targeting oncogenic protein sythesis in myc-driven b cell

89
University of Nebraska Medical Center University of Nebraska Medical Center DigitalCommons@UNMC DigitalCommons@UNMC Theses & Dissertations Graduate Studies Spring 5-4-2019 Targeting Oncogenic Protein Sythesis in MYC-driven B Cell Targeting Oncogenic Protein Sythesis in MYC-driven B Cell Lymphoma Lymphoma Xuan Zhang University of Nebraska Medical Center Follow this and additional works at: https://digitalcommons.unmc.edu/etd Part of the Cancer Biology Commons Recommended Citation Recommended Citation Zhang, Xuan, "Targeting Oncogenic Protein Sythesis in MYC-driven B Cell Lymphoma" (2019). Theses & Dissertations. 341. https://digitalcommons.unmc.edu/etd/341 This Dissertation is brought to you for free and open access by the Graduate Studies at DigitalCommons@UNMC. It has been accepted for inclusion in Theses & Dissertations by an authorized administrator of DigitalCommons@UNMC. For more information, please contact [email protected].

Upload: others

Post on 19-Oct-2021

4 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Targeting Oncogenic Protein Sythesis in MYC-driven B Cell

University of Nebraska Medical Center University of Nebraska Medical Center

DigitalCommons@UNMC DigitalCommons@UNMC

Theses & Dissertations Graduate Studies

Spring 5-4-2019

Targeting Oncogenic Protein Sythesis in MYC-driven B Cell Targeting Oncogenic Protein Sythesis in MYC-driven B Cell

Lymphoma Lymphoma

Xuan Zhang University of Nebraska Medical Center

Follow this and additional works at: https://digitalcommons.unmc.edu/etd

Part of the Cancer Biology Commons

Recommended Citation Recommended Citation Zhang, Xuan, "Targeting Oncogenic Protein Sythesis in MYC-driven B Cell Lymphoma" (2019). Theses & Dissertations. 341. https://digitalcommons.unmc.edu/etd/341

This Dissertation is brought to you for free and open access by the Graduate Studies at DigitalCommons@UNMC. It has been accepted for inclusion in Theses & Dissertations by an authorized administrator of DigitalCommons@UNMC. For more information, please contact [email protected].

Page 2: Targeting Oncogenic Protein Sythesis in MYC-driven B Cell

P a g e | I

TARGETING ONCOGENIC PROTEIN SYNTHESIS IN MYC-DRIVEN B

CELL LYMPHOMA

by

Xuan Zhang

A DISSERTATION

Presented to the Faculty of

the University of Nebraska Graduate College

In Partial Fulfillment of the Requirements

For the Degree of Doctor of Philosophy

Pathology and Microbiology

Graduate Program

Under the Supervision of Professor Kai Fu

University of Nebraska Medical Center

Omaha, Nebraska

January, 2019

Supervisory Committee:

Matthew Lunning, DO Robert Lewis, Ph.D. Rakesh K. Singh, Ph.D. Shantaram S Joshi, Ph.D.

Page 3: Targeting Oncogenic Protein Sythesis in MYC-driven B Cell

II

ACKNOWLEDGEMENTS

The Ph.D program at UNMC is valuable to my career development and also greatly

changed the way I think. This five years of training transformed me from a learner of

science to an explorer of science. As I am writing this dissertation now, I feel proud that

my efforts could contribute to a better understanding of this disease. Yet little could be

achieved without the help and support from people around me, herein I would like to

express my sincere gratitude to them.

First I want to thank my advisor Dr.Fu for his overall guidance of my study. Dr.Fu has

always been encouraging and often inspires me with his own experience in his Ph.D

training years. He is exceptional knowledgeable in the field of hematopathology from

whom I leant a lot of B cell malignancy morphology and pathogenesis. As a chief

pathologist in the department of Pathology at UNMC, Dr.Fu established the connection

between laboratory work and clinical practices, which is utterly advantageous for our

translational research. Therefore, he provides me a lot of opportunities to work with

clinical labs and availability of patient samples, from which we established numerous

patient derived xenograft models and valuable profiling data.

I would like to extend my appreciation to my committee members. For their guidance

and criticism, especially in my committee meetings and comprehensive exam.

Secondly, I would like to thank Dr. Chengfeng (Andy) Bi as he bequeaths the

knowledge of experimental techniques and skills to me. Andy is the instructor in our lab

from whom I learnt the basics like PCR and western when I first came, and together we

Page 4: Targeting Oncogenic Protein Sythesis in MYC-driven B Cell

III

studied and developed new techniques and pipelines. I shall not forget the days when

there were only two research personnel in the lab and how we worked back to back. I

would also like to thank Ph.D. students Tian Tian, and Cheng Wang for their constructive

suggestions and support on my research. In addition, I am grateful for the contributions

to my project from visiting Ph.D student Ting Lu and summer student Ting Yue.

Next, I want to express my gratitude towards our collaborators. Dr. Jianguo Tao and

Dr. Xiaohong Zhao in Moffitt Cancer Center helped me generating the drug screening

data and I also participated in several of their projects. Dr. John A. Porco and his lab in

Boston University provided rocaglates for this study. Dr. Christopher D. Carey and Dr.

Scott J. Rodig in Brigham and Women’s hospital generated classifier score and

collaborated with us in the nanostring B cell classification study.

In addition, I really appreciate the quality service from our core facilities and clinical

labs. Dr. Jian Jiang and Dr. Lijun Sun in tissue science core facility. Ronald Redder and

faculty at UNMC sequencing core. Dr. Sophie Alvarez Y Albala and faculty of protein core

at UNL. Victoria B. Smith, B.S, Samantha Wall, B.S and faculty of flow cytometry core at

UNMC. Faculty of UNMC frozen section lab.

Finally, I would like to thank NIH and UNMC department of pathology for funding

support. I want to thank my family and friend for their caring and faith in me.

Page 5: Targeting Oncogenic Protein Sythesis in MYC-driven B Cell

IV

ABSTRACT

Burkitt lymphoma (BL) and diffuse large B-cell lymphoma (DLBCL) are aggressive

tumors of mature B cells that can be distinguished based on histomorphological,

phenotypic, and genetic features. B-cell lymphoma unclassifiable, with features

intermediate between BL and DLBCL (BCL-U) is a subset of mature B cell lymphoma

contains one or more features that overlap BL and DLBCL. Previous molecular analyses

prove there is a biological continuum between BL and DLBCL based on the activity of

MYC. Herein, in order to resolve the molecular heterogeneity of BCL-U, we tested

whether a targeted expression profiling panel could categorize tumors as BL and DLBCL.

Among these MYC driven lymphomas, those with concurrent MYC and BCL2

dysregulation are especially a challenge in clinical practice due to rapid disease

progression, resistance to standard chemotherapy and high risk of refractory. MYC plays

a central role by coordinating hyperactive protein synthesis with upregulated

transcription in order to support the rapid proliferation. Translation initiation inhibitor

rocaglates were identified as the most potent drug across all MYC driven lymphomas as

they efficiently inhibit MYC expression and tumor cell viability. Surprisingly, we found

this class of compounds does not inhibit translation by depleting the abundance of eIF4A

in eIF4F, but more likely acts as a selective roadblock to prevent ribosome scanning.

Furthermore, proteome wide quantification demonstrated rocaglate induced selective

repression of multiple critical oncoproteins beside MYC in B cell lymphoma including

NEK2, MCL1, AURKA, PLK1 and several transcription factors that generally considered

Page 6: Targeting Oncogenic Protein Sythesis in MYC-driven B Cell

V

undruggable. Finally, (-)-SDS-1-021, the most promising rocaglate, was confirmed highly

potent as single agent, and displayed significant synergy with BCL2 inhibitor ABT199 in

inhibiting tumor growth and survival in patient derived xenograft mouse models.

Overall, our findings support the strategy of using rocaglates to target oncoprotein

synthesis in MYC-driven lymphomas.

Page 7: Targeting Oncogenic Protein Sythesis in MYC-driven B Cell

VI

TABLE OF CONTENTS

List of Figures Figure 1.1. Illustration of Updated B cell Lymphoma Classification ................................. 12 Figure 2.1. Results of the MYCKEY Diagnostic classifier for the Aggressive B-cell lymphoma test cohort ................................................................................................................. 19 Figure 2.2. Diagnostic scores (0-1) for each tumor as profiled in UNMC, Omaha (x-axis) and BWH, Boston (y-axis) .......................................................................................................... 20 Figure 3.1 Inhibitors targeting protein synthesis exhibit highest potency in MYC driven lymphomas .................................................................................................................................. 40 Figure 3.2. Rocaglates but not mTOR inhibitors efficiently repress MYC expression. ..... 44 Figure 3.3. Impact of translation inhibitors on oncoprotein expression in MYC driven lymphomas. ................................................................................................................................ 45 Figure 3.4. silvestrol strongly inhibits cap-dependent and IRES translation of MYC ...... 49 Figure 3.5. Expression of eIF4A1/2 and eIF4A knockdown ................................................. 51 Figure 3.6. Rocaglates increase MYC mRNA binding to eIF4A, instead of depleting eIF4A in eIF4F ............................................................................................................................. 54 Figure 3.7. silvestrol increases rocaglate target mRNA enrichment on eIF4A1/2 ............. 57 Figure 3.8. Rocaglates repress multiple critical oncoproteins in DHL ............................... 60 Figure 3.9. Downregulated proteins pathway enrichment analysis and target validation in Torin1 treatment ..................................................................................................................... 61 Figure 3.10. Potency of rocalgates and combined treatment with ABT199 in vitro .......... 64 Figure 3.11 Structure and EC50 of silvestrol and (-)-SDS-1-021 ........................................... 65 Figure 3.12. (-)-SDS-1-021 displayed high potency as single agent in DEL PDX model .. 68 Figure 3.13. (-)-SDS-1-021 displayed high potency, and synergy with ABT199 DHL PDX model ............................................................................................................................................ 69

List of Tables Table 2.1. Summary of results .................................................................................................. 19 Table S1A. List of antibodies used for immunoblots ............................................................ 37 Table S1B. List of qRT primers ................................................................................................. 38

Page 8: Targeting Oncogenic Protein Sythesis in MYC-driven B Cell

P a g e | 7

LIST OF ABBREVIATIONS

BL Burkitt Lymphoma

DLBCL Diffuse Large B cell lymphoma

NOS Non other specific

BCR B cell receptor

GC Germinal center

FDC Follicular dendritic cells

FL Follicular lymphoma

PBL Plasmablastic lymphoma

IGH Immunoglobin heavy chain

FISH Fluorescent in situ hybridization

DHL Double hit lymphoma

IHC Immunochemistry

ABC Activated B cell

GCB Germinal center B cell

RPMI Roswell Park Memorial Institute

ASCT Autologous stem cell transplantation

IRES Internal ribosome entry site

ITAF IRES trans-acting factors

mTOR Mammalian target of rapamycin

Page 9: Targeting Oncogenic Protein Sythesis in MYC-driven B Cell

8

eIF Eukaryotic initiation factor

4EBP1 eIF4E binding protein 1

RPS6 Ribosome protein 6

UTR Untranslated region

PDX Patient derived xenograft

Page 10: Targeting Oncogenic Protein Sythesis in MYC-driven B Cell

9

Chapter One: MYC driven B cell lymphoma and Double hit lymphoma

MYC is a master regulator of transcription. This basic helix–loop–helix leucine zipper

protein dimerizes with MAX to bind enhancer box sequences (E-boxes) and regulation

transcription by recruiting specific coactivators and repressors. MYC controls cellular

processes necessary for normal cell growth and proliferation via transcriptional and non-

transcriptional mechanisms including cell cycle and DNA replication, ribosome

biogenesis, metabolism and apoptosis. Hence MYC was thought be expressed in all

tissues undergoing rapid cellular expansion and renewal. In the past decade, the function

of MYC as a transcription factor and its targets have been redefined several times. Current

understanding is MYC drives differential expression of its bona fide target gene subsets1,

meanwhile it could also enhance the expression of existing genetic background2-4.

Overexpression of MYC activates and cooperates with multiple oncogenic pathways,

therefore strongly promotes malignant transformation and disease progression5.

MYC is essential and tightly regulated in normal B cell development and affinity

maturation6,7. Transient MYC expression is induced in a subgroup of mature B cells

through the interaction with antigen and T cells to initiate the germinal center, and then

repressed by BCL6 when GC is formed. Re-expression of MYC happens in a subgroup of

activated B cells with high BCR affinity in the light zone that are ready to reenter dark

zone for proliferation. In these cells, MYC promotes G0/G1-S transition by activating of

genes encoding cyclin-dependent kinase (CDK) complex proteins, including Cyclin D2

(CCND2), as well as by repressing CDK inhibitors. In the dark zone, BCL6 represses MYC

Page 11: Targeting Oncogenic Protein Sythesis in MYC-driven B Cell

10

transcription meanwhile interacts with transcription MIZ1, a binding partner of MYC, to

maintain proliferation independent of MYC and Cyclin D2. Therefore, dysregulated MYC

expression contributes to the transformation of many B cell malignancies, most commonly

as a result of translocation or amplification. Generally, MYC driven lymphomas refers to

Burkitt lymphoma (BL), diffuse large B cell lymphoma, non-other specific (DLBCL, NOS),

B cell lymphoma, unclassifiable with features intermediate between BL and

DLBCL(BCLU), plasmablastic lymphoma (PBL), anaplastic lymphoma kinase (ALK)–

positive large B-cell lymphoma, B-cell lymphoblastic leukemia/lymphoma, and a group

of aggressive mantle cell lymphoma. MYC translocation with immunoglobulin heavy

(IGH) or light chain (IGL) typically detected by fluorescent in situ hybridization (FISH)

was initially found in BL and later became the hallmark of this disease. BL and most

lymphomas that overexpress MYC usually exhibit high proliferation rate and aggressive

clinical presentation. MYC/IG rearrangements are present in ~15% of diffuse DLBCL and

up to 50% of BCL-U which are always found associated with inferior outcome8-11. Notably,

majority of MYC translocated high grade B cell lymphoma also possess a t(14;18)(q32;q21)

IGH/BCL2 rearrangement, or less frequently IGH/BCL6 translocations12. High grade B cell

lymphoma with concurrent MYC and BCL2 translocations (DHL), and those with MYC

and BCL2 double high expression (DEL) which represents a much larger group of cases,

particularly present with late clinical stage and dismal outcome. The median overall

survival time of patients with DHL/DEL ranges from less than 12 months to 21.9 months

due to extremely rapid disease progression and poor response to standard R-CHOP

regimen9,11,13,14. Actually, in the 2016 WHO revision of classification of lymphoid

Page 12: Targeting Oncogenic Protein Sythesis in MYC-driven B Cell

11

neoplasms, any high grade B cell lymphoma with MYC and BCL2 translocation,

regardless of morphology, are classified under a newly entity high-grade B-cell

lymphoma with translocations involving MYC and BCL2 or BCL6(DHL/THL)

category15(Figure 1.1). It should also be noted that indolent lymphoma particular follicular

lymphoma which carries BCL2 translocation in the first place could transform to DLBCL,

and 21% of them were found double hit.

DHL is currently the most challenging type of aggressive B cell lymphoma in clinical

practice. Firstly, FISH test for MYC break apart and BCL2 translation are required for the

diagnosis of DHL. However, not every institution performs FISH in all B cell lymphoma

cases as it will substantially increase the diagnostic cost. So far, no consensus is made on

which large B-cell lymphomas should undergo this testing, though some researchers

suggest performing FISH in GCB subtype as determined by IHC or nanostring lymph2Cx,

highly proliferative lymphomas measured by ki67 or cases that are positive for MYC

staining. Secondly, majority of DHL patients experienced disease progression after

standard R-CHOP treatment. It is clear that standard R-CHOP is not sufficient for the

induction in this type of lymphoma. BL, the disease characterized by hallmark MYC

rearrangement responses to intense chemotherapy well and usually presents good

outcome, several study groups have proposed BL like aggressive regimen for patients

with DHL16. Several intense chemotherapies were found advantageous for treating DHL

in retrospective studies. Dose-adjusted R-EPOCH (rituximab, etoposide, prednisone,

vincristine, cyclophosphamide, doxorubicin) is currently the most recognized regimen

and has been accepted as the backbone immunochemotherapy in a few clinical trials17.

Page 13: Targeting Oncogenic Protein Sythesis in MYC-driven B Cell

12

Other regimens that are initially designed for pediatric BL were found less tolerable in

elder patients including CODOX-M/IVAC (cyclophosphamide, vincristine, doxorubicin

and high-dose methotrexate/ifosfamide, etoposide and high-dose cytarabine) and

HyperCVAD/MA (rituximab, hyperfractionated cyclophosphamide, vincristine,

doxorubicin, dexamethasone, alternating with cytarabine plus methotrexate). Autologous

stem cell transplantation (ASCT) is generally recommended although how much it will

benefit patients with DHL remains controversial in different studies16,18. Despite

aggressive regimen R-EPOCH is reported leverage two-year event free survival to 67%

compared to 25% in R-CHOP treated DHL patients, DHL and DEL largely remain

incurable due to relapse and refractory. Novel strategies and therapeutic agents are

urgently in need.

Page 14: Targeting Oncogenic Protein Sythesis in MYC-driven B Cell

13

Figure 1.1. Illustration of Updated B cell Lymphoma Classification

Page 15: Targeting Oncogenic Protein Sythesis in MYC-driven B Cell

14

Chapter Two: Resolving the molecular heterogeneity of B-cell lymphoma,

unclassifiable, with features intermediate between DLBCL and BL (BCL-U)

2.1 Introduction

BL and DLBCL are two distinct entities of aggressive mature B cell lymphoma that

usually can be differentiated based on morphology, immunophenotype and genetic

features. BL typically consist of monotonous medium-sized blastic lymphoid cells that

show multiple centrally located nucleoli with clumped chromatin in round nuclei.

Tingible body macrophages containing apoptotic debris are commonly found among

rapid proliferating BL cells which leads to the ‘starry sky’ pattern. Immunochemistry

staining usually shows >95% Ki67 positive and germinal center markers including CD20,

CD10, and BCL6. In addition, MYC translocation with IGH t(8;14)(q24;q32) is required for

BL diagnosis. Despite the aggressive biological behaviors, BL is largely considered

curable. Patients with BL usually response well to intensity chemotherapy and five-year

overall survival is 70-80%. On the other hand, DLBCL is characterized by the proliferation

of heterogeneous B-cells comprising centroblastic, immunoblastic, T-cell/histiocyte-rich

and anaplastic morphological variants. Immunophenotype can either show a germinal

center or activated B cell pattern.

Although it seems these two disease can be well distinguished, a large number of cases

that contains one or more characteristics overlapping BL and DLBCL cannot be placed

either in BL or DLBCL, for example, a case morphology resembles classical BL yet has

BCL2 expression. This subgroup of non-Hodgkin lymphomas was called B-cell

Page 16: Targeting Oncogenic Protein Sythesis in MYC-driven B Cell

15

lymphoma unclassifiable, with features intermediate between BL and DLBCL (BCL-U) in

WHO 2008 revision of classification of non-Hodgkin lymphoma. Retrospectively, cases

originally diagnosed with a variety of diagnosis may fall in this category, including

Burritt-like lymphoma (BLL), atypical Burritt lymphoma, small non-cleaved B cell

lymphoma, greyzone lymphoma and high grade B cell lymphoma, NOS.

BCL-U cases are heterogeneous in both morphology and genetics. The optimal treatment

for BCL-U patients has not been established. Previous studies have reported poor

outcomes with the standard therapies used for DLBCL, as well as with more intensive

regimens19. While using a single regimen for all BCL-U cases is clearly implausible,

subgrouping BCL-U into BL and DLBCL based their biological resemblance may help to

direct an appropriate treatment plan either towards intensive chemotherapy (BL) or R-

CHOP (DLBCL).

2.2 Method and Materials

2.2.1 Experiment design

In collaboration with Brigham and Women’s hospital, we investigated a series of

difficult to classify aggressive B-cell lymphomas, in addition to typical DLBCLs and BLs,

and compared digital gene expression profiling in 2 independent sites (Boston, MA and

Omaha, NE), to the expert pathological review provided by pathologists.

2.2.2 Composition of Patient series

Page 17: Targeting Oncogenic Protein Sythesis in MYC-driven B Cell

16

A total 94 aggressive B cell lymphoma cases were collected from both sites, including

tumors with the following original pathological diagnoses: BL (14), DLBCL (30), BCL-U /

small non-cleaved, non-Burkitt (29), BL-like / Atypical BL (12), and ‘High grade NHL’ (9).

Twenty three of these tumors are genetic DHLs, combining a MYC rearrangement with

either a BCL2- (18), a BCL6- (4) rearrangement, or both (1).

2.2.3 Expert Review by Five Hematopathologists

Each hematopathologist independently reviewed H&E-stained tissue sections online

at 20x magnification, together with patient demographic information, details of

immunohistochemistry and FISH data (for MYC, BCL2, BCL6 loci, as available). For every

tumor each pathologist provided a final diagnosis, from a selection of (i) BL, (ii) DLBCL,

(iii) BCL-U, (iv) Other, or (v) Unable to classify. We interpreted a 4/5 agreement as

‘consensus classification’ for this study.

2.2.4 RNA Extraction

FFPE tissue blocks were divided into sections or cores were punched immediately

before the RNA extraction. For isolation from sections, the initial section (10 μm thick)

was discarded and three to four subsequent sections (10 μm thick) were taken. For

isolation from cores, cores (1mm in diameter) were punched from tumor rich region. Total

RNA was isolated using the Qiagen RNeasy FFPE kit (Qiagen) and quantified using a

Nanodrop spectrophotometer (Thermo Scientific). RNA was diluted to 150 to 200 ng/5 μL,

aliquoted, and stored at −80°C until use.

2.2.5 Quantitative Target Gene Expression

Page 18: Targeting Oncogenic Protein Sythesis in MYC-driven B Cell

17

For the multiplexed, digital gene expression analysis, 150 to 200 ng of RNA for each

sample was hybridized with 20 μL of reporter probes/reaction buffer and 5 μL of capture

probes at 65°C for 20 hours. The hybridized samples were then processed on the

NanoString nCounter preparation station for 2.5 hours and expression data were

subsequently generated on the NanoString nCounter digital analyzer (NanoString

Technologies) using the 600 fields of view setting over 4 hours. Total RNA from 94 cases

was extracted from FFPE tissue blocks and tested on the Nanostring platform in 2

independent laboratories, using the ‘MYCKEY2’ profiling panel and diagnostic classifier

which provides a final probability score of 0-1, as previously described20. Tumors were

classified as molecular BL (mBL; score >0.75), molecularly intermediate (m-INT; 0.25-0.75)

and molecular DLBCL (mDLBCL; <0.25).

2.3 Results

RNA samples from 94 unique ABCLs were profiled in 2 independent laboratories. The

MYCKEY diagnostic classification concurred in 92/94 (97.9%) tumors. The molecular

classifier resulted in 38 (40.4%) tumors being classified as mBL, 45 (47.8%) as mDLBCL,

and 11 (11.7%) as m-INT.

On the other hand, 66/94 (70.2%) tumors were classified by the panel of

hematopathologists with ≥ 4/5 consensus. 15/15 (100%) ‘consensus BLs’ were classified as

mBL. 27/29 (93.1%) ‘consensus DLBCLs’ were classified as mDLBCL. Two (6.9%)

‘consensus DLBCLs’ were classified as m-INT. There were 22 ‘consensus BCL-Us’, 7

Page 19: Targeting Oncogenic Protein Sythesis in MYC-driven B Cell

18

molecularly classified as mBL, 4 as m-INT, and 9 as mDLBCL. For a further 2 tumors the

molecular classification was either m-INT or mDLBCL across the 2 sites.

For 28/94 (29.8%) tumors there was a lack of consensus in pathological diagnosis. The

molecular classifier was able to classify 25/28 (89.3%) of these tumors as mBL or mDLBCL

with high probability (16 of these tumors profiled as mBL, 9 as mDLBCL, and 3 as m-INT).

Page 20: Targeting Oncogenic Protein Sythesis in MYC-driven B Cell

19

Table 2.1. Summary of results

Page 21: Targeting Oncogenic Protein Sythesis in MYC-driven B Cell

20

Figure 2.1. Results of the MYCKEY Diagnostic classifier for the Aggressive B-cell lymphoma test cohort. Tumors categorized according to the molecular diagnostic score for laboratory 1 (bar graph), the relative expression of the indicated transcripts (heat map), including the relative contribution of each to the classifier (horizontal shaded bar graphs), the MYC, BCL2 and BCL6 rearrangement status, the assigned molecular scores according to different test laboratories, and the final diagnoses by each of the 5 expert hematopathologists (tumors with ≥ 4/5 consensus are indicated with a red box).

Page 22: Targeting Oncogenic Protein Sythesis in MYC-driven B Cell

21

Figure 2.2. Diagnostic scores (0-1) for each tumor as profiled in UNMC, Omaha (x-axis) and BWH, Boston (y-axis). Spearman's rank correlation coefficient was calculated(r=0.996) with a p value<0.0001

Page 23: Targeting Oncogenic Protein Sythesis in MYC-driven B Cell

22

2.4 Discussion

The expert opinion of pathologists remains the gold standard for accurately

diagnosing aggressive B-cell lymphomas, according to WHO criteria. However, variation

in IHC staining protocol and personal subjectivity make it difficult to standardize across

institutions. Also, it is impractical to evaluate all tumors with a panel of pathologists in

routine clinical practice. In this study, we validated an established molecular classifier,

profiling RNA derived from FFPE tissue blocks, is able to accurately classify 95.5% of BL

and DLBCL, into mBL and mDLBCL groups, respectively. The classifier provides a

standardized approach, with objective outputs, to further subgroup the enigmas BCL-U,

which also demonstrated high reproducibility between independent laboratories. Herein,

we propose the MYCKEY diagnostic classifier as a highly valuable companion test, which

provides clarity of classification and tumor biology in BCL-Us, as well as other difficult to

diagnose aggressive B cell lymphomas.

However, BCL-U is a vague diagnosis as cases that fall into this category are still

heterogeneous which has little influence on deciding the appropriate regimen. Therefore,

in 2016 WHO classification, only cases with concurrent translocation of MYC and BCL2

that clearly indicate a poor prognosis and may benefit from BL like high intensive

chemotherapy are highlighted and defined as high-grade B-cell lymphoma with

rearrangements of MYC and BCL2 and/or BCL6. Nonetheless, classification of BCL-Us

that do not possess genetic rearrangements remain unclear.

Page 24: Targeting Oncogenic Protein Sythesis in MYC-driven B Cell

23

2.5 Conclusion and Pitfalls

This test could be used, in combination with standard pathological assessment, to

support clinical trials in patients with BCL-U. However, the number of BCL-U cases

included in this study is limited and some cases are still unclassifiable. Further

prospective clinical trials are needed to validate whether molecularly redefined BCL-U

would benefit from either treatment regimen and clearly indicate a choice of treatment.

Page 25: Targeting Oncogenic Protein Sythesis in MYC-driven B Cell

24

Chapter three: Targeting oncoprotein synthesis with rocaglates in MYC driven lymphoma

3.1 Introduction

Given the central role MYC plays in aggressive B cell lymphomas, several strategies

have been proposed to indirectly target this “undruggable” transcription factor.

Disrupting MYC transcription by bromodomain (BRD)-4 inhibitors have been widely

investigated in both preclinical models and clinical trials. However, early clinical data

showed BRD4 inhibitors failed to provide durable cytotoxic effects in human cancers as

single agent21. Reversal of MYC inhibition was found among relapsed patients22, and

reactivation of multiple oncogenic pathway was detected in BRD4 inhibitors resistant

cells23,24. Since transcription adaption and adaptive kinome reprogramming are often the

cause of target treatment resistance, alternative approaches for suppressing MYC or its

function are urgently needed. Augmented protein sythesis is utterly required to

accommodate the rapid proliferation in MYC driven lymphoma. Previous studies have

shown protein synthesis following MYC activation is a rate-limiting determinant of cancer

initiation25. Translation inititation is the rate limiting step of protein sythesis, which is

tightly regulated by the interactions between mRNA 5’UTR and initiation factors in a cap-

dependent manner or internal ribomse trans acting factors(ITAF) in internal ribosome

entry site (IRES)-dependent initiation. MYC as a transcription factor could directly induce

the expression of components in eukaryotic initiation factor 4F (eIF4F), activate

mammalian target of rapamycin (mTOR) pathway and susequently upregulate cap-

dependent translation initiation26. Meanwhile, targeting translation initiation also

Page 26: Targeting Oncogenic Protein Sythesis in MYC-driven B Cell

25

provides specificity mRNA with complex 5’UTR secondary structures such as cyclins and

MYC that rely on eIF4F for efficient translation could preferentially take advantage of

increased cap-dependent translation27,28, thereby forming a feed-forward loop for

oncoprotein production in MYC driven lymphomas. Therefore, strategies directed against

oncogene translation initiation may cut this oncogenic loop and create a chokepoint for

the much abundant MYC mRNA, thus haboring great theraputical potentials.

Rocaglate is a class of natural products isolated from plants of the Aglaia genus.

Rocaglate and its derivates are currently the most potent class of translation initiation

inhibitors which presents selective activity against B cells and can be well tolerated in

vivo29,30. These natural compounds and deviates have been demonstrated to potently

inhibit protein translation initiation via eIF4A31. Several rocaglates including silvestrol

and Rocaglamide showed selective inhibition upon multiple oncoproteins e.g, MYC,

MCL1, and exhibited significant cytotoxic effect on a variety of tumors in vitro and in

vivo30,32-35. The use of rocaglates for anti-cancer treatment was limited due to the scarcity

of these natural products e.g. silvestrol. In recent years, total synthesis of silvestrol was

made possible and further chemical modification and screening have unveiled a few

synthetic rocaglates that are more potent than silvestrol such as CR-1-31-B and (-)-SDS-1-

021 36-38. Furthermore, multigram scale synthesis of aglain derivatives was achieve by

using continuous flow photoreactors to perform the excited state intramolecular proton

transfer (ESIPT) [3+2]-photocycloaddition39, which unlocked the potential use of

rocaglates for industrial production and clinical applications.

Page 27: Targeting Oncogenic Protein Sythesis in MYC-driven B Cell

26

In this study, we identified translation initiation inhibitors as strong candidates for

MYC-driven B cell lymphomas, and then investigated the differential impact of mTOR

inhibitors and silvestrol on oncoprotein translation. We probed MYC translational

dependence on eIF4A1/2, and studied the mode of action of rocaglates. Moreover, we

found rocaglates potently deplete multiple B cell related oncoproteins, and demonstrated

the efficacy of the most potent rocaglate derivate (-)-SDS-1-021 in vitro and in preclinical

PDX models.

3.2 Method and Materials

3.2.1 Study Design

This study was designed to (i) identify potent small molecule inhibitors for MYC

driven lymphoma treatment. (ii) investigate the impact of rocaglates on oncoprotein

synthesis in MYC driven lymphoma, especially DHL and its underlying mechanism. (iii)

evaluate the preclinical efficacy of synthetic rocaglate (-)-SDS-1-021 in vitro and in vivo

PDX models. A cell titer blue based small molecule screen consisting of 29 FDA approved

drug and silvestrol was performed in 11 MYC driven cell lines, and an IC50 of each

compound was generated and mapped. Compounds targeting protein synthesis were

found among most potent across all cell lines. However, we found rocaglates can

efficiently repress both cap and IRES dependent translation of key oncoprotein MYC,

whereas TORKi cannot due to the abundant eIF4E in DHL. Furthermore, the abundance

of eIF4A in eIF4F complex is measured in rocaglate-treated cells and compared with that

in eIF4A knock down by siRNA, we conclude rocaglates do not repress MYC translation

Page 28: Targeting Oncogenic Protein Sythesis in MYC-driven B Cell

27

by depleting eIF4A in eIF4F complex. TMT-MS was performed to quantitatively describe

the proteome change induced by (-)-SDS-1-021. Downregulated proteins were analyzed

using online pathway enrichment database tool GSEA MSigDB and String2, and validated

by immunoblots and RT-qPCR. The effect of silvestrol and (-)-SDS-1-021 and in

combination with BCL2 inhibitor ABT199 was assessed by Prestoblue cell viability assay

and flow cytometry in vitro, and determined in two DEL/DHL preclinical xenograft mice

models.

For experiments in patient-derived mouse models, NSG mice were randomly assigned

to vehicle control and treatment groups with equal number of both genders in each group.

Group size (n=6) for tumor growth measurement was determined by power calculation

P<0.05 and an actual power>0.80 using G power 3.1(http://www.gpower.hhu.de/) based

an exploratory study in which various concentration of inhibitors were administrated to

smaller group of tumor bearing animals. Body weight, tumor volume, overall of well-

being and sign of distress was monitored every day. Humane endpoint is set at when

tumor diameter reaches 2cm or animal exhibits deteriorating body condition.

3.2.2 High-throughput small-molecule drug screen

Using a semi-automated platform as previously described40, we tested an annotated

focused library of 30 small molecules in multiple MYC-driven lymphoma cell lines. Cell

viability was measured by incubating with resazurin (R&D Systems, Minneapolis, MN,

US). Cells were seeded in 384-well plates with 2,000 cells per well in 30 μL medium, and

cultured in the presence of the different compounds at serial 3-fold diluted concentrations.

Page 29: Targeting Oncogenic Protein Sythesis in MYC-driven B Cell

28

After 3 days of treatment, 6 μL of the dye resazurin was added into each well and the

plate was incubated for 4 hours. Plates were read at a wavelength of 560/590 nm to

evaluate cell proliferation.

3.2.3 Cell culture

Dohh2, Val, Ros50, LY19, U2904, U2932 and Carnaval were purchased from DSMZ.

Namalwa, Raji, Ramos, sp53 and P493-6 were purchased from ATCC. All lymphoma cells

were cultured in RPMI 1640 medium supplemented 15% Fetal Bovine Serum (FBS) and L-

glutamine and 1% Penicillin-Streptomycin (Thermo Fisher Scientific). Isogenic BCL2

expressing BL cells were generated as previously described40. 4EBP1-4A transduced Val

and Ros50 cells and P493-6 cells were cultured in RPMI 1640 supplemented with 15% TET

free FBS and L-glutamine. To induce exogenous gene expression, 1 μg/ml doxycycline

was added to tet-on/off cells, and protein level change was detected by immunoblotting

at 48 hours. 239T and GP2-293T cells were cultured in Dulbecco’s Modified Eagle’s

Medium supplemented with 10~20% FBS. CD19+ cells were isolated from fresh healthy

human tonsil using CD19 MicroBeads (Miltenyi Biotec, Bergisch Gladbach, Germany) and

cultured in RPMI 1640 supplemented with 20% FBS, IL4 and CD40L. Primary lymphoma

cells are co-cultured with follicular fibroblasts in RPMI 1640 with 20% FBS and IL4. Patient

1 is a DLBCL case, with MYC, BCL2 and IGH complex rearrangement. Patient 2 is an ABC-

DLBCL case, with BCL2/IGH translocation, BCL6 rearrangement and MYC amplification.

Patient 3 is an ABC-DLBCL case, with MYC amplification and BCL2/IGH translation.

Patient 4 is a DLBCL case, with MYC/IGH and BCL2/IGH translocations.

Page 30: Targeting Oncogenic Protein Sythesis in MYC-driven B Cell

29

3.2.4 Protein and RNA Isolation

Cells were lysed using M-PER Mammalian Protein Extraction Reagent (Thermo Fisher

Scientific) or NP40 lysis buffer (NaCl, 150 mM, NP-40, 1.0%, Tris-Cl 50 mM pH 8.0)

supplemented with Halt Protease and Phosphatase Inhibitor Cocktail (Thermo Fisher

Scientific) for 15 or 30 mins on ice. Cell lysates were then centrifuged at 14,000g for 10

mins at 4°C. Protein concentration was measured using Pierce Bradford Protein Assay kit

(Thermo Fisher Scientific). Total RNA was isolated using the RNeasy Mini Kit (Qiagen,

Hilden, Germany) or RNAzol RT (Molecular Research Center Cincinnati, OH, USA), and

quantified by NanoDrop ND1000 spectrophotometer (Thermo Fisher Scientific).

3.2.5 Immunoblot analysis

Isolated protein was denatured for 10 mins at 70°C in NuPAGE LDS Sample Buffer

with reducing buffer (Thermo Fisher Scientific). Protein electrophoresis was performed

using Bolt 4~12% Bis-Tris Plus Gels and MES SDS running buffer (Thermo Fisher

Scientific). Proteins were then transferred onto nitrocellulose membrane, blocked with

Odyssey TBS Blocking Buffer (LI-COR, Lincoln, NE, USA), and incubated with primary

antibody overnight at 4°C. Membrane was washed and incubated with secondary

antibody for 1h at room temperature, washed again and scanned on Odyssey CLx imager

(LI-COR). Protein was quantified based on band intensity according to manufacturer’s

manual. Protein expression change was calculated by normalizing with β-actin or target

protein input. Antibodies used for immunoblotting are listed in Supplemental Table S1A.

3.2.6 Sucrose gradient fractionation

Page 31: Targeting Oncogenic Protein Sythesis in MYC-driven B Cell

30

Cells grown in T75 flask were treated with Torin1 (200nM) or silvestrol (125nM) for 2

hours. Inhibitor treated cells were incubated with cyclohexamide (100 μg/ml) for

10 minutes prior to harvesting and subsequently lysed in 600 μl 1% Triton-X100 lysis

buffer containing RNasin Plus(Promega, Madison, WI, USA) and 2 mM DTT. After

centrifugation, 500 μl supernatant was layered onto 15–45% sucrose gradients containing

10 mM HEPES, pH 7.4, 100 mM KCl, 5 mM MgCl2, 100 μg/mL of cycloheximide, 2 mM

DTT. After centrifugation at 37500 r.p.m. for 2 hours using SW55Ti rotor (Beckman),

fractions were collected with BR-188 Density Gradient Fractionation System (Brandel) and

monitored at 254 nm with Biologic LP controller (Bio-Rad). RNA in each fraction was

extracted using RNAzol RT and subsequently analyzed by qRT-PCR.

3.2.7 Quantitative real-time PCR

RNA isolated from cells, sucrose gradients and pull down elution were used for cDNA

synthesis using ProtoScript II First Strand cDNA Synthesis Kit (New England Biolabs).

mRNA expression was quantified by quantitative PCR performed on Bio-Rad CFX96

Real-Time PCR Detection System. Relative expression fold change was calculated by 2^-

ΔΔCT method. GAPDH and RPL13A were used as housekeeping gene. PCR primers used

were listed in Supplemental Table S1B.

3.2.8 Cell viability Assays

Cells were treated with translation inhibitor and/or ABT199 at several concentrations.

Cell viability was measured using Prestoblue (Thermo Fisher Scientific) at day 0, 1, 2 and

3. Relative fluorescence units was detected by Infinite M200 Pro plate reader (Tencan). To

Page 32: Targeting Oncogenic Protein Sythesis in MYC-driven B Cell

31

measure cell apoptosis, cells were stained using FITC Annexin V Apoptosis Detection Kit

(BD biosciences) and detected by flow cytometry.

3.2.9 In vitro plasmid transfection and transduction

Dual luciferase construct pRF was a kind gift from Dr. Gera (Greater Los Angeles VA

Healthcare System, USA). pURF was created by cloning MYC 5’UTR without IRES

element in front of renilla luciferase in pRF backbone. pURF-IRES was constructed based

on pURF by cloning MYC IRES element into restriction site ahead of firefly luciferase. For

transfection, equal number of 293T cells were seeded in 12 well plate. pURF and pURF-

IRES were introduced into 293T cells in the aid of TransIT-LT1 transfection reagent (Mirus

Bio LLC). Inhibitor treatment was conducted at 24h after transfection. For transduction,

pURF and pURF-MYC was modified into a retroviral expression plasmid pMIP-w

containing WRE element and puromycin resistant gene as selection marker, which was

modified from pMIG-w, a gift from Luk Parijs (Addgene plasmid # 12282). PCW57.1-

4EBP1_4xAla was a gift from David Sabatini (Addgene plasmid # 38240). Expression

plasmid was mixed with pCL-Ampho retrovirus packaging vector (Novus) or lentiviral

env plasmid pMD2.G (Addgene plasmid #12259) and lentiviral packaging plasmid

psPAX2 (Addgene plasmid#12260) in the presence of TransIT-LT1 transfection reagent,

and then were transfected into GP2-293T cells or 293T cells. Virus containing culture

medium was collected from GP2-293T or 293T cells at 48 and 72h post transfection. Virus

was concentrated using PEG-it Virus Precipitation Solution (System Biosciences) and

Page 33: Targeting Oncogenic Protein Sythesis in MYC-driven B Cell

32

resuspended with RPMI medium and Transdux (System Biosciences) to infect DHL cells.

Stable cell line was established by puromycin selection twice.

3.2.10 siRNA electroporation

Dicer-Substrate siRNA targeting eIF4A1, eIF4A2 and eIF4E were purchased from IDT.

Optimization was done for Val, Ros50 and U2904 cells using TYE 563 Transfection Control

DsiRNA according to manufacturer’s protocol. Three siRNA (20nM) targeting different

locations on the same mRNA transcript were pooled together. siRNA pools and negative

control were introduced into DHL cells by electroporation using Neon Transfection

System(Thermo Fisher Scientific). Knockdown efficiency confirmation by

immunoblotting and following experiments were performed at 48h post electroporation.

3.2.11 Dual Luciferase Assay

Stably transduced DHL cells or luciferase construct transfected 293T cell were treated

with inhibitors for 2h and then harvested in passive lysis buffer. Cell debris were spun

down and 10 μl supernatant was added into 96 well white plate for Dual-Luciferase

Reporter Assay (Promega). Luciferase activity was read by Tecan plate reader. Cell

number was used to normalize luciferase reading among independent repeats.

3.2.12 TMT-MS

Val cells were treated with SDS-1-021 (100 nM) or vehicle control for 4 hours. Proteins

were extracted using Radioimmunoprecipitation assay buffer (RIPA buffer) with

proteinase and phosphatase inhibitor cocktail. Proteins from three biological replicates

Page 34: Targeting Oncogenic Protein Sythesis in MYC-driven B Cell

33

were labelled using 6-plex TMT kit (Thermo Fisher Scientific) according to manufacturer’s

protocol and then pooled together and fractionated by high-pH reverse phase

chromatography. Dried fractions were redissolved and run by nanoLC-MS/MS using a 2-

hour gradient on a 0.075mmx250mm C18 Waters CSH column feeding into a Q-Exactive

HF mass spectrometer (Thermo Fisher Scientific).

3.2.13 Native RNA Immunoprecipitation

Native RIP was carried out as described previously41 with a few modifications. To be

brief, Torin1 (200nM) or silvestrol (125nM) was added to cells grown in T75 flask. Cells

were harvested at specific time point and lysed in PLB buffer. Normal rabbit IgG, Anti-

eIF4A2 antibody (ab31218) or Anti-eIF4A1 antibody (ab31217) were immobilized to

Dynabead Protein-G magnetic beads (Thermo Fisher Scientific) by incubation at room

temperature for 1 hour. Cell lysate supernatant was then incubated with immobilized

antibody overnight at 4°C to form pull down protein-RNA complexes. RNA was isolated

by RNAzol RT after protein K digestion of precipitated protein-RNA complexes, and

quantified by RT-qPCR.

3.2.14 m7GTP-pulldown assays

siRNA KD cells and inhibitor treated cells were solubilized in NP40 lysis buffer with

proteinase and phosphatase inhibitor cocktail. 500-1000μg total protein was incubated

with prewashed immobilized γ-aminophenyl-m7GTP agarose (Jena Bioscience) by head

to tail rotation at 4°C overnight. After washing three times using lysis buffer, 30μl LDS

loading buffer with reducing agent was added to agarose beads. Protein bound with

Page 35: Targeting Oncogenic Protein Sythesis in MYC-driven B Cell

34

agarose beads were eluted and denatured by incubation for 10 mins at 70°C, and then

diluted with lysis buffer for gel electrophoresis and immunoblotting.

3.2.15 In vivo patient derived xenograft

MYC/BCL2 double expression PDX cell is a gift from Dr. Wang Yuzhuo in Living

Tumor Laboratory. MYC/BCL2 double translocation PDX line is a gift from Dana-Farber’s

bank of patient-derived tumor xenografts (Dana-Farber). 106~5x106 second passage PDX

cells (LTL025/DFBL20954) were inoculated to the flank of 6-8 week old male and female

NSG mice (JAX lab). After tumors reached palpable size, tumor bearing mice were

separated into vehicle ctrl group (5.2%Tween 80/5.2% PEG400 in water) and SDS group

(0.2mg/kg). Additional ABT group (1 mg/kg) and combined group (SDS

0.2mg/kg+ABT199 1mg/kg) were added for DFBL20954 PDX model. Each group contains

six mice. Drug or vehicle was given i.p daily for 10 consecutive days. Animals were

weighted and tumor size was measured with caliper daily. Tumor volume was calculated

as V = (W2 × L)/2. At day 10 or endpoint, mice were euthanized and tumor was isolated,

weighed and pictures were taken. Trimmed tumor was fixed in formaldehyde for 24 hours

and then embedded in paraffin. Slides were cut from tumor FFPE blocks, H&E, C-myc

and Ki67 IHC staining was performed at UNMC tissue science facility.

3.2.16 Study ethics approval

Human tissues was collected or transferred under a tissue banking and utilization

protocol approved and supervised by University of Nebraska Medical Center

Institutional Review Board. Animal study was conducted in accordance with an approved

Page 36: Targeting Oncogenic Protein Sythesis in MYC-driven B Cell

35

protocol of University of Nebraska Medical Center Institute Animal Care and Use

Committee.

3.2.17 Statistics and data analysis

Data were analyzed using Prism 5 program (GraphPad Software) and Prism online

tools. Data shown with the mean ± SD are from at least 3 independent experiments.

Unpaired t-test was used to compare data from two independent groups. One-way

analysis of variance (ANOVA) was used to compare data from three or more independent

groups. P values of less than 0.05 were considered significant.

TMT-MS samples were processed using Proteome Discoverer 2.2 software (Thermo

Fisher Scientific) linked to the Mascot Server MSMS search software (Matrix Science,

London, UK; version 2.6.1). Mascot was set up to search the common contaminants

database cRAP_20150130.fasta (117 entries) and the uniprot-human_20180625 database

(72792 entries). Mascot was searched with a fragment ion mass tolerance of 0.06 Da and a

parent ion tolerance of 10.0 PPM. TMT and carbamidomethyl were specified as fixed

modification. Oxidation of methionine was specified in Mascot as a variable modification.

The data was searched using a decoy database to set the false discovery rate to 1% (high

confidence) and 5% (medium confidence). The protein quantification was processed using

Proteome Discoverer (Thermo Fisher Scientific, version 2.2). The co-isolation threshold

was set to 50%, and the average S/N to 10. A total of 6385 proteins were identified and

quantified. 177 proteins depleted over 20% were submitted for pathway enrichment

Page 37: Targeting Oncogenic Protein Sythesis in MYC-driven B Cell

36

analysis on String website (https://string-db.org/) and GSEA/MSigDB database

(http://software.broadinstitute.org/gsea/msigdb).

Drug combination index (CI) was calculated using Compusyn which based on Chou-

Talalay method for drug combination42. CI offers quantitative definition for additive effect

(CI = 1), synergism (CI < 1), and antagonism (CI > 1).

Percentage of positive cell was measured by counting positive cells vs negative cell in

5 high power (400x) fields.

Page 38: Targeting Oncogenic Protein Sythesis in MYC-driven B Cell

37

Table S1A. List of antibodies used for immunoblots

Target protein Manufacturer Cat# Dilution

eIF4A2 Novus Biologicals NBP2-24612SS 1:1000

eIF4A1 Abcam ab31217 1:1000

β-Actin Santa Cruz sc-1616 1:500

MCL1 Santa Cruz sc-819 1:500

eIF4G1 Santa Cruz sc-373892 1:500

BCL2 Santa Cruz sc-492 1:500

MYC CST 5605 1:1000

eIF4E CST 9742 1:1000

4EBP1 CST 6944 1:1000

p-4EBP1(T37/46) CST 2855 1:1000

RPS6 CST 2217 1:1000

p-RPS6(S235/236) CST 4858 1:1000

PARP CST 9542 1:1000

Vinculin CST 13901 1:1000

Page 39: Targeting Oncogenic Protein Sythesis in MYC-driven B Cell

38

Table S1B. List of qRT primers

Target Forward Reverse

AURKA GGAATATGCACCACTTGGAACA TAAGACAGGGCATTTGCCAAT

AURKB CGCAGAGAGATCGAAATCCAG AGATCCTCCTCCGGTCATAAAA

BCL2 GGCTGGGATGCCTTTGTG GCCAGGAGAAATCAAACAGAGG

BCL6 GGAGTCGAGACATCTTGACTGA ATGAGGACCGTTTTATGGGCT

eIF4A1 ATGGCACTAGGAGACTACATGG CCACGGCTTAACATTTCGTCA

eIF4A2 GAAGCCTTCCGCTATTCAGCA CTTGGGTCTCCTTGAACTCAATC

MYC GGTGCTCCATGAGGAGACA CCTGCCTCTTTTCCACAGAA

NEK2 GATGACTCAGTTGACTCTGGC TCCCGATGCAATACGGTATGA

PLK1 CTCAACACGCCTCATCCTC GTGCTCGCTCATGTAATTGC

TCF3 CCGACTCCTACAGTGGGCTA CGCTGACGTGTTCTCCTCG

MCL1 GGACATCAAAAACGAAGACG GCAGCTTTCTTGGTTTATGG

GAPDH CCACTCCTCCACCTTTGAC ACCCTGTTGCTGTAGCCA

NDUFS7 GAGGTGTCCATCAGAGCGTG AGCTTGGCCACCACATACTC

Page 40: Targeting Oncogenic Protein Sythesis in MYC-driven B Cell

39

3.3 Results

3.3.1 Inhibitors targeting protein synthesis exhibit high potency in MYC driven

lymphomas

To identify the candidate drug for MYC driven lymphoma treatment, drug screen

comprising of silvestrol and 29 FDA approved small molecule inhibitors targeting

variously B cell lymphoma oncogenic pathways was performed in 11 aggressive MYC

driven B cell lymphoma cell lines, including BL cell line Namalwa, Raji and Ramos and

those with isogenic BCL2 expression, DHL cell line CJ, Val and Dohh2, DEL cell line

U2932, and mantle cell lymphoma cell line Sp53. Translation initiation inhibitor silvestrol,

PI3K inhibitor PIK75, mTOR inhibitor AZD80555 and BEZ235 and PLK1 inhibitor

Volasertib exhibited highest potency across all MYC driven lymphomas tested (Figure

3.1A). We validated that both AZD8055 and silvestrol inhibit DHL cell proliferation at a

dose dependent manner, although all three DHL cell lines are more sensitive to silvestrol

(Figure 3.1B). Based on these findings and the role of MYC in coordinating transcription

and translation, targeting mRNA translation initiation especially by silvestrol seems very

promising in MYC driven lymphomas.

Page 41: Targeting Oncogenic Protein Sythesis in MYC-driven B Cell

40

Figure 3.1 Inhibitors targeting protein synthesis exhibit highest potency in MYC driven lymphomas

(A) Functional drug screens in MYC driven lymphomas (Namalwa, Raji, Ramos, Namalwa-BCL2, Raji-BCL2, Ramos BCL2, CJ, Val, Dohh2, U2932 and Sp53). Small molecules inhibitors and corresponding targets are clustered according biology function and represented in an IC50 (μM) heatmap format. (B) mTOR inhibitor AZD8055 and silvestrol decrease DHL cell viability. DHL cell Dohh2, Val and Ros50 are treated with AZD8055 or silvestrol at various concerntrations. Cell viability is determined by prestoblue luciferase unit (PFU) at 48 hours. Data presented show the mean ± SD of at least 3 independent experiments.

Page 42: Targeting Oncogenic Protein Sythesis in MYC-driven B Cell

41

3.3.2 Rocaglate but not mTOR inhibitors efficiently inhibit MYC expression

The mTOR pathway is the sensor of nutrition and master regulator of growth in

eukaryotic cells. mTORC1, the better characterized one of the two mTOR complexes,

directly phosphorylates S6 kinase 1 (S6K1) and the translation regulator eukaryotic

translation initiation factor 4E (eIF4E) binding protein 1 (4EBP1). Phosphorylated 4EBP1

then dissociates from eIF4E, which frees eIF4E for the binding to mRNA 5’ cap. eIF4E

recruits the scaffold protein eIF4G which together with dead box helicase eIF4A and other

initiation factors forms the eIF4F complex. Therefore, mTOR inhibitors may actively

inhibit cap-dependent translation, and subsequently decrease oncoproteins that depend

on 5’cap for efficient translation. To evaluate the inhibition of mTOR inhibitors on mRNA

translation in double hit lymphoma cell lines, Val and Ros50 cells were treated with mTOR

inhibitors Torin1, temsirolimus and AZD8055 for 24 hours. As expected, second

generation ATP competitive mTOR inhibitor Torin1 and AZD8055 decreased

phosphorylated 4EBP1 and phosphorylated ribosomal protein S6 (RPS6), the substrate of

S6K1. First generation rapamycin analog temsirolimus decreased phosphorylated RPS6

but had no effect on 4EBP1. Surprisingly, ATP competitive mTOR inhibitors remarkably

reduced MCL1, but only marginally decreased MYC in Val and Ros50 while not affecting

BCL2 at all (Figure 3.2A). Similar results were observed in P493-6 cells, a Burkitt

lymphoma cell line in which expression of MYC is controlled by a tetracycline promoter,

where Torin1 again failed to repress MYC expression (Figure 3.2C). mTOR inhibitors did

not induce much cell apoptosis as indicated by cleaved PARP. MCL1 has very short half-

life therefore any subtle change on protein synthesis or degradation would greatly affect

Page 43: Targeting Oncogenic Protein Sythesis in MYC-driven B Cell

42

MCL1 level. MYC and BCL2 mRNA contain long and complex 5’UTR that were thought

to heavily rely on cap-dependent translation actually do not respond to mTOR inhibition

very well in MYC driven lymphomas.

mTOR inhibitors repress cap-dependent translation by blocking 4EBP1

phosphorylation, therefore insufficient endogenous 4EBP1 may not fully sequester eIF4E

from eIF4F complex. To rule out this possibility, we introduced a doxycycline-inducible

allele of 4EBP1 that carries alanine substitutions at four serine/threonine residues (4EBP1-

4A) into DHL cells (Figure 3.2B). These four residues are the targets of mTORC1 and can

otherwise be phosphorylated. As a result, 4EBP1-4A constitutively binds to eIF4E

regardless of mTORC1 activity and acts as a dominant cap-dependent translation

initiation inhibitor43. Upon ectopic expression of 4EBP1-4A, MYC expression was only

marginally decreased (Figure 3.2B), implying either eIF4E is still relatively abundant or

eIF4E is dispensable for MYC translation. To figure out whether MYC translation is

dependent on eIF4E, we next disrupted eIF4E by siRNA knockdown. eIF4E knockdown

showed a significant depletionof eIF4E at 48 hours following siRNA electroporation, and

MYC expression was remarkably decreased, which validated eIF4E is crucial for MYC

mRNA translation (Figure 3.2C). Briciclib binds to eIF4E and was reported to block cap-

dependent translation of cyclin D1, MYC and VEGF44. The small molecule 4EGI-1

pharmacologically mimics 4E-BP function and inhibits mRNA cap-dependent translation

by competing for eIF4E/eIF4G interaction45. By contrast, an insufficient inhibition by eIF4E

inhibitor Briciclib or 4EG-I, similar to TORKi, was unable to efficiently deplete MYC

expression (Figure 3.3A). These findings argue eIF4E is excessively abundant in MYC

Page 44: Targeting Oncogenic Protein Sythesis in MYC-driven B Cell

43

driven lymphomas that even increasing 4EBP1 ectopically is not able to reach the point

where MYC can be efficiently inhibited. The level of eIF4E is usually associated with the

demand for protein synthesis and cell proliferation. Lymph node, especially germinal

center presents a high expression of eIF4E compared to other tissue. Further

overexpression of eIF4E is commonly found in B cell malignancies and associated with

agressiveness46. Therefore, although MYC depends on eIF4E for cap-dependent

translation initiation, the inhibitory effect of 4EBP1 and thus mTOR inhibitors could be

very limited in DHL due to the abundance of eIF4E.

In contrast, silvestrol treatment dramatically decreased MYC expression in DHL and

DEL cells (Figure 3.2D-E) at 5 nM, and abolished MYC expression at higher biologically

relevant concentrations. Moreover, we tested silvestrol in P493-6 cells in which MYC

expression is tetracycline-repressible, and found low concentration of silvestrol was able

to abrogate MYC expression comparable to doxycycline (Figure 3.3C). Furthermore,

silvestrol exhibited more potent inhibition of MCL1 expression compared to TORKi, yet

neither significantly decreased BCL2 in DHL cells (Figure 3.2A and 3.2D). The findings

are consistent with the fact that MCL1 has a short half-life and as such any subtle change

in protein synthesis or degradation would greatly affect the protein level, whereas the

BCL2 protein is stable with a half-life of approximate 20 hours47,48. Taken together, we

demonstrated that silvestrol potently repressed MYC expression in all MYC deregulated

B cell lymphoma lines; however the inhibitory effect of mTOR inhibitors was

compromised due to eIF4E overexpression.

Page 45: Targeting Oncogenic Protein Sythesis in MYC-driven B Cell

44

Figure 3.2. Rocaglates but not mTOR inhibitors efficiently repress MYC expression.

(A) Effect of mTOR inhibitors on oncoprotein MYC, MCL1 and BCL2. Val and Ros50 cells treated with Torin1, temsirolimus (TEM) and AZD8055 at indicated concentrations for 24 hours. Protein levels were examined by immunoblotting. (B) 4EBP1-4A was introduced into Val and Ros50 cells by retrovirus infection and the ectopic expression was induced by doxycycline (1 μg/ml) for 48 hours. (C) siRNA targeting eIF4E was transfected into Val and Ros50 cells by electroporation. MYC protein level was determined at 48 hours post-transfection. DHL cell lines Val and Ros50 (D), and DEL cell lines U2932 and LY19(E) were treated with increasing concentrations of silvestrol for 24 hours. MYC, MCL1, BCL2 and PARP were measured by immunoblotting.

Page 46: Targeting Oncogenic Protein Sythesis in MYC-driven B Cell

45

Figure 3.3. Impact of translation inhibitors on oncoprotein expression in MYC driven lymphomas. (A) Impact of eIF4E inhibitor briciclib on MYC protein. Val and Ros50 cells were treated with increasing concentrations of briciclib for 24h. (B) Impact of eIF4E-eIF4G interaction inhibitor 4EG-I on MYC protein. Val and Ros50 cells were treated with increasing concentrations of 4EG-I for 24h. (C) Assessment of MYC protein level in Torin1 and silvestrol-treated P493-6 cells. P493-6 cells were treated Torin1, silvestrol, doxycycline or vehicle control for 24h. Data shown are representative of at least 3 independent experiments.

Page 47: Targeting Oncogenic Protein Sythesis in MYC-driven B Cell

46

3.3.4 silvestrol strongly inhibits cap-dependent translation of MYC

To provide insight into the differential impact of Torin1 and silvestrol on oncogene

mRNA translation, we performed polysome fractionation to analyze mRNA distribution

on ribosomes. Polysome fractionation separates mRNA based on the number of ribosomes

bound. Generally, efficiently translated mRNA are associated with multiple ribosomes

(polysome) and appear in heavier factions(7-9), while poorly translated mRNA are

associated with much less ribosomes (monosomes and lighter polysomes) and found in

lighter fractions(4-6). To elucidate the immediate translational change caused by

inhibitors, DHL cells were treated for 2 hours with DMSO control, Torin 1 or silvestrol.

Cells were incubated with cycloheximide and then sucrose density gradient

centrifugation was performed. A total of nine fractions were collected at one fraction per

minute. The abundance of mRNA in fraction 3-9 was measured by real-time reverse

transcription polymerase chain reaction (qRT-PCR), and normalized to total input. We

found silvestrol had stronger overall translational inhibition than Torin1 in both DHL cell

lines as indicated by an elevation of monosome peaks and reduced polysome peaks

(Figure 3.4A). Moreover, MYC mRNA was redistributed from actively translating

polysomes to monosomes in silvestrol-treated cells but not in Torin1 treated ones. Both

inhibitors induced MCL1 mRNA redistribution towards monosomes although the impact

of silvestrol seemed higher. BCL2 mRNA enrichment in fractions was not noticeably

altered by either inhibitor (Figure 3.4B). These observations confirmed our findings from

immunoblotting, rocaglates efficiently repress MYC translation while mTOR inhibition

cannot.

Page 48: Targeting Oncogenic Protein Sythesis in MYC-driven B Cell

47

MYC mRNA contains an IRES element in 5’UTR, which is capable of initiating

translation by directly recruiting ribosome independent of 5’cap structure. IRES

translation may be activated to maintain MYC protein expression under stress eg.

translation inhibition. To measure MYC cap-dependent and IRES translation activity

respectively, we generated two dual luciferase reporter vectors pURF and pURF-IRES

(Figure 3.4C). The translation of renilla luciferase is regulated by MYC 5’UTR without the

IRES element, meanwhile translation of firefly luciferase is driven by MYC IRES. silvestrol

and Torin1 were added to 293T cells transfected with reporter constructs. After 4 hours

incubation, luciferase activity were measured and normalized to cell number. Although

both inhibitors were able to decrease IRES and cap-dependent translation, silvestrol

displayed much stronger inhibition especially on cap-dependent translation (Figure

3.4D). Next, we stably transduced DHL cells with dual luciferase constructs using

retrovirus. Similar to our observation in 293T cells, silvestrol showed stronger inhibition

than Torin1 in DHL cells, especially on cap-dependent translation. silvestrol reduced

~31% firefly and ~55% renilla luciferase activity in Val, while Torin1 only decreased ~13%

and ~21% respectively. In reporter construct transduced Ros50 cells, silvestrol showed

~30% reduction of firefly and ~44% of renilla luciferase activity, compared to ~14% and

~16% in Torin1 treated cells (Figure 3.4E). Interestingly, 200nM Torin1 and 25nM silvestrol

treated cells exhibited comparable reduction of firefly luciferase activity yet silvestrol

decreased 20% more renilla luciferase activity. While 25nM silvestrol treatment nearly

depleted MYC protein, cells treated with 200nM Torin1 only showed a marginal decrease

(Figure 3.2A, Figure 3.3A). These findings argue the strong translational inhibition on

Page 49: Targeting Oncogenic Protein Sythesis in MYC-driven B Cell

48

MYC by rocaglates is mainly facilitated through cap-dependent translation repression,

although rocaglates suppress both IRES and cap-dependent translation.

Page 50: Targeting Oncogenic Protein Sythesis in MYC-driven B Cell

49

Figure 3.4. silvestrol strongly inhibits cap-dependent and IRES translation of MYC

(A) Polysome fractionation of Val and Ros50 cell, treated with Torin1 (200nM), silvestrol (125nM) or solvent control for 2 hours. Collection is set at one fraction per minute. Data shown is representative of 3 independent experiments. (B) Distributes MYC, MCL1, BCL2 mRNA in polysome and monosome fractions. Relative mRNA content per fraction is measured by RT-qPCR. Data presented show the mean ± SD of 3 independent experiments. (C) Schematic illustration describing luciferase reporters used in this study. pURF contains MYC 5’UTR without IRES element inserted proximal to renilla luciferase coding sequence. pURF-IRES is generated by inserting MYC IRES element before firefly luciferase coding sequence in pURF construct. (D) 293T cells transfected with pURF and pURF-IRES are treated with Torin1 and silvestrol for 4 hours. Luciferase activity is measured and normalized to cell number (per 106 cells). (E) Val and Ros50 cells transduced with pURF-MYC are treated with Torin1 and silvestrol for 4 hours. Luciferase activity is measured and normalized to cell number and that of vehicle control. Data presented in D and E show the mean ± SD of at least 3 independent experiments.

Page 51: Targeting Oncogenic Protein Sythesis in MYC-driven B Cell

50

Page 52: Targeting Oncogenic Protein Sythesis in MYC-driven B Cell

51

3.3.5 Rocaglates overcome eIF4A abundance

Previous studies have disclosed rocaglates as selective inhibitors of eIF4A through

diminishing its availability in eIF4F complex35,49-51, however recent evidence demonstrated

that rocaglates directly bind at the interface formed between eIF4A and polypurine

sequences on targeted mRNA, forming a stable structure to prevent ribosome

scanning52,53. Herein, we were interested to determine through which mechanism

rocaglates achieved efficient MYC repression in DHL/DEL. First we probed the expression

of eIF4A in DHL/DEL cells. The eIF4A family consists of three helicases in which eIF4A1

and eIF4A2 share high resemblance and were reported to interact with silvestrol and

episilvestrol54, whereas eIF4A3 is distant from these isoforms in both amino acid sequence

and function55. We found that eIF4A1 but not eIF4A2 is highly up-regulated in DHL/DEL

cells as compared to normal B cells (Figure 3.5A). Consistently, eIF4A1 mRNA was found

to be much more abundant than eIF4A2, ranging from 2.11 fold in Carnaval to 9.63 fold

in Val (Figure 3.5B), and knockdown of eIF4A1, but not eIF4A2, had significant impact on

cell viability (Figure 3.5C). These data are in accordance with the common notion that

eIF4A1 is the major helicase required in cancers.

Page 53: Targeting Oncogenic Protein Sythesis in MYC-driven B Cell

52

Figure 3.5. Expression of eIF4A1/2 and eIF4A knockdown. (A) Protein expression of initiation factor eIF4A1, eIF4A2, eIF4E, eIF4G, and MYC in normal CD19+ B and DHL/DEL cells. (B) Ratio of eIF4A1 to eIF4A2 mRNA expression in CD19+ normal B and DHL/DEL cells. (C) Impact of eIF4A1, eIF4A2 or combined eIF4A1/2 knockdown on cell proliferation. Data shown in A are representative of at least 3 independent experiments. Data presented in B and C show the mean ± SD of 3 independent experiments.

Page 54: Targeting Oncogenic Protein Sythesis in MYC-driven B Cell

53

Next, we compared the impact of eIF4A knockdown (Figure 3.6A) versus rocaglate

treatment (Figure 3.6B) in two aspects, eIF4A abundance in eIF4F and MYC expression.

We successfully introduced siRNA into DHL cells via electroporation and knockdown

efficiency reached over 60% in both cell lines. Following siRNA-mediated eIF4A1

knockdown, consistent with previous reports34,56, we observed a marked increase of

eIF4A2 expression while eIF4A1 showed no changes to eIF4A2 depletion Interestingly,

Val and Ros50 responded differently to eIF4A knockdown (Figure 3.6C). m7GTP pull-

down assay confirmed depletion of eIF4A1 and eIF4A2 in the eIF4F complex in single

knockdown cells. eIF4A1/2 double knockdown significantly decreased eIF4A abundance

in eIF4F complex in Ros50, however such reduction is not as prominent in Val (Figure

3.6A), as reduced eIF4A may still be more abundant than the relatively low level of eIF4E

in this cell line (Figure 3.5A). Notably, Val cells transfected with negative control siRNA

showed similar amount of eIF4A2 in cap pull down to that in eIF4A2 knockdown,

suggesting eIF4A1 predominately occupies eIF4E in since eIF4A1 is far more abundant

than eIF4A2 in this cell line. Cell viability assay confirmed that Ros50 is more vulnerable

to eIF4A depletion than Val (Figure 3.5C). We then analyzed MYC expression upon eIF4A

knockdown and were surprised to find that the MYC protein level was only marginally

decreased in these cell lines (Figure 3.6C). eIF4A2 knockdown actually increased MYC

expression in Val, which is likely due to the upregulation of eIF4E(Figure 3.6C, D).

Previous studies demonstrated eIF4A2 is required for miRNA mediated gene

downregulation57, we speculate eIF4A2 may play different roles based on its abundance

in eIF4F complex, although further study is needed. Dual luciferase assay revealed that

Page 55: Targeting Oncogenic Protein Sythesis in MYC-driven B Cell

54

eIF4A knockdown failed to induce comparable reduction of luciferase activity (Figure

3.6D) to silvestrol treatment as shown in Figure 3.4E. These findings suggest that eIF4A1

is highly abundant in MYC-driven lymphoma wherein even up to 70% depletion had

minimal effect on MYC translation. In contrast, the abundance of eIF4A1 and eIF4A2 in

the eIF4F complex was barely affected by a high concentration of silvestrol or (-)-SDS-1-

021, a synthetic rocaglate that is more potent as a translation inhibitor than silvestrol30,58

(Figure 3.6B and 3.7). Unlike eIF4A knockdown, rocaglates efficiently repressed MYC at

a low concentration (Figure 3.2D), and had no additive effect with eIF4A knockdown in

repressing MYC (Figure 3.6C). In addition, rocaglates appeared to be much more effective

on MYC repression than hippuristanol, an ATP-competitive inhibitor of eIF4A which

impairs its helicase function (Figure 3.6E). These findings suggest that the potent MYC

suppression by rocaglates is unlikely a consequence of eIF4A depletion in eIF4F complex

in MYC-driven lymphoma cells.

Page 56: Targeting Oncogenic Protein Sythesis in MYC-driven B Cell

55

Figure 3.6. Rocaglates increase MYC mRNA binding to eIF4A, instead of depleting eIF4A in eIF4F.

(A)Evaluation of total eIF4A1 and eIF4A2 as well as their abundance in eIF4F following siRNA knockdown in Val and Ros50 cells. Knockdown efficiency indicated in lane. (B) Evaluation of eIF4A1 and eIF4A2 abundance in eIF4F after 4 hours treatment with 125nM silvestrol (Silv), 100 nM (-)-SDS-1-021(SDS) or vehicle control in Val and Ros50 cells. (C) Alterations of MYC expression after eIF4A1, eIF4A2 or combined knockdown, and with silvestrol added. siRNA transfected Val and Ros50 cells were incubated with vehicle control or silvestrol (125 nM) for 4 hours. (D) Dual luciferase assay performed in eIF4A knock down cells. Luciferase activity measured at 48 hours post-transfection and normalized to cell number. (E) eIF4A inhibitor hippuristanol (Hipp) and (-)-SDS-1-021 (SDS)(24 hours) induced reduction of MYC protein. (F) Native RNA immunoprecipitation

Page 57: Targeting Oncogenic Protein Sythesis in MYC-driven B Cell

56

measuring MYC mRNA enrichment on eIF4A1 and eIF4A2. Data shown in A, B, C and E are representative of at least 3 independent experiments. Data presented in D and F show the mean ± SD of at least 3 independent experiments.

Page 58: Targeting Oncogenic Protein Sythesis in MYC-driven B Cell

57

Finally, to test whether rocaglates directly alter eIF4A and MYC mRNA binding, we

conducted native RNA immunoprecipitation (RIP). We found MYC mRNA was

significantly enriched on eIF4A1 and eIF4A2 upon rocaglate treatment (Figure 3.6F),

while the previously described rocaglate-insensitive mRNA NDUFS7 was not53 (Figure

3.7), suggesting that the rocaglate selectively stabilized the eIF4A-MYC mRNA interface.

It has been shown that the eIF4A-mRNA-rocaglate structure is very stable that may persist

even after ATP hydrolysis, therefore directly prevents ribosome scanning in translation

initiation 52,53. Collectively, we demonstrated that eIF4A exists in excess in MYC-driven

lymphoma cells, and that moderate depletion is insufficient to repress MYC translation.

Apparently, rocaglates efficiently repress MYC translation by increasing MYC mRNA

binding to eIF4A, instead of depleting it in eIF4F complex, and therefore overcome eIF4A

abundance.

Page 59: Targeting Oncogenic Protein Sythesis in MYC-driven B Cell

58

Figure 3.7. silvestrol increases rocaglate target mRNA enrichment on eIF4A1/2

Enrichment of target mRNA MYC, NEK2, TCF3, BCL6, PLK1, AURKA, AURKB and negative control NDUSFS7 on eIF4A1 (A) and eIF4A2(B). Val and Ros50 cells were treated with 125nM silvestrol (Silv), 200nM Torin1 or vehicle control for 2 hours. eIF4A1/2 binding mRNA was isolated by native RNA immunoprecipitation and subsequently quantified by RT-qPCR. Data presented show the mean ± SD of 3 independent experiments.

Page 60: Targeting Oncogenic Protein Sythesis in MYC-driven B Cell

59

3.3.6 Rocaglates repress multiple critical oncoprotein expression in DHL

To better characterize translation inhibitory effect of rocaglate in DHL, we performed

6-plex tandem mass tag based mass spectrometry (TMT-MS) to quantitatively describe (-

)-SDS-1-021 induced proteome change. To elucidate primary immediate effect of

translation inhibition caused by (-)-SDS-1-021, Val cells were treated with 100nM (-)-SDS-

1-021 or DMSO ctrl for 4 hours. Total protein from three biological replicates was isobaric

tagging labelled, fractionated and detected by nanoLC-MS/MS. 6385 proteins were

identified and quantified(FDR<5%), of which 177 were found depleted over

20%(p<0.05)(Figure 3.8A). Proteins that have been validated to play crucial roles in B cell

lymphoma biology among top 200 depleted are highlighted (Figure 3.8B, Figure 3.9A).

Among the most depleted proteins, several critical oncoproteins were found including

MYC, MCL1 and cyclins, which is consistent with previous reports in other cancers30,35.

We also identify NEK2, AURKA, AURKB, PLK1, WEE1 and CHEK1 which are kinases

within MYC regulation network. NEK2 is a mitotic regulator which prevents B cell

maturation when overexpressed59,60. AURKA and AURAKB are essential in mitosis and

required for the maintenance of Myc-driven B-cell lymphoma61. CHK1 and WEE1 are key

regulators of DNA damage surveillance pathways. Inhibition of CHK1 and WEE1 were

reported to create synergetic killing effect on mantle cell lymphoma and DLBCL62,63. PLK1

stabilizes MYC by inducing autoubiquitylation and proteasome degradation of the E3

ubiquitin ligase FBW764. MYC in turn activates PLK1 transcription, forming a feed-

forward loop in DHL40. In addition, multiple B cell related transcription factors were also

significantly decreased, including EST1, TCF3, BCL6 and NOTCH1. ETS1 promotes

Page 61: Targeting Oncogenic Protein Sythesis in MYC-driven B Cell

60

premature B cell differentiation into antibody-secreting cells, and is frequently

dysregulated in DLBCL65. TCF3 is constitutively active in BL which activates antigen-

independent B-cell-receptor (BCR) signaling for cell survival66. NOTCH1 cooperates MYC

and plays a oncogenic role in lymphoid malignancies67. BCL6 is a transcriptional repressor

that regulates germinal center formation, which is frequently activated in the

pathogenesis of B cell lymphoma derived from germinal center68. qRT-PCR results

demonstrated mRNA expression of MYC, AURAKA, AURAKB, PLK1, NEK2, and MCL1

were upregulated by (-)-SDS-1-021 treatment, whereas expression of TCF3 is remarkably

decreased and BCL6 unchanged (Figure 3.8C). These observations argue the depletion of

these proteins are mainly through translational inhibition. Pathway enrichment analysis

confirmed cell cycle and PLK1 pathway activity were mostly involved (Figure 3.7D,

Figure 3.8B). Immunoblotting validated MYC, AURAKA, PLK1, NEK2, TCF3, BCL6, and

MCL1 were uniformly depleted in four DHL cell lines by silvestrol and (-)-SDS-1-021 at

relatively low concentration (25nM) (Figure 3.8E). On the other hand, Torin1 (200nM) was

able to moderately decrease NEK2, MCL1, PLK1, AURKA but increase TCF3, and results

vary among cell lines (Figure 3.9C). Overall, these findings indicate rocaglates not only

directly inhibit MYC translation but also disrupt MYC regulation network. Rocaglates

also strongly deplete multiple critical B cell lymphoma related oncoproteins besides MYC

in including transcription factors that are generally hard to target.

Page 62: Targeting Oncogenic Protein Sythesis in MYC-driven B Cell

61

Figure 3.8. Rocaglates repress multiple critical oncoproteins in DHL

(A) Quantitative proteomics demonstrating the impact of (-)-SDS-1-021 treatment (100 nM) relative to vehicle control in Val cells after 4 hour incubation (three biological replicates TMT labelled). 6385 proteins were identified, of which 177 were significantly depleted for more than 20% by (-)-SDS-1-021 (P < 0.05). Several critical oncoprotein in B cell lymphoma are highlighted in red including NEK2, MCL1, MYC, TCF3, PLK1 and AURKA. (B) Change of B cell lymphoma related oncoproteins are presented in heatmap format (log2 transformation of relative abundance ratio). (C) mRNA expression change induced by rocaglates. Val cells were treated with 125nM silvestrol(Silv), 100nM (-)-SDS-1-021(SDS) or vehicle control for 4 hours. Relative expression level to control of MYC, AURKA, AURKB, PLK1, NEK2, TCF3, BCL6 and MCL1 are measured by RT-qPCR. (D) The 177 depleted proteins were analyzed in MSigDB C2 canonical pathway database, indicating an enrichment of cycle and PLK1 pathway. (E) Validation of oncoproteins identified in TMT-MS by immunoblot. Val, Ros50, U2904, Carnaval cells were treated with silvestrol (25nM) or (-)-SDS-1-021(25nM) or vehicle control for 24 hours.

Page 63: Targeting Oncogenic Protein Sythesis in MYC-driven B Cell

62

Figure 3.9. Downregulated proteins pathway enrichment analysis and target validation in Torin1 treatment

(A) Top 200 downregulated proteins by (-)-SDS-1-021 in Val cells. Data from three biological replicates is shown in heatmap format (log2 transformed relative abundance ratio). (B) Go pathway enrichment analysis of most downregulated proteins. Top 15 categories of GO biological process and cellular components, and top 5 GO molecular function are shown and ranked according –log (FDR). (C) Change of oncoprotein MYC, AURKA PLK1, NEK2, TCF3, BCL6, MCL1 and BCL2 by Torin treatment. Val, Ros50, U2904 and Carnaval were treated with 200nM Torin1 for 24 hours. Data shown are representative of at least 3 independent experiments.

Page 64: Targeting Oncogenic Protein Sythesis in MYC-driven B Cell

63

3.3.7 Rocaglates synergistically induce cell death with ABT199 but spare normal B

cells

BCL2 belongs to a large family that regulates cell apoptosis by altering mitochondria

membrane permeability. This anti-apoptotic protein is excessively expressed in DHL cells

due to chromosome t(14,18) translocation. Enforced BCL2 expression relieves cells from

the pro-apoptotic stress caused by MYC driven proliferation69. MYC and BCL2 are the two

major driven oncogenes in DHL as both are highly overexpressed due to primary genetic

abnormalities. DHL cells are addicted to MYC as well as BCL2 since these cells are primed

to such genetic background during pathogenesis. ABT199 is a novel selective small-

molecule inhibitor of BCL2 that has been shown effective in B cell malignancies in

multiple pre-clinical models and clinical trials69. Indeed, our drug screen showed DHL

and DEL lines are sensitive to ABT199 whereas BL lines with isogenic BCL2 expression

are not. Also, we previously demonstrated DHL cells specifically depend on BCL2 for

survival but not on other anti-apoptotic proteins in this family, and ABT199 resistant DHL

cells exhibited adaptive reliance on MCL140. As inhibition of BCL2 mRNA translation

cannot be achieved with either mTOR inhibitors or rocaglates (Figure 3.2A, 3.3A), we

proposed the addition of ABT199 could synergize with rocaglates for an even more

efficient tumor cell killing. To validate the synergy between ABT199 and rocalgates, Val

and Ros50 cells were treated with silvestrol, ABT199 or combined at various

concentrations for 48 hours. Cell apoptosis was detected by flow cytometry with FITC-

AnnexinV/PI staining. Both inhibitors showed dose-dependent effects of inducing cell

Page 65: Targeting Oncogenic Protein Sythesis in MYC-driven B Cell

64

apoptosis, and combined treatment displayed significant synergy as indicated by a

combination index (CI) value less than 1(Figure 3.10A). While collaborative disruption of

both drive oncogene could explain the devastating impact on cell survival, it should also

be noted that such synergy may partially come from simultaneous inhibition of BCL2 and

MCL1, as silvestrol inhibits MCL1 protein synthesis. This combined strategy may also

reduce the potential of ABT199 resistance which is commonly due to acquired

dependence on MCL1. Since rocaglates repress multiple B cell related oncoproteins and (-

)-SDS-1-021 exhibit impressive potency in DHL cells, we next wanted to determine

whether normal B cells would be similarly affected. We found (-)-SDS-1-021 impressively

reduced cell viability at single digit nanomolar in DHL cells whereas CD19+ normal B

cells could tolerate (Figure 3.10B). In our hand (-)-SDS-1-021 is even more potent that

silvestrol in DHL cells (Figure 3.11B), exhibiting a 2.85 and 8.21 fold smaller EC50 in

decreasing cell viability in Val and Ros50 respectively (Figure 3.11C). In addition, (-)-SDS-

1-021 completely stalled cell proliferation at low concentration (2.5nM) and exhibited

strong killing effect at higher concentrations in all DHL lines (Figure 3.10C). We then

employed (-)-SDS-1-021 and combination treatment in a series of primary DEL/DHL cells

and found (-)-SDS-1-021 potently decreased cell viability whilst the inhibitory effect was

further enhanced when combined with ABT199 (Figure 3.10D). These findings imply

DHL/DEL cells are prone to rocaglates mediated translation inhibition, yet normal B cells

could tolerate as they do not rely dysregulated oncoproteins for survival.

Page 66: Targeting Oncogenic Protein Sythesis in MYC-driven B Cell

65

Figure 3.10. Potency of rocalgates and combined treatment with ABT199 in vitro

(A) Cell apoptosis induced by silvestrol, ABT199 and combined. Val and Ros50 cells were treated with vehicle control, silvestrol, ABT199 or combined for 48 hours. Percentage of apoptosis cells was assessed by flow cytometry with FITC-AnnexinV/PI staining and normalized to vehicle control. Drug synergy is calculated by combination index (CI) as indicated. (B) Impact of (-)-SDS-1-021(SDS) on cell viability. CD19+ normal B cells and DHL cells were treated with (-)-SDS-1-021 at various concentrations for 48 hours. Cell viability was assessed by prestoblue staining and normalized with that of vehicle control. (C) Effect of silvestrol and (-)-SDS-1-021 on cell viability in Val and Ros50 cells. (D) EC50 of silvestrol and (-)-SDS-1-021 in Val and Ros50 cells. (E) (-)-SDS-1-021 inhibits cell proliferation a single digit nanomolar. Val, Ros50, U2904 and Carnaval cells were treated with (-)-SDS-1-021 for three days. Cell viability was measured by prestroblue every day and normalized to day 0. Data shown in A-E represent mean ± SD of at least 3 independent experiment.

Page 67: Targeting Oncogenic Protein Sythesis in MYC-driven B Cell

66

Figure 3.11. Structure and EC50 of silvestrol and (-)-SDS-1-021. (A)Structures of silvestrol and (-)-SDS-1-021. (B) Effects of silvestrol and (-)-SDS-1-021(SDS) on cell viability in Val and Ros50 cells. (C) EC50 of silvestrol and (-)-SDS-1-021 in Val and Ros50 cells. Data shown in B represent mean ± SD of at least 3 independent experiments.

Page 68: Targeting Oncogenic Protein Sythesis in MYC-driven B Cell

67

3.3.8 (-)-SDS-1-021 suppresses tumor growth, and synergizes with ABT199 in PDX

models

To assess the efficacy of (-)-SDS-1-021 and combined treatment in vivo, we adopted

two established PDX mice models by injecting tumor cells into the flank of male and

female NOD-scid IL2Rgammanull(NSG) mice subcutaneously. Previous study showed (-)-

SDS-1-021 could prolong survival and be well tolerated at 0.7mg/kg twice a week for a

duration of 8 weeks in multiple myeloma xenograft mice30. Here, we are interested in the

impact of this novel compound on tumor growth at exponential state in vivo which

resembles a clinical onset. After tumor reached palpable size (100~200mm3), mice were

randomized into vehicle control and treatment groups with equal number of both genders

in each. To mimic an intense short-term first line chemotherapy that is commonly used

for treating highly aggressive B cell lymphoma such as BL and DHL, vehicle and

inhibitors were administrated to tumor bearing mice daily via intraperitoneal injection for

10 days. LTL025 is a patient derived DLBCL with MYC and BCL2 double expression not

due to translocations. (-)-SDS-1-021 at 0.2 mg/kg was able to potently repress tumor

growth (Figure 3.12A), and significantly reduced tumor burden (Figure 3.12B, C).

Immunohistochemistry staining confirmed a drastic decrease of ki67 staining and

completely abrogation of MYC protein expression (Figure 3.12B, D). These findings

suggest (-)-SDS-1-021 as single agent is effective for MYC inhibition and suppressing

tumor progression in the double expression scenario. DFBL20954 is a patient derived

DLBCL with FISH confirmed MYC and BCL2 double translocation. This DHL model

Page 69: Targeting Oncogenic Protein Sythesis in MYC-driven B Cell

68

exhibited more aggressive biological behaviors. Expeditious tumor growth, blood vessel

infiltration and local invasion were observed (Figure 3.13A, C). ABT199 at 1mg/kg

moderately delayed tumor growth and tumor burden (Figure 3.13 A-C), yet had no effect

on MYC expression or proliferation (ki67) (Figure 3.13D). In (-)-SDS-1-021 0.2mg/kg

treatment group, potent tumor repression and significant reduced tumor burden were

observed (Figure 3.13A-C). IHC staining showed substantial inhibition of MYC

expression (>95% vs <10%) and decrease of ki67 (100% vs 30%)( Figure 3.13D). (-)-SDS-1-

021 and ABT combine treatment induced profound cytotoxicity on tumor. Rapid tumor

regression was observed on day 3 and day 4. Xenograft tumor became impalpable and

remained so till end point. Resolved tumor and surrounding fatty tissue were collected.

Large area of necrosis and elimination of MYC expression were validated by IHC (Figure

3.13 A-D). Moderate body weight loss (~10%) was observed in mice treated with (-)-SDS-

1-021 and ABT199 accompanied with rapid tumor regress yet body weight was recovered

later when the tumor resolved. No significant body weight loss was observed in other

single treatment groups. Our findings indicate synthetic rocaglate derivate (-)-SDS-1-021

could potently inhibit MYC expression as well as tumor progression, and synergize with

ABT199 for rapid tumor regression in clinical relevant PDX models.

Page 70: Targeting Oncogenic Protein Sythesis in MYC-driven B Cell

69

Figure 3.12. (-)-SDS-1-021 displayed high potency as single agent in DEL PDX model

(A) Tumor growth of double expression lymphoma PDX LTL025 in NSG mice. (-)-SDS-1-021(0.2mg/kg) or vehicle control was administered by intraperitoneal (i.p.) injections daily for 10 days. (B) Weight of tumor from vehicle control or (-)-SDS-1-021 treated DEL PDX mice at endpoint. P<0.0001. (C) Representative images of the tumors from DEL PDX therapeutic study shown in (A) (B). (D) Representative IHC staining of MYC, Ki67 in DEL PDX therapeutic study. Magnification, ×20.

Page 71: Targeting Oncogenic Protein Sythesis in MYC-driven B Cell

70

Figure 3.13. (-)-SDS-1-021 displayed high potency, and synergy with ABT199 in DHL PDX model

(A) Tumor growth of double hit lymphoma PDX DFBL20954 in NSG mice. (-)-SDS-1-021(0.2mg/kg), ABT199 (1mg/kg), combined or vehicle control was administered by intraperitoneal (i.p.) injections daily for 10 days. (B) Weight of tumor from vehicle control or DHL PDX mice in therapeutic groups at endpoint. P value between combined and ctrl group P <0.0001, ABT199 group P =0.0013 (-)-SDS-1-021 group P =0.0019. (C) Representative images of the tumors from DEL PDX therapeutic study shown in (A) (B). (D) Representative IHC staining of MYC and Ki67 in DHL PDX, and impact of (-)-SDS-1-021, ABT199 or combined treatment on their expression. Magnification, ×20.

Page 72: Targeting Oncogenic Protein Sythesis in MYC-driven B Cell

71

3.4 Discussion

Deregulated protein synthesis a hallmark of many cancers which often correlates with

genetic aberrations to promote tumor survival and progression. eIF4F is the key regulator

of cap-dependent ribosome recruitment in translation initiation. Components in the eIF4F

complex (eIF4E, eIF4A, eIF4G) are commonly found overexpressed in cancers due to

activation of oncogenic signaling such as MYC, mTOR, Ras and MAPK51. In MYC driven

lymphomas, hyperactivated protein synthesis is required to sustain the rapid

proliferation, and therefore contributes to the unique aggressive behaviors. MYC could

coordinate transcription and translation by upregulating eIF4E, eIF4A and eIF4G70.

Upregulated cap-dependent translation in turn promotes excessive production of MYC,

thereby forming a feed-forward loop between MYC overexpression and active mRNA

translation. As a result, targeting mRNA translation initiations seems especially

promising for treating MYC driven lymphomas. Indeed, our drug screening identified

protein synthesis exhibiting highest potency in a variety of MYC driven lymphoma cell

lines. However, we found mTOR inhibitors were not able to efficiently repress MYC

mRNA translation. eIF4E is the least abundant initiation factor in eIF4F and thus

considered the limiting factor for cap-dependent translation. Initiation factors exist in

excess under physiological conditions, previous study showed 50% reduction of eIF4E did

not alter global polysome profiles and was fully compatible with normal organismal

development and physiology in eIF4E haploinsufficiency mice71. We found in MYC

driven lymphoma eIF4E does also exist in excess. The observation that Torin1 failed to

repress MYC is likely due to the vastly abundant eIF4E in DHL cells, as introducing

Page 73: Targeting Oncogenic Protein Sythesis in MYC-driven B Cell

72

dominant negative 4EBP1-4A only marginally decreased MYC and directly knocking

down 80% eIF4E only reduced MYC by half. On the other hand, rocaglates uniformly

abolished MYC mRNA translation at low concentration in all MYC driven B cell

lymphoma we tested, and showed strong inhibition on both IRES and cap-dependent

translation of MYC. It is intriguing to see rocaglates would inhibit MYC and other

oncogene in such potent manner by disrupting another initiation factor eIF4A that is

generally considered even more abundant in eIF4F.

eIF4A1 and eIF4A2 are the targets of rocaglates. While most studies investigating cap-

dependent translation initiation focused on eIF4A1 as it is more abundant, eIF4A2 was

thought simply functional redundant and sometimes mistaken as an isoform of eIF4A1

due to their high sequence similarity in protein coding. We found both eIF4A1 and eIF4A2

may participate in MYC cap-dependent translation initiation, and the preference for

which is determined by their abundance in eIF4F complex, as a consequence of their

relative expression ratio. That being said, eIF4A2 seems to play a completely different role

in and out of eIF4F. eIF4A2 acts similar to eIF4A1 and regulates cap-dependent translation

initiation when it is present in eIF4F, however when sequestered out of eIF4F by

overwhelming amount of eIF4A1, eIF4A2 displayed a translational inhibitory effect as

knockdown of eIF4A2 increased MYC, which is possibly through microRNA mediated

gene regulation57. It does make sense since eIF4A2 expression in not regulated by MYC

and consistent among normal B cells and DHL cells whereas eIF4A and eIF4E are highly

overexpressed in DHL. Although future studies are required to validate this theory, these

Page 74: Targeting Oncogenic Protein Sythesis in MYC-driven B Cell

73

observations may help to explain the controversial role of eIF4A2 in predicting disease

outcome72,73.

Under physiological conditions, eIF4A is the most abundant initiation factor in eIF4F,

and is likely still excessive than required in pathological condition where MYC leverages

the entire translation machinery. Consistent with this assumption, we found over 50%

knock down of either eIFA1, eIF4A2 or combined did not significantly reduce MYC

protein. Rocaglates increase binding affinity between eIF4A and mRNA by clamping

eIF4A on polypurine rich mRNA53,54, resulting two theories of the mechanism of action

“ribosome scanning prevention” and “eIF4A cap depletion”. Since “eIF4A cap depletion”

theory is based on the evidence that the abundance of eIF4A in eIF4F was decreased by

rocaglates74,75, we performed m7-cap pull down to compare the change of eIF4A in the

presence of rocaglates with that in eIF4A knock down. Surprisingly, rocaglates or eIF4A

knockdown did not necessarily deplete eIF4A in situation where immensely abundant

eIF4A compete for relative limited eIF4E. And the decrease of eIF4A in eIF4F by rocaglates

is much less than that by eIF4A knock down, which actually had little impact on MYC

expression. These findings argue the decrease of eIF4A in eIF4F observed in rocaglates

treated cells may not be biologically relevant to its potent inhibition of MYC. Therefore,

rocaglates do not repress MYC, and possible other mRNA translation by depleting eIF4A

for cap-dependent translation. Potent translation inhibition is more likely through

blocking ribosome scanning in which eIF4A merely acts as a selective roadblock. For that

reason, it only requires a fairly small amount of eIF4A to be clamped on the outnumbered

active translating mRNA to suppress the translation initiation, as rocaglates stabilize

Page 75: Targeting Oncogenic Protein Sythesis in MYC-driven B Cell

74

eIF4A-mRNA binding. On the other hand, eIF4A ATPase inhibitors Hippuristanol and

elatol work in a different mechanism by directly disrupting eIF4A helicase function, and

studies have shown eIF4A ATPase inhibitor induced proteome change is different from

that by rocaglates53. Since the majority of eIF4A exists in free form not in eIF4F complex,

these inhibitors may have to saturate a gigantic target pool in order to incapacitate the

small proportion of eIF4A in eIF4F and subsequently disable cap-dependent translation

initiation, which explains the low potency of Hippuristanol observed. Previous studies

reported varied response to elatol in non-Hodgkin lymphomas, while silvestrol uniformly

displayed higher potency at 5 to over 1000 fold76. Overall, rocaglates should be considered

as selective translation initiation inhibitors not eIF4A inhibitors because the translation

inhibition they inflicted is not related to the function of eIF4A in eIF4F complex. This

unique mode of action is actually advantageous because the minimal amount eIF4A

required for oncogenic mRNA translation is sufficient for rocaglates to utilize while

excessive expression of eIF4A in MYC driven lymphomas cannot rescue.

Rocaglates not only manifest strong inhibition on MYC translation but also suppress

multiple other oncoproteins. Repression of AURAKA, AURAKB and PLK1 destabilizes

MYC, together with inhibition of NEK2, CHEK1, WEE1 and other cell cycle regulators

such as cyclins, collaboratively suppress tumor cell proliferation. Rocaglates also deplete

several B cell lymphoma related transcription factor that are non-targetable including

TCF3, BCL6 and NOTCH1. Notably, inhibition of individual oncoprotein identified above

such as CHEK1, WEE1 and PLK1 by kinase inhibitors has showed profound impact on

MYC driven lymphoma cell survival in our initial drug screening and in other previous

Page 76: Targeting Oncogenic Protein Sythesis in MYC-driven B Cell

75

studies40,60,63-65,67. The ability to target multiple critical oncoproteins may partially explain

the high potency of rocaglates in DHL. While inhibitors targeting a single component or

pathway are likely to fail due to adaption like transcriptional rewiring, the broad

oncogenic protein inhibition by rocaglates confers a comprehensive blockage on multiple

oncogenic pathways that is difficult for tumor cells to tolerate in the first place and

generate adaption. Unfortunately, rocaglates cannot inhibit translation of BCL2, the

hallmark anti-apoptotic protein overexpressed in DHL. Therefore, we propose the

combination of ABT199 with rocaglate for DHL treatment. (-)-SDS-1-021, the most potent

synthetic rocaglate derivate, efficiently inhibits tumor cell proliferation at single-digit

nanomolar but spares normal B cells in vitro. A 0.2mg/kg dose stalls in vivo tumor

proliferation as single agent and nearly diminishes MYC expression, and induces rapid

tumor regression combined with ABT199 in DHL PDX models.

Concisely, in pursuit of a novel approach for treating MYC driven lymphoma,

especially the most challenging type DHL, we identified rocaglate as the most potent and

suitable class of translation initiation inhibitor. Several characteristics have made

rocaglates very promising for this type of disease, and possible all aggressive cancers.

First, MYC-driven lymphomas overexpress translation initiation factors in excess to

accommodate the rapid proliferation. As shown here, moderate functional depletion of

these initiation factors may not impair MYC protein synthesis. Rocaglates present a

unique mode of translation inhibition which relates little if any to eIF4A function in cap-

dependent translation, therefore they can efficiently repress oncoprotein synthesis

regardless of eIF4A expression level in all cell lines. Second, rocaglates selectively inhibit

Page 77: Targeting Oncogenic Protein Sythesis in MYC-driven B Cell

76

multiple oncoproteins that are critical for B cell lymphoma. Collaborative suppression of

MYC regulation network and oncogenic pathways could minimize the possibility of

developed drug resistance commonly observed in target therapies. Finally, large scale

total synthesis of rocaglate derivatives was achieved recently, and hence the limited

bioavailability from natural resources will no longer hinder the potential use of this class

of inhibitors. Besides, despite silvestrol has shown favorable pharmacokinetics in mice77,

the pharmacokinetics and pharmacodynamics of rocalgate derivate such as (-)-SDS-1-021

may require further investigation in clinical studies. Based the preclinical findings

presented here, we believe this class of inhibitor is a favorable candidate for treating

aggressive B cell lymphoma and may be used combination with ABT199 specific for DHL

management.

Page 78: Targeting Oncogenic Protein Sythesis in MYC-driven B Cell

77

Conclusion and Pitfalls

In this study, we provided rationales for using synthetic rocaglates to treat MYC

driven lymphomas, from a mechanistic aspect and in preclinical settings. We

demonstrated rocaglates, a class of translation initiation inhibitor is particular promising

as they efficiently repress the expression of MYC and many other oncoproteins required

for B cell lymphoma survival by drug screen and proteomic profiling. More importantly,

unlike mTOR inhibitors, eIF4A helicase inhibitor hippuristanol and similar therapeutic

strategies that attempt to deplete eukaryotic translation initiation factors, rocaglates

directly prevent translation by increasing target mRNA binding to eIF4A, therefore their

efficacy is not compromised by the overly abundant eIF4A in MYC-driven lymphomas.

Finally, we proved (-)-SDS-1-021, the most potent synthetic rocalgate, is highly potent and

exhibits strong synergy with the BCL2 inhibitor ABT199 in a series of primary double

hit/double expression lymphoma cells in vitro and in patient derived xenograft models of

double hit and double expression lymphoma in vivo.

Notably, the use of rocaglates or derivatives for antitumor therapy was underappreciated

because of their limited bioavailability in the past. Our collaborator John Porco Jr. and his

colleagues have recently unveiled a few promising synthetic rocoglates and achieved total

synthesis at multigram level, which unlocks the potential use for clinical applications.

Indeed, this class of compounds have drawn attention from both academia and

pharmaceutical companies, as one phase 1 clinical trial is planned in 2019. This study is

one of the few pioneers highlighting the therapeutic value of rocaglates in hematopoietic

Page 79: Targeting Oncogenic Protein Sythesis in MYC-driven B Cell

78

malignancies, which will encourage more clinical studies and bring innovations to current

treatment for this disease.

This study also has a few limitations. The drug screen we used is a relative small pool,

there might be inhibitors that are more potent than silvestrol in MYC-driven lymphomas.

In the second part, a positive control eg.MCL1 expression should be tested in 4EBP1-4A

transduced cells. In DHL cells transduced with dual luciferase reporter construct, the

number of respective luciferase mRNA transcripts is not strictly controlled. It would be

better to transfect fixed amount of in vitro synthesized mRNA however it is nearly

impossible to transfect full length luciferase construct mRNA into B cells. While native

RIP provides an easy way to measure mRNA enrichment, the elution method is not

stringent which may cause a lot of non-specific binding. Enhanced crosslinking RNA

immunoprecipitation (eCLIP) may help to determine the exact eIF4A1 and MYC binding

site. In animal study, due to insufficient (-)-SDS-1-021 compound, we were not able to

perform long term survival study, therefore the long term safety is unknown.

Based on these limitations, two experiments would improve the quality of this study.

Polysome fractions could be divided into monosome and heavy polysome groups, and

RNA-seq could be performed to profile the entire mRNA pool impacted by silvestrol

treatment. eCLIP could be performed to determine the eIF4A1 and MYC binding site, and

followed by RNA sequencing, may help to discovery the mRNA motif required for this

interfacial binding.

Page 80: Targeting Oncogenic Protein Sythesis in MYC-driven B Cell

79

Bibliography

1. Sabo A, Kress TR, Pelizzola M, et al. Selective transcriptional regulation by Myc in

cellular growth control and lymphomagenesis. Nature. 2014;511(7510):488-492.

2. Nie Z, Hu G, Wei G, et al. c-Myc is a universal amplifier of expressed genes in

lymphocytes and embryonic stem cells. Cell. 2012;151(1):68-79.

3. Lin CY, Loven J, Rahl PB, et al. Transcriptional amplification in tumor cells with

elevated c-Myc. Cell. 2012;151(1):56-67.

4. Sander S, Calado DP, Srinivasan L, et al. Synergy between PI3K signaling and

MYC in Burkitt lymphomagenesis. Cancer Cell. 2012;22(2):167-179.

5. Ott G, Rosenwald A, Campo E. Understanding MYC-driven aggressive B-cell

lymphomas: pathogenesis and classification. Blood. 2013;122(24):3884-3891.

6. Calado DP, Sasaki Y, Godinho SA, et al. The cell-cycle regulator c-Myc is essential

for the formation and maintenance of germinal centers. Nat Immunol.

2012;13(11):1092-1100.

7. Dominguez-Sola D, Victora GD, Ying CY, et al. The proto-oncogene MYC is

required for selection in the germinal center and cyclic reentry. Nat Immunol.

2012;13(11):1083-1091.

8. Scott DW, King RL, Staiger AM, et al. High-grade B-cell lymphoma with MYC and

BCL2 and/or BCL6 rearrangements with diffuse large B-cell lymphoma

morphology. Blood. 2018;131(18):2060-2064.

Page 81: Targeting Oncogenic Protein Sythesis in MYC-driven B Cell

80

9. Savage KJ, Johnson NA, Ben-Neriah S, et al. MYC gene rearrangements are

associated with a poor prognosis in diffuse large B-cell lymphoma patients treated

with R-CHOP chemotherapy. Blood. 2009;114(17):3533-3537.

10. Klapper W, Stoecklein H, Zeynalova S, et al. Structural aberrations affecting the

MYC locus indicate a poor prognosis independent of clinical risk factors in diffuse

large B-cell lymphomas treated within randomized trials of the German High-

Grade Non-Hodgkin's Lymphoma Study Group (DSHNHL). Leukemia.

2008;22(12):2226-2229.

11. Barrans S, Crouch S, Smith A, et al. Rearrangement of MYC is associated with poor

prognosis in patients with diffuse large B-cell lymphoma treated in the era of

rituximab. J Clin Oncol. 2010;28(20):3360-3365.

12. Aukema SM, Siebert R, Schuuring E, et al. Double-hit B-cell lymphomas. Blood.

2011;117(8):2319-2331.

13. Johnson NA, Savage KJ, Ludkovski O, et al. Lymphomas with concurrent BCL2

and MYC translocations: the critical factors associated with survival. Blood.

2009;114(11):2273-2279.

14. Petrich AM, Gandhi M, Jovanovic B, et al. Impact of induction regimen and stem

cell transplantation on outcomes in double-hit lymphoma: a multicenter

retrospective analysis. Blood. 2014;124(15):2354-2361.

15. Swerdlow SH, Campo E, Pileri SA, et al. The 2016 revision of the World Health

Organization classification of lymphoid neoplasms. Blood. 2016;127(20):2375-2390.

Page 82: Targeting Oncogenic Protein Sythesis in MYC-driven B Cell

81

16. Oki Y, Noorani M, Lin P, et al. Double hit lymphoma: the MD Anderson Cancer

Center clinical experience. Br J Haematol. 2014;166(6):891-901.

17. Friedberg JW. How I treat double-hit lymphoma. Blood. 2017;130(5):590-596.

18. Puvvada SD, Stiff PJ, Leblanc M, et al. Outcomes of MYC-associated lymphomas

after R-CHOP with and without consolidative autologous stem cell transplant:

subset analysis of randomized trial intergroup SWOG S9704. Br J Haematol.

2016;174(5):686-691.

19. Perry AM, Crockett D, Dave BJ, et al. B-cell lymphoma, unclassifiable, with

features intermediate between diffuse large B-cell lymphoma and burkitt

lymphoma: study of 39 cases. Br J Haematol. 2013;162(1):40-49.

20. Carey CD, Gusenleitner D, Chapuy B, et al. Molecular classification of MYC-

driven B-cell lymphomas by targeted gene expression profiling of fixed biopsy

specimens. J Mol Diagn. 2015;17(1):19-30.

21. Doroshow DB, Eder JP, LoRusso PM. BET inhibitors: a novel epigenetic approach.

Ann Oncol. 2017;28(8):1776-1787.

22. Stathis A, Zucca E, Bekradda M, et al. Clinical Response of Carcinomas Harboring

the BRD4-NUT Oncoprotein to the Targeted Bromodomain Inhibitor OTX015/MK-

8628. Cancer Discov. 2016;6(5):492-500.

23. Pawar A, Gollavilli PN, Wang S, Asangani IA. Resistance to BET Inhibitor Leads

to Alternative Therapeutic Vulnerabilities in Castration-Resistant Prostate Cancer.

Cell Rep. 2018;22(9):2236-2245.

Page 83: Targeting Oncogenic Protein Sythesis in MYC-driven B Cell

82

24. Kurimchak AM, Shelton C, Duncan KE, et al. Resistance to BET Bromodomain

Inhibitors Is Mediated by Kinome Reprogramming in Ovarian Cancer. Cell Rep.

2016;16(5):1273-1286.

25. Barna M, Pusic A, Zollo O, et al. Suppression of Myc oncogenic activity by

ribosomal protein haploinsufficiency. Nature. 2008;456(7224):971-975.

26. Elkon R, Loayza-Puch F, Korkmaz G, et al. Myc coordinates transcription and

translation to enhance transformation and suppress invasiveness. EMBO Rep.

2015;16(12):1723-1736.

27. Hinnebusch AG, Ivanov IP, Sonenberg N. Translational control by 5'-untranslated

regions of eukaryotic mRNAs. Science. 2016;352(6292):1413-1416.

28. Topisirovic I, Svitkin YV, Sonenberg N, Shatkin AJ. Cap and cap-binding proteins

in the control of gene expression. Wiley Interdiscip Rev RNA. 2011;2(2):277-298.

29. Lucas DM, Edwards RB, Lozanski G, et al. The novel plant-derived agent silvestrol

has B-cell selective activity in chronic lymphocytic leukemia and acute

lymphoblastic leukemia in vitro and in vivo. Blood. 2009;113(19):4656-4666.

30. Manier S, Huynh D, Shen YJ, et al. Inhibiting the oncogenic translation program is

an effective therapeutic strategy in multiple myeloma. Sci Transl Med. 2017;9(389).

31. Pan L, Woodard JL, Lucas DM, Fuchs JR, Kinghorn AD. Rocaglamide, silvestrol

and structurally related bioactive compounds from Aglaia species. Nat Prod Rep.

2014;31(7):924-939.

Page 84: Targeting Oncogenic Protein Sythesis in MYC-driven B Cell

83

32. Oblinger JL, Burns SS, Huang J, et al. Overexpression of eIF4F components in

meningiomas and suppression of meningioma cell growth by inhibiting

translation initiation. Exp Neurol. 2018;299(Pt B):299-307.

33. Cencic R, Carrier M, Galicia-Vazquez G, et al. Antitumor activity and mechanism

of action of the cyclopenta[b]benzofuran, silvestrol. PLoS One. 2009;4(4):e5223.

34. Oblinger JL, Burns SS, Akhmametyeva EM, et al. Components of the eIF4F

complex are potential therapeutic targets for malignant peripheral nerve sheath

tumors and vestibular schwannomas. Neuro Oncol. 2016;18(9):1265-1277.

35. Wiegering A, Uthe FW, Jamieson T, et al. Targeting Translation Initiation Bypasses

Signaling Crosstalk Mechanisms That Maintain High MYC Levels in Colorectal

Cancer. Cancer Discov. 2015;5(7):768-781.

36. Stone SD, Lajkiewicz NJ, Whitesell L, Hilmy A, Porco JA, Jr. Biomimetic kinetic

resolution: highly enantio- and diastereoselective transfer hydrogenation of aglain

ketones to access flavagline natural products. J Am Chem Soc. 2015;137(1):525-530.

37. Rodrigo CM, Cencic R, Roche SP, Pelletier J, Porco JA. Synthesis of rocaglamide

hydroxamates and related compounds as eukaryotic translation inhibitors:

synthetic and biological studies. J Med Chem. 2012;55(1):558-562.

38. Wang W, Cencic R, Whitesell L, Pelletier J, Porco JA, Jr. Synthesis of Aza-

Rocaglates via ESIPT-Mediated (3+2) Photocycloaddition. Chemistry.

2016;22(34):12006-12010.

Page 85: Targeting Oncogenic Protein Sythesis in MYC-driven B Cell

84

39. Yueh H, Gao Q, Porco JA, Jr., Beeler AB. A photochemical flow reactor for large

scale syntheses of aglain and rocaglate natural product analogues. Bioorg Med

Chem. 2017;25(23):6197-6202.

40. Ren Y, Bi C, Zhao X, et al. PLK1 stabilizes a MYC-dependent kinase network in

aggressive B cell lymphomas. J Clin Invest. 2018.

41. Gagliardi M, Matarazzo MR. RIP: RNA Immunoprecipitation. Methods in molecular

biology. 2016;1480:73-86.

42. Chou TC. Drug combination studies and their synergy quantification using the

Chou-Talalay method. Cancer Res. 2010;70(2):440-446.

43. Rong L, Livingstone M, Sukarieh R, et al. Control of eIF4E cellular localization by

eIF4E-binding proteins, 4E-BPs. Rna. 2008;14(7):1318-1327.

44. Park IW, Reddy MV, Reddy EP, Groopman JE. Evaluation of novel cell cycle

inhibitors in mantle cell lymphoma. Oncogene. 2007;26(38):5635-5642.

45. Moerke NJ, Aktas H, Chen H, et al. Small-molecule inhibition of the interaction

between the translation initiation factors eIF4E and eIF4G. Cell. 2007;128(2):257-

267.

46. Pettersson F, Del Rincon SV, Miller WH, Jr. Eukaryotic translation initiation factor

4E as a novel therapeutic target in hematological malignancies and beyond. Expert

Opin Ther Targets. 2014;18(9):1035-1048.

47. Yang T, Buchan HL, Townsend KJ, Craig RW. MCL-1, a member of the BLC-2

family, is induced rapidly in response to signals for cell differentiation or death,

but not to signals for cell proliferation. J Cell Physiol. 1996;166(3):523-536.

Page 86: Targeting Oncogenic Protein Sythesis in MYC-driven B Cell

85

48. Raynaud FI, Orr RM, Goddard PM, et al. Pharmacokinetics of G3139, a

phosphorothioate oligodeoxynucleotide antisense to bcl-2, after intravenous

administration or continuous subcutaneous infusion to mice. J Pharmacol Exp Ther.

1997;281(1):420-427.

49. Chu J, Galicia-Vazquez G, Cencic R, et al. CRISPR-Mediated Drug-Target

Validation Reveals Selective Pharmacological Inhibition of the RNA Helicase,

eIF4A. Cell Rep. 2016;15(11):2340-2347.

50. Bordeleau ME, Robert F, Gerard B, et al. Therapeutic suppression of translation

initiation modulates chemosensitivity in a mouse lymphoma model. J Clin Invest.

2008;118(7):2651-2660.

51. Pelletier J, Graff J, Ruggero D, Sonenberg N. Targeting the eIF4F translation

initiation complex: a critical nexus for cancer development. Cancer Res.

2015;75(2):250-263.

52. Iwasaki S, Iwasaki W, Takahashi M, et al. The Translation Inhibitor Rocaglamide

Targets a Bimolecular Cavity between eIF4A and Polypurine RNA. Mol Cell. 2018.

53. Iwasaki S, Floor SN, Ingolia NT. Rocaglates convert DEAD-box protein eIF4A into

a sequence-selective translational repressor. Nature. 2016;534(7608):558-561.

54. Chambers JM, Lindqvist LM, Webb A, Huang DC, Savage GP, Rizzacasa MA.

Synthesis of biotinylated episilvestrol: highly selective targeting of the translation

factors eIF4AI/II. Org Lett. 2013;15(6):1406-1409.

55. Andreou AZ, Klostermeier D. The DEAD-box helicase eIF4A: paradigm or the odd

one out? RNA Biol. 2013;10(1):19-32.

Page 87: Targeting Oncogenic Protein Sythesis in MYC-driven B Cell

86

56. Galicia-Vazquez G, Cencic R, Robert F, Agenor AQ, Pelletier J. A cellular response

linking eIF4AI activity to eIF4AII transcription. Rna. 2012;18(7):1373-1384.

57. Meijer HA, Kong YW, Lu WT, et al. Translational repression and eIF4A2 activity

are critical for microRNA-mediated gene regulation. Science. 2013;340(6128):82-85.

58. Chu J, Cencic R, Wang W, Porco JA, Jr., Pelletier J. Translation Inhibition by

Rocaglates Is Independent of eIF4E Phosphorylation Status. Molecular cancer

therapeutics. 2016;15(1):136-141.

59. Gu Z, Zhou W, Huang J, et al. Nek2 is a novel regulator of B cell development and

immunological response. Biomed Res Int. 2014;2014:621082.

60. Fang Y, Zhang X. Targeting NEK2 as a promising therapeutic approach for cancer

treatment. Cell Cycle. 2016;15(7):895-907.

61. den Hollander J, Rimpi S, Doherty JR, et al. Aurora kinases A and B are up-

regulated by Myc and are essential for maintenance of the malignant state. Blood.

2010;116(9):1498-1505.

62. Chila R, Basana A, Lupi M, et al. Combined inhibition of Chk1 and Wee1 as a new

therapeutic strategy for mantle cell lymphoma. Oncotarget. 2015;6(5):3394-3408.

63. Restelli V, Vagni M, Arribas AJ, Bertoni F, Damia G, Carrassa L. Inhibition of

CHK1 and WEE1 as a new therapeutic approach in diffuse large B cell lymphomas

with MYC deregulation. Br J Haematol. 2018;181(1):129-133.

64. Xiao D, Yue M, Su H, et al. Polo-like Kinase-1 Regulates Myc Stabilization and

Activates a Feedforward Circuit Promoting Tumor Cell Survival. Mol Cell.

2016;64(3):493-506.

Page 88: Targeting Oncogenic Protein Sythesis in MYC-driven B Cell

87

65. Bonetti P, Testoni M, Scandurra M, et al. Deregulation of ETS1 and FLI1

contributes to the pathogenesis of diffuse large B-cell lymphoma. Blood.

2013;122(13):2233-2241.

66. Schmitz R, Ceribelli M, Pittaluga S, Wright G, Staudt LM. Oncogenic mechanisms

in Burkitt lymphoma. Cold Spring Harb Perspect Med. 2014;4(2).

67. Lobry C, Oh P, Mansour MR, Look AT, Aifantis I. Notch signaling: switching an

oncogene to a tumor suppressor. Blood. 2014;123(16):2451-2459.

68. Basso K, Dalla-Favera R. BCL6: master regulator of the germinal center reaction

and key oncogene in B cell lymphomagenesis. Adv Immunol. 2010;105:193-210.

69. Adams JM, Cory S. The BCL-2 arbiters of apoptosis and their growing role as

cancer targets. Cell Death Differ. 2018;25(1):27-36.

70. Malka-Mahieu H, Newman M, Desaubry L, Robert C, Vagner S. Molecular

Pathways: The eIF4F Translation Initiation Complex-New Opportunities for

Cancer Treatment. Clin Cancer Res. 2017;23(1):21-25.

71. Truitt ML, Conn CS, Shi Z, et al. Differential Requirements for eIF4E Dose in

Normal Development and Cancer. Cell. 2015;162(1):59-71.

72. Shaoyan X, Juanjuan Y, Yalan T, Ping H, Jianzhong L, Qinian W. Downregulation

of EIF4A2 in non-small-cell lung cancer associates with poor prognosis. Clin Lung

Cancer. 2013;14(6):658-665.

73. Chen Z-H, Wu Q, Lu J, et al. Effect of knockdown of eukaryotic initiation factor

4A2 (eIF4A2) on growth, metastasis, and oxaliplatin sensitivity in colorectal

cancer. 2018;36(15_suppl):e15658-e15658.

Page 89: Targeting Oncogenic Protein Sythesis in MYC-driven B Cell

88

74. Bordeleau ME, Matthews J, Wojnar JM, et al. Stimulation of mammalian

translation initiation factor eIF4A activity by a small molecule inhibitor of

eukaryotic translation. Proceedings of the National Academy of Sciences of the United

States of America. 2005;102(30):10460-10465.

75. Low WK, Dang Y, Schneider-Poetsch T, et al. Inhibition of eukaryotic translation

initiation by the marine natural product pateamine A. Mol Cell. 2005;20(5):709-722.

76. Peters TL, Tillotson J, Yeomans AM, et al. Target-Based Screening against eIF4A1

Reveals the Marine Natural Product Elatol as a Novel Inhibitor of Translation

Initiation with In Vivo Antitumor Activity. Clin Cancer Res. 2018;24(17):4256-4270.

77. Saradhi UV, Gupta SV, Chiu M, et al. Characterization of silvestrol

pharmacokinetics in mice using liquid chromatography-tandem mass

spectrometry. AAPS J. 2011;13(3):347-356.