circulation. Cancer. 2010;116(9):2201-2207.

60. Rosich L, Saborit-Villarroya I, Lopez-Guerra M, et al. The phosphatidylinositol-3-kinase inhibitor NVP-BKM120 overcomes resistance signals derived from microenvironment by regulating the Akt/ FoxO3a/Bim axis in chronic lymphocytic leukemia cells. Haematologica. 2013;98(11):1739-1747. 61. Lopez-Royuela N, Balsas P, Galan-Malo P, Anel A, Marzo I, Naval J. Bim is the key mediator of

glucocorticoid-induced apoptosis and of its potentiation by rapamycin in human myeloma cells.

Biochim Biophys Acta. 2010;1803(2):311-322.

62. Chi H. Regulation and function of mTOR signalling in T cell fate decisions. Nat Rev

Immunol. 2012;12(5):325-338.

63. Ali K, Soond DR, Pineiro R, et al. Inactivation of PI(3)K p110delta breaks regulatory T-cell-mediated immune tolerance to cancer. Nature. 2014;510(7505):407-411.

64. Riabinska A, Daheim M, Herter-Sprie GS, et al. Therapeutic targeting of a robust non-oncogene addiction to PRKDC in ATM-defective tumors. Sci Transl Med. 2013;5(189):189ra178.

(23)

3

TORK/DNA-PK INHIBITION IN CLL Ta b le S 1. P at ie nt s’ c ha ra ct er is tic s Patient Age Gender   IgVH mutation WBC 10^9/L % lymphocytes %CD19+/ CD5+ cells FISH Mutation Tr eatment befor e sampling 1 73 M M ND ND 13.3 13q-  none 2A 30 M U 286 ND 65.4 11q-FCR 2B 30 M U 424.2 99 94.6 11q-FCR 2C 27 M U 160.5 94.6 93.8 11q-AT M none 3A 81 M M 80.8 90.5 90.6 13q-  none 3B 80 M M 86.4 87 93.4 13q-  none 4A 64 F M 175 95.1 94 13q-  none 4B 62 F M 62 87 90.7 13q-  none 5 77 F M 120 94 93.3 ND   none 6 60 M M 64.3 ND   13q-  none 7 42 M M 66.6 92 86.3 13q-  none 8A 63 F M 32.2 86.7 80.6 13q-  none 8B 66 F M 23.8 80 88.8 13q-  none 9 74 F M 102 86.7 99.8 none NOTCH1 chlorambucil + pr ednisone 10 unknown unknown unknown ND ND ND none   unknown 11 unknown unknown unknown ND ND ND none   unknown 12 44 M U ND ND ND 11q-, 13q-AT M none 13 76 M U 46.4 93.3 91.09 11q-, 13q-AT M chlorambucil 14 63 unknown U ND ND ND 11q-AT M none 15 70 unknown U ND ND ND 11q-AT M

fludarabine + cyclophosphamide, fludarabine

16 74 M U 127 ND 93 none   none 17 69 M M ND ND 95.2 13q-  chlorambucil 18 62 M U 192.1 92.9 94.9 13q-  none 19A 63 F M 153.0 96.0 95.7 13q-, 17p-  chlorambucil, FCR 19B 62 F M 173.0 ND 99.9 13q-  none 20 70 M U 422 ND 92.8 17p-  chlorambucil, CVP/COP , FCR 21 56 F M ND ND 94.7 13q-  none

SUPPLEMENTARY MATERIALS

(24)

3

TORK/DNA-PK INHIBITION IN CLL Ta b le S 1. (co nt inu ed ) Patient Age Gender IgVH mutation WBC 10^9/L % lymphocytes %CD19+/ CD5+ cells FISH Mutation Tr eatment befor e sampling 22 67 F M 88.7 ND 86.9 13q-  chlorambucil + pr ednison, fludarabine 23 62 F M 84.8 ND 99.9 13q-  none 24 88 F M 53.8 87.2 87.1 ND   none 25 83 F M 56.8 87.6 88.1 13q-, tris12   none 26 61 F M 46.1 ND 91.1 13q-  none 27 81 F M 23.1 87.3 93.3 ND   none 28 44 F M 115 ND 92.9 13q-  chlorambucil 29 46 M M 12.8 72.4 99.5 ND   none 30 50 M M 56.5 90 95.2 ND   none 31 86 M M 90.5 90.8 81 13q-  chlorambucil 32A 56 M U 78.1 ND 89.6 tris12   chlorambucil 32B 50 M U 149 94 90 tris12   chlorambucil 33A 72 F U 115.3 ND 90.3 tris12   none 33B 73 F U 89.9 ND 91.4 tris12   none 33C 74 F U 89.9 ND 91.4 tris12   none 34A 60 M U 69.5 ND 94 13q-  none 34B 60 M U ND ND 91 13q-  none 35A 73 M U 116.8 ND 98.4 ND   none 35B 74 M U 166.6 ND 84.2 ND   chlorambucil 36 60 M U 21.9 75.1 99.2 ND   none 37 76 M U 49.7 ND 88.6 13q-  none 38 52 M U 132 92.1 86.2 tris12   none 39 65 M M ND ND 92.1 none   none 40 60 F U 265 ND 99.8 13q-  FCR 41 41 M M 40 84 91.4 13q-  none 42 58 M U 55.7 83 94.4 13q- 

chlorambucil + fludarabine + cyclofosfamide

43A 55 F U 272 ND 95.4 none   FCR

(25)

3

TORK/DNA-PK INHIBITION IN CLL Ta b le S 1. (co nt inu ed ) Patient Age Gender IgVH mutation WBC 10^9/L % lymphocytes %CD19+/ CD5+ cells FISH Mutation Tr eatment befor e sampling 43B 55 F U 180 ND 95.1 none   FCR 44 71 F M 47.3 91.1 95.9 ND   none 45 60 M M 85.5 94 95.8 13q-  none 46 65 F U 89.9 90 89 ND   none 47 53 M U 31.1 95.8 82.1 11q-AT M chlorambucil, radiotherapy , CVP/COP , FCR 48 44 M U ND ND ND 13q-, 11q-AT M none 49 68 unknown U ND ND ND 11q-AT M none 50 80 unknown M ND ND ND 11q-AT M none 51 70 unknown U ND ND ND 11q-AT M

fludarabine + cyclophosphamide, fludarabine

52 77 unknown U ND ND ND 11q-AT M none 53 68 unknown U ND ND ND 11q-AT M none 54 44 unknown U ND ND ND 11q-AT M none 55 64 unknown U ND ND ND 11q-AT M

fludarabine + cyclophosphamide, alemtuzumab

56 36 M U 82.5 75.1 85.13 17p-TP53 CVP/COP , FCR, alemtuzumab, R-CHOP , R-DHAP 57 59 M U ND ND ND 17p-TP53 none 58 82 M M 137 94.8 83.5 13q-, 17p-TP53 chlorambucil, alemtuzumab 59 58 M M 155 ND 87.9 tris12, 17p-TP53 chlorambucil + pr ednison, R-CHOP , FCR, Rituximab, R-DHAP 60 63 unknown U ND ND ND 17p-TP53

fludarabine + cyclophosphamide, R-CHOP

, alemtuzumab 61 75 unknown U ND ND ND 17p-, 11q-TP53 chlorambucil, FCR, alemtuzumab 62 67 F U ND ND 99.6 none SF3B1 chlorambucil, radiotherapy , CVP/COP , fludarabine 63 69 M M 11.4 32.7 18.59 none SF3B1 none 64 82 F M 20.6 65.9 76.22 ND SF3B1 chlorambucil

(26)

3

TORK/DNA-PK INHIBITION IN CLL Ta b le S 1. (co nt inu ed ) Patient Age Gender IgVH mutation WBC 10^9/L % lymphocytes %CD19+/ CD5+ cells FISH Mutation Tr eatment befor e sampling 65 60 M U ND ND ND 11q-SF3B1 none 66 72 M unknown 87.9 86 82.4 13q-, 11q-NOTCH1 chlorambucil, R-CVP , FCR + R-CHOP dasatinib + fludarabine 67 76 M M 31.9 96.1 91.74 none NOTCH1 chlorambucil 68 73 M M 47.3 86.8 89.29 none NOTCH1 none 69 57 F M 33.9 85.5 85.7 ND   none 70 76 M M 74.1 76 99.1 ND   none 71 78 M U 100 74 94.1 tris12   none 72 58 F M 170.9 ND 97.9 13q-, tris12  

chlorambucil, fludarabine + cyclophosphamide, fludarabine

73 63 F M 92.7 ND 93.7 none   none 74 68 F U 71.7 83.2 97 ND   none 75 78 M U 100 74 94.1 tris12   none 76 75 F M 92.9 91 85.9 ND   none 77 63 F M 79.2 91,0 92.3 ND   none 78 80 M M 149.4 92 98.2 none   chlorambucil 79 68 F U 100.5 89 84.8 tris12   none 80 66 M M 93.4 ND 90.8 none   none 81 73 M U 108 94.7 96.4 17p- 

chlorambucil, fludarabine, alemtuzumab, FCR

82 65 F M 59 92 87.6 ND   none 83 50 F M 80.2 96 91.4 ND   none 84 48 M unknown ND ND ND 13q-R-CHOP 85 58 F unknown ND ND ND 11q-AT M none 86 76 F unknown ND ND ND 11q-, 13q-AT M none 87 72 M unknown ND ND ND 11q-AT M none 88 44 M unknown ND ND ND 11q-AT M none

(27)

3

TORK/DNA-PK INHIBITION IN CLL Ta b le S 1. (co nt inu ed ) Patient Age Gender IgVH mutation WBC 10^9/L % lymphocytes %CD19+/ CD5+ cells FISH Mutation Tr eatment befor e sampling 89 63 M unknown ND ND ND none none 90 52 M unknown ND ND ND none none 91 75 F unknown ND ND ND none none 92 68 M unknown ND ND ND none none

Gender M: Male; F: Female, IgVH mutation M: Mutated; U: Unmutated, ND: not deter

mined A, B, C r

efer to dif

fer

ent sampling times of

the

same patient;

FCR: fludarabine + cyclophosphamide + rituximab; CVP/COP: cyclophosphamide + vincristine/oncovin + pr

ednisolone

R-CHOP: rituximab + cyclophosphamide + vincristine/oncovin + pr

(28)

3

TORK/DNA-PK INHIBITION IN CLL Ta bl e S 2. P at ie nt c ha ra ct er is tic s f o r c lin ic al s tu d y C C -1 15 -S T-00 1, C LL & S LL s ub je ct s  Patient Age Gender IgVH mutation WBC 10^9/L % lymphocytes FISH Mutation Prior T reatment Best Response 0011034* 62 F unknown 4.8 27.0 11q-, 13q-AT M   R-CHOP , FCR, R, BR, NE anti CD20 Ab 2011004 54 M unknown 100.7 90.7 11q-A TM** FC, alemtuzumab PR 2011005 60 M unknown 64.9 91.2 11q-AT M FCR PD 2011007 50 F U  65.3 89.5 11q-A TM** FCR SD 3011019 75 F unknown 17.4 89.4 11q-, 17p-A TM** CHOP , F , FCR, bendamustine, ofatumumab, R-CHOP , BR, PR with lyphocytosis 4011001 57 M U 32.2 61 11q-, 1q-DER 17;22 AT M  

FC, BR, Dex alemtuzumab, lenalidomide

PR with lyphocytosis 4011002 54 M unknown 65.8 64 11q-AT M   FC, BR PR with lyphocytosis 4011003 55 M U 17.6 90 11q-AT M   FCR, CHOP NE

* SLL subject **Likely or potentially deleterious mutation in r

emaining A

TM

U: Unmutated, NE: not evaluable, PR: partial r

esponse, PD: pr

ogr

essive disease, SD: stable disease

FCR: fludarabine + cyclophosphamide + rituximab; CVP/COP: cyclophosphamide + vincristine/oncovin + pr

ednisolone; Dex:dexamethsone

R-CHOP: rituximab + cyclophosphamide + doxorubicin + vincristine/oncovin + pr

ednisolone;

(29)

3

TORK/DNA-PK INHIBITION IN CLL

Table S3. Primer sequences

Primer sequence p53_exon4_Fw 5’-TGTAAAACGACGGCCAGTCTGGTAAGGACAAGGGTTGG-3’ p53_exon4_Rv 5’-CAGGAAACAGCTATGACCGATACGGCCAGGCATTGAAG-3’ p53_exon5_Fw 5’-TGTAAAACGACGGCCAGTCTAGCTCGCTAGTGGGTTGC-3’ p53_exon5_Rv 5’-CAGGAAACAGCTATGACCCACTCGGATAAGATGCTGAG-3’ p53_exon6_Fw 5’-TGTAAAACGACGGCCAGTCCACCATGAGCGCTGCTCAG-3’ p53_exon6_Rv 5’-CAGGAAACAGCTATGACCCCCTTAGCCTCTGTAAGCTTC-3’ p53_exon7_Fw 5’-TGTAAAACGACGGCCAGTGCCTCCCCTGCTTGCCACAG-3’ p53_exon7_Fw2 5’-TGTAAAACGACGGCCAGTCCTCCCCTGCTTGCCACAG-3’ p53_exon7_Rv 5’-CAGGAAACAGCTATGACCGGGAGCAGTAAGGAGATTCC-3’ p53_exon8&9_Fw 5’-TGTAAAACGACGGCCAGTTTCCTTACTGCCTCTTGCTT-3’ p53_exon8&9_Rv 5’-CAGGAAACAGCTATGACCAGAAAACGGCATTTTGAGTG-3’ p53_exon10_Fw 5’-TGTAAAACGACGGCCAGTCAATTGTAACTTGAACCATC-3’ p53_exon10_Rv 5’-CAGGAAACAGCTATGACCGGATGAGAATGGAATCCTAT-3’

Referenties

GERELATEERDE DOCUMENTEN

contrast early life dampness or mold exposure was associated with higher level of lung 362. function at both ages 12

In the subsequent survival experiments, we demonstrated that this technique could indeedd prevent hind limb neurologic deficit after one hour of aortic cross-clamping (chapters 44

Transcranial myogenic motor evoked potentialss (tc-MEPs) seem to meet these requirements. Thee present thesis described the application of various protective techniques for the spinal

Two-way univariate analyses of variance with as factors language of the narrative (L1/L2) and nationality of the reader (Dutch/German) were carried out in order to measure

Het verschil in effect op de Mate waarin de advertentie angstaanjagend werd gevonden voor de aan- en afwezigheid van een humoristische appeal in de advertentie bleek alleen op

The method used to plant these legumes is hydroseeding (Pt. The next step is to plant trees as a method to prevent soil erosion over longer periods of time. The trees that

Les participants qui ont regardé la vidéo instructive dans la langue maternelle ou ceux qui ont regardé la vidéo instructive avec sous-titrage, ont plus souvent utilisé la

recognise that public participation needs to be more integrated in the Netherlands, as it has not only practical implications, but on scientific research that underpins