<<

Downloaded from molecularcasestudies.cshlp.org on October 9, 2021 - Published by Cold Spring Harbor Laboratory Press

1 Clinical response of the novel activating ALK-I1171T mutation in

2 neuroblastoma to the ALK inhibitor ceritinib.

3

4 Jikui Guan1*, Susanne Fransson2*, Joachim Tetteh Siaw1*, Diana Treis4*, Jimmy Van den 5 Eynden1, Damini Chand1, Ganesh Umapathy1, Kristina Ruuth9, Petter Svenberg4, Sandra 6 Wessman5, Alia Shamikh5, Hans Jacobsson6, Lena Gordon7, Jakob Stenman8, Pär-Johan 7 Svensson8, Magnus Hansson3, Erik Larsson1, Tommy Martinsson2#, Ruth H. Palmer1#, Per 8 Kogner5#, Bengt Hallberg1#.

9

10 Departments of 1Medical Biochemistry and Cell Biology, 2Pathology and Genetics, 3Pediatrics and 11 Pathology, Institute of Biomedicine, The Sahlgrenska Academy, University of Gothenburg, 12 Gothenburg, Sweden, 4Childhood Cancer Research Unit, Department of Women’s and Children’s 13 Health, and Pediatric Oncology Program Karolinska University Hospital, 5Department of Oncology- 14 Pathology, Karolinska Institutet, and Dept. of Clinical Pathology, Karolinska University Hospital, 15 6Department of Radiology, Karolinska University Hospital, 7Dept. of Pediatric Radiology, Astrid 16 Lindgren Children’s Hospital, Karolinska University Hospital, 8Department of Pediatric Surgery, 17 Astrid Lindgren Children’s Hospital, Karolinska University Hospital, Stockholm, Sweden and 18 9Institute of Molecular Biology, Umeå University, Umeå, Sweden.

19

20

21 Running title: Clinical response to ceritinib in ALK-positive neuroblastoma.

22

23 #Correspondence: Bengt Hallberg ([email protected]), Ruth. H. Palmer 24 ([email protected]), Tommy Martinsson ([email protected]) and Per Kogner 25 ([email protected]).

26

27

1

Downloaded from molecularcasestudies.cshlp.org on October 9, 2021 - Published by Cold Spring Harbor Laboratory Press

1 Abstract

2

3 Tumors with Anaplastic Kinase (ALK) fusion rearrangements, including

4 non-small cell and anaplastic large cell lymphoma, are highly sensitive to ALK

5 tyrosine kinase inhibitors (TKIs), underscoring the notion that such cancers are addicted to

6 ALK activity. While mutations in ALK are heavily implicated in childhood neuroblastoma,

7 response to the ALK TKI has been disappointing. Embryonal tumors in patients

8 with DNA repair defects such as Fanconi anemia (FA) often have a poor prognosis, due to

9 lack of therapeutic options. Here we report a child with underlying FA and ALK mutant high-

10 risk neuroblastoma responding strongly to precision therapy with the ALK TKI ceritinib.

11 Conventional treatment caused severe, life-threatening toxicity. Genomic

12 analysis of the initial biopsy identified germ-line FANCA mutations as well as a novel ALK-

13 I1171T variant. ALK-I1171T generates a potent gain-of-function mutant, as measured in

14 PC12 cell neurite outgrowth and NIH3T3 transformation. Pharmacological inhibition

15 profiling of ALK-I1171T in response to various ALK TKIs identified an 11-fold improved

16 inhibition of ALK-I1171T with ceritinib when compared with crizotinib. Immunoaffinity-

17 coupled LC-MS/MS phosphoproteomics analysis indicated a decrease in ALK signaling in

18 response to ceritinib. Ceritinib was therefore selected for treatment in this child. Mono-

19 therapy with ceritinib was well tolerated and resulted in normalized catecholamine markers

20 and tumor shrinkage. After 7.5 months treatment, residual primary tumor shrunk, was

21 surgically removed and exhibited hallmarks of differentiation together with reduced Ki67

22 levels. Clinical follow-up after 21 months treatment revealed complete clinical remission

23 including all metastatic sites. Therefore, ceritinib presents a viable therapeutic option for

24 ALK-positive neuroblastoma.

25

26

2

Downloaded from molecularcasestudies.cshlp.org on October 9, 2021 - Published by Cold Spring Harbor Laboratory Press

1 Introduction

2 Anaplastic Lymphoma Kinase (ALK) is a (RTK) comprising an

3 extracellular ligand-binding domain, a transmembrane domain and an intracellular tyrosine

4 kinase domain (Iwahara, Fujimoto et al. 1997, Morris, Naeve et al. 1997). ALK was first

5 described in 1994 as a fusion partner in the t(2;5) chromosomal translocation in anaplastic

6 large cell lymphoma (ALCL) (Morris, Kirstein et al. 1994). Vertebrate ALK is activated by

7 the recently described small secreted ALKAL (FAM150/AUG) ligands, which potently

8 activate ALK signaling (Zhang, Pao et al. 2014, Guan, Umapathy et al. 2015, Reshetnyak,

9 Murray et al. 2015, Mo, Cheng et al. 2017, Fadeev, Mendoza-Garcia et al. 2018). ALK

10 activates multiple signaling pathways, such as the PI3K-AKT, CRKL-C3G, MEKK2/3-

11 MEK5-ERK5, JAK-STAT and MAPK pathways (Hallberg and Palmer 2016).

12 Mutation of full-length ALK has been well described in the neural crest derived

13 pediatric cancer neuroblastoma (Caren, Abel et al. 2008, Chen, Takita et al. 2008, George,

14 Sanda et al. 2008, Janoueix-Lerosey, Lequin et al. 2008, Mosse, Laudenslager et al. 2008,

15 Hallberg and Palmer 2013). Of the more than 35 mutations in ALK described, the majority

16 are point mutations in the kinase domain, although deletions in the extracellular domain and

17 translocations have also been reported (Okubo, Takita et al. 2012, Cazes, Louis-Brennetot et

18 al. 2013, Hallberg and Palmer 2013, Bresler, Weiser et al. 2014, Fransson, Hansson et al.

19 2015). Within the kinase domain three hotspot residues, F1174, F1245 and R1275, represent

20 most patient mutations (De Brouwer, De Preter et al. 2010, Hallberg and Palmer 2013,

21 Bresler, Weiser et al. 2014). In addition to ALK mutations in treatment-naïve neuroblastoma,

22 examination of ALK status in relapsed neuroblastoma samples has highlighted an increase in

23 activating ALK mutations appearing later in the disease course (Martinsson, Eriksson et al.

24 2011, Schleiermacher, Javanmardi et al. 2014, Eleveld, Oldridge et al. 2015).

3

Downloaded from molecularcasestudies.cshlp.org on October 9, 2021 - Published by Cold Spring Harbor Laboratory Press

1 In neuroblastoma, ALK signaling has been shown to act synergistically with MYCN to

2 drive tumor development (Berry, Luther et al. 2012, Heukamp, Thor et al. 2012, Schonherr,

3 Ruuth et al. 2012, Zhu, Lee et al. 2012, Cazes, Lopez-Delisle et al. 2014, Ueda, Nakata et al.

4 2016), with combined occurrence of MYCN amplification and ALK mutations being

5 associated with particularly bad prognosis (De Brouwer, De Preter et al. 2010). The

6 importance of ALK has lead to the suggestion that inhibition of ALK with small molecule

7 tyrosine kinase inhibitors (TKIs) may offer clinical benefit in neuroblastoma. The ALK TKI

8 crizotinib was approved for clinical use in patients with ALK-positive non-small cell lung

9 cancer (NSCLC) in 2011 ((FDA) 26th of August, 2011, Kwak, Bang et al. 2010), based on a

10 robust response in this patient population. Although significant problems with resistance to

11 ALK TKIs occur, a trial comparing crizotinib with chemotherapy concluded that crizotinib is

12 superior in patients with previously treated, advanced ALK-positive NSCLC (Shaw, Kim et

13 al. 2013). Similarly, treatment with crizotinib resulted in a strong response in a phase I

14 crizotinib monotherapy trial of pediatric patients with ALK-fusion positive tumors,

15 although patient responses in pediatric ALK mutant neuroblastoma were less

16 encouraging (Mosse, Lim et al. 2013, Mosse, Voss et al. 2017). It is not clear whether this

17 relates to clinical factors unique to neuroblastoma or to issues of efficacy of inhibition of

18 ALK by crizotinib. The clinical data thus far motivates exploration of alternative strategies

19 in neuroblastoma, including ALK monotherapy with next-generation TKIs and combination

20 strategies (Berry, Luther et al. 2012, Moore, Azarova et al. 2014, Umapathy, El Wakil et al.

21 2014, Guan, Tucker et al. 2016, Infarinato, Park et al. 2016, Krytska, Ryles et al. 2016).

22 Other ALK TKIs include ceritinib, , and (Christensen, Zou

23 et al. 2007, Katayama, Khan et al. 2011, Sakamoto, Tsukaguchi et al. 2011, Marsilje, Pei et al.

24 2013, Chia, Mitchell et al. 2014, Johnson, Richardson et al. 2014, Katayama, Lovly et al.

25 2015) (for latest details of clinical trials employing ALK TKIs, please see Clinicaltrials.gov).

4

Downloaded from molecularcasestudies.cshlp.org on October 9, 2021 - Published by Cold Spring Harbor Laboratory Press

1 These ALK inhibitors bind slightly differently within the ATP-binding pocket of the ALK

2 kinase domain, show varying abilities to cross the blood brain barrier and have differing

3 profiles of inhibition for the wild-type ALK kinase domain compared with the various ALK

4 kinase mutants. These properties have important implications for potential treatment of ALK-

5 positive neuroblastoma in which ALK mutations are present in treatment-naïve tumors.

6 Ceritinib gained US Food and Drug Administration approval in 2014 following accelerated

7 review for the treatment of patients with ALK-positive (ALK+) metastatic NSCLC who have

8 progressed on, or are intolerant to, crizotinib ((FDA) 26th of May, 2017, (FDA) 29th of April,

9 2014, Shaw, Kim et al. 2014).

10 Here we report the robust response of an ALK-positive neuroblastoma patient to

11 ceritinib treatment. The patient received chemotherapy according to protocol after being

12 diagnosed with high-risk neuroblastoma but displayed severe hematological failure early after

13 initial treatment. This led to suspicion of Fanconi anemia (FA), that was confirmed by

14 chromosomal breakage assessment and identification of FANCA gene mutations. FA is a rare

15 recessive genetic disorder clinically characterized by congenital abnormalities and progressive

16 bone marrow failure (Kutler, Singh et al. 2003), although some patients may show only subtle

17 symptoms or no phenotype at all (Neveling, Endt et al. 2009). FA is caused by mutations in

18 one of at least 21 different FA genes encoding proteins that function in interstrand cross-link

19 and double strand DNA repair (Mamrak, Shimamura et al. 2017). Due to the impaired DNA

20 damage response, FA patients have significantly increased cancer susceptibility, particularly

21 to acute myeloid , head and neck squamous cell carcinoma (Kutler, Auerbach et al.

22 2003, Kutler, Singh et al. 2003) and more rarely, embryonic tumors such as Wilms tumor,

23 hepatoblastoma or neuroblastoma (Abbondanzo, Manz et al. 1986, Bissig, Staehelin et al.

24 2002, Berrebi, Lebras et al. 2006, Kopic, Eirich et al. 2011, Malric, Defachelles et al. 2015).

25 The management of cancer in FA patients is challenging as defective DNA repair leads to

5

Downloaded from molecularcasestudies.cshlp.org on October 9, 2021 - Published by Cold Spring Harbor Laboratory Press

1 hypersensitivity to both radiation and crosslinking agents such as mitomycin C and cisplatin

2 with increased risk of developing severe toxicity (Walsh, Chang et al. 2017)

3 (http://fanconi.org/index.php/publicaionts/guidelines). In parallel to identification of FANCA

4 gene mutations in this neuroblastoma patient, we also noted the presence of an ALK mutation

5 at residue I1171.

6 Preclinical analysis showed that the mutation of ALK-I1171 to threonine (T), which has

7 not previously been reported in neuroblastoma, generates a potent gain-of-function mutant.

8 PC12 cells expressing ALK-I1171T display ligand-independent activation of ALK, neurite

9 outgrowth and activation of downstream signaling. In addition, ALK-I1171T drives foci

10 formation in NIH3T3 cells. We were able to identify ceritinib as a next-generation ALK TKI

11 that effectively inhibits ALK-I1171T in a preclinical setting, and based on these experimental

12 data, the patient was treated with ceritinib. A dramatic response was observed and 7.5 months

13 after initiation of treatment, tumor material was surgically removed, exhibiting reduced Ki67

14 levels and upregulation of markers of differentiation. Based on this case, we suggest that

15 ceritinib presents a viable therapeutic option for patients with ALK-positive neuroblastoma .

16

17

6

Downloaded from molecularcasestudies.cshlp.org on October 9, 2021 - Published by Cold Spring Harbor Laboratory Press

1 Materials and methods

2

3 Patient

4 A 16-month-old boy presented at the pediatric emergency unit with neurologic symptoms

5 including facial palsy, impaired balance and strabismus. There was no past medical history of

6 note except for examination by a hand surgeon and a dysmelia team due to slightly deviant

7 thumbs which had been present since birth. At the emergency visit, exophthalmus and an

8 abdominal mass were also noted, and imaging using MIBG scintigraphy, CT scans and MRI

9 revealed a metastatic neuroblastoma with metastases in bone, lungs and intracranially (Fig. 1,

10 Fig. S1) but with no neuroblastoma detected in the bone marrow. Biological workup of the

11 diagnostic biopsy showed a number of unfavorable chromosomal aberrations, including 11q

12 loss, 17q gain, 2q loss, 4p loss,7q and 9q gain (Fig. 2). Treatment was initiated according to

13 the high-risk neuroblastoma protocol HR-NBL-1 SIOPEN (Ladenstein, Potschger et al. 2017)

14 (ethical permit 02-294). In response to this treatment, the patient developed protracted

15 neutropenia, anemia and thrombocytopenia necessitating repeated transfusions of erythrocytes

16 and platelets for almost 6 months (Fig. 3). Furthermore the patient experienced repeated

17 episodes of severe septicemia and typhlitis requiring intensive care. Based on the suspicion of

18 underlying FA (Walsh, Chang et al. 2017), chromosomal breakage assessment was perfomed

19 and found positive. Further genomic analysis revealed inherited germline mutations in the

20 FANCA gene, confirming FA diagnosis (Fig. 2B, Table S1). Detailed genomic analysis

21 further identified a somatic ALK-I1171T mutation in the patient, raising the possibility of

22 ALK TKI treatment (Fig. 2C, Table S1). Following preclinical investigation of the ALK-

23 I1171T mutation, a clinical multidisciplinary conference, an Institutional Ethical Board

24 meeting and a Clinical Ethical Board meeting endorsed application for a Medical Products

25 Agency (MPA) license (according to LVFS 2008:1). In addition, written informed consent

7

Downloaded from molecularcasestudies.cshlp.org on October 9, 2021 - Published by Cold Spring Harbor Laboratory Press

1 from the patient’s guardians was obtained and the patient was included in the

2 compassionate use Individual Patient Program for ceritinib (CLDK378A2003M). Sampling

3 and analyses of tumor tissues and linkage to clinical information were performed according to

4 the ethical permit 2009/1369-31/1. Written informed consent was obtained from the

5 patient’s guardians allowing scientific research publication.

6

7 Immunohistochemistry

8 Staining for NB84 (Novocastra, NCL-NB84), NF (DAKO, 2F11) and Ki-67 (DAKO, MIB-1)

9 was performed using a DAKO Autostainer. Slides were rehydrated with Xylene followed by a

10 series of alcohol dilutions and a rinse in buffer (DAKO 8007). PTLINK (DAKO) was used

11 for enzyme antigen retrieval. Endogenous enzyme block was performed for 5 minutes with

12 EnVision FLEX Peroxidase-Blocking Reagent (DAKO). The antibodies (diluted at DAKO

13 Company) were incubated at room temperature for 20 minutes. After 5 minutes rinse in buffer

14 the labeled polymer, EnVision FLEX/HRP (DAKO), was applied and incubated for 20

15 minutes with two additional rinses before the slides were incubated with substrate-

16 chromogen, Substrate Working Solution (DAKO), for 10 minutes. Slides were ultimately

17 rinsed and counterstained in EnVision FLEX Hematoxylin prior to mounting.

18

19 Genomics profile with SNP array

20 Microarray analyses of DNA from the tumor samples were performed using Affymetrix

21 Human Cytoscan High Density arrays essentially as described earlier (Fransson, Ostensson et

22 al. 2016). For primary data analysis the GDAS software (Affymetrix) was used, while

23 genomic profiles were generated using CNAG (Copy Number Analyzer for

24 AffymetrixGeneChip Mapping arrays) version 3.3 (Genome Laboratory, Tokyo University;

25 http://www.genome.umin.jp; 11).

8

Downloaded from molecularcasestudies.cshlp.org on October 9, 2021 - Published by Cold Spring Harbor Laboratory Press

1

2 Detection of ALK mutation with massive parallel DNA sequencing

3 DNA was extracted from tumor sample using standard procedures and evaluated through

4 fluorometric quantitation and DNA integrity assessment on Agilent Tapestation (Agilent,

5 Santa Clara, CA) prior to exome sequencing. Exome sequencing was performed at GATC

6 (GATC, Konstanz, Germany) through paired-end sequencing (101 bp read length) on

7 Illumina platforms after enrichment with Agilent SureSelect Human All Exon v5 enrichment

8 giving an average coverage of 85X (sequencing details are supplied in supplemental

9 information file: ‘Exome sequencing’). Alignment against hg19 was performed using BWA

10 with GATK local realignment followed by SNV calling using SNPeff and Annovar

11 annotation. Visualization of mapped reads and manual QC of called variants was done

12 through IGV (Robinson, Thorvaldsdottir et al. 2011). Only variants located at regions covered

13 by at least 10 unique reads were kept. A systematic filtering approach was used to identify

14 critical variants by removal of common variants (e.g. showing an allele frequency above

15 0.005 in 1000 genomes or exome variant server) as well as excluding all synonymous variants

16 or variants in non-coding regions except those affecting canonical splice sites. PolyPhen 2

17 (PP2) and SIFT were used for prediction of functional relevance of called SNVs. Sanger

18 sequencing of the patient’s constitutional and tumor DNA verified presence of a somatic ALK

19 mutation in the tumor. Sanger sequencing of ALK exons was performed as described

20 previously (Caren, Abel et al. 2008). All genomic positions are specified according

21 hg19/GRCh37.

22

23 Generation of human ALK mutant constructs in PC12 cells

24 The ALK-I1171T mutation was generated based on pcDNA3-ALK-WT (NM_004304.3) by

25 Eurofins Genomics (Ebersberg, Germany). ALK-F1174L has been described previously

9

Downloaded from molecularcasestudies.cshlp.org on October 9, 2021 - Published by Cold Spring Harbor Laboratory Press

1 (Martinsson, Eriksson et al. 2011). All mutations generated in the kinase domain were

2 confirmed by sequencing from both directions.

3

4 Neurite outgrowth assay

5 PC12 (2 x 106) cells were co-transfected with 0.5 µg of empty pcDNA3 vector, pcDNA3-

6 ALK-WT, pcDNA3-ALK-I1171T or pcDNA3-ALK-F1174L respectively together with 0.5

7 µg of pEGFP-N1 (Clonetech) as indicated by electroporation using Amaxa electroporator

8 (Amaxa Biosystems) and Ingenio electroporation solution (Mirus Bio LCC). After

9 electroporation, cells were kept in minimum essential medium (MEM) supplemented with 7%

10 horse serum and 3% fetal bovine serum and seeded into 24-well plates. For samples

11 stimulated with ALK ligand, purified ALKAL1 (FAM150A) protein was added to the well

12 with a final concentration of 1µg/ml (Guan, Umapathy et al. 2015). After 48 hours of

13 incubation, the fraction of Green fluorescent protein (GFP)-positive and neurite carrying cells

14 versus GFP-positive cells was observed under a Zeiss Axiovert 40 CFL microscope. To be

15 judged as a neurite carrying cell, the neurite of the cell should be at least twice the diameter of

16 a normal cell body. Experiments were performed in triplicate, and each sample within an

17 experiment was performed in triplicate.

18

19 Transformation assay

20 NIH 3T3 cells (5 × 104) were seeded into collagen-coated 12-well plates and transfected with

21 0.55μg of pcDNA3 vector, pcDNA3-ALK-WT, pcDNA3-ALK-I1171T, or pcDNA3-ALK-

22 F1174L respectively using Lipofectamine 2000 according to the manufacturer’s protocol

23 (Invitrogen). Twenty-four hours later, three fifths of the cells from each well were transferred

24 to wells in 6-well plates and kept in DMEM supplemented with 10% FBS and 0.5 mg/ml

10

Downloaded from molecularcasestudies.cshlp.org on October 9, 2021 - Published by Cold Spring Harbor Laboratory Press

1 G418 until the cells reached confluence. Thereafter, cells were kept in DMEM supplemented

2 with 5% FBS and 0.25 mg/ml G418 for another 10 days, with replacement of medium every

3 second day. Plates were washed with PBS and air-dried, fixed with methanol for 20 minutes,

4 and cells were stained with 0.2% crystal violet in 20% ethanol for 20 minutes, followed by

5 three short rinses in water.

6

7 Cell culture, viability, lysis, and immunoblotting

8 Cell viability was assessed as relative redox metabolic activity using aresazurin-based assay.

9 Cell lines employed were CLB-BAR, CLB-GE, CLB-GA, Kelly, SK-N-AS, SK-N-BE, SK-

10 N-DZ, IMR32 and CLB-PE neuroblastoma cells (Umapathy, Guan et al. 2017). Cells (4 ×104)

11 were plated on collagen-coated 48-well plates and treated with either ceritinib or crizotinib

12 with indicated concentrations. Cell viability was determined after 72 hours with 55μM

13 resazurin (Sigma, Stockholm, Sweden). After 3h at 37°C, the amount of metabolized

14 resazurin was analyzed as relative fluorescence with an Infinit200 plate reader (TEKAN,

15 Männedorf, Switzerland). Results were from one of three representative experiments, with

16 each experiment being performed in triplicate. GraphPad Prism 6.0 was used to calculate IC50

17 values by fitting data to a log (inhibitor concentration) versus normalized response (variable

18 slope) equation.

19 PC12 cells were transfected with 0.5 µg of empty pcDNA3 vector, pcDNA3-ALK-WT, or

20 pcDNA3-ALK variants as specified respectively by electroporation as described above. Cells

21 were serum-starved for 36 hours before lysis. For samples stimulated with ALK ligand, they

22 were treated with 1μg/ml of purified ALKAL1 protein for 30 minutes prior to lysis. Cells

23 were washed twice with ice-cold PBS prior to harvest in lysis buffer [25mmol/L Tris-Cl,

24 pH7.5, 150mmol/L NaCl, 1% (v/v) Triton X-100, 1mmol/L DTT, protease inhibitor cocktail

11

Downloaded from molecularcasestudies.cshlp.org on October 9, 2021 - Published by Cold Spring Harbor Laboratory Press

1 tablet (Roche)]. Cell lysates were cleared by centrifugation at 14,000 rpm for 15 minutes at

2 4°C. Samples were boiled in 1x SDS sample buffer and analyzed by immunoblotting. Primary

3 antibodies used for immunoblotting were: anti-pan-ERK (1:10,000), purchased from BD

4 Transduction Laboratories (Franklin Lakes, NJ), anti-pALK (Y1604) and anti-pERK1/2

5 (T202/Y204) from Cell Signaling Technology (Danvers, MA). Monoclonal antibody 135

6 (anti-ALK) was produced in the Hallberg laboratory against the extracellular domain of ALK

7 as described (Moog-Lutz, Degoutin et al. 2005, Schonherr, Ruuth et al. 2012). Horseradish-

8 peroxidase-conjugated secondary antibodies goat anti-rabbit IgG and goat anti-mouse IgG

9 (1:5,000) were from Thermo Scientific (Waltham, MA).

10

11 ALK inhibitor profiles on ALK variants

12 PC12 cells (2×106) were transfected with 1.0 μg of pcDNA3-ALK-WT, or pcDNA3-ALK

13 variants as described (Guan, Tucker et al. 2016, Siaw, Wan et al. 2016). Cells from three

14 electroporations were pooled together, mixed and equally seeded into 9 wells of one 24-well

15 plate. After 24-36 hours culture, cells were treated with serial dilutions of the indicated

16 inhibitors for 1 hour before lysis. Cells were washed with cold 1X PBS and lysed with 1X

17 SDS sample buffer and samples were boiled at 95ºC for 5 minutes. Phospho-ALK (Y1604)

18 antibody was used to detect ALK phosphorylation and ALK mAb135 was used to detect total

19 ALK. The intensity of pALK (Y1604) and total ALK bands was quantified with Image Studio

20 Lite 3.1 software. Data were normalised to the 0 nM inhibitor samples. GraphPad Prism 6.0

21 was used to calculate IC50 values by fitting data to a log (inhibitor concentration) vs.

22 normalized response (variable slope) equation.

23

24 Phosphoproteomics

12

Downloaded from molecularcasestudies.cshlp.org on October 9, 2021 - Published by Cold Spring Harbor Laboratory Press

1 Tyrosine and serine/threonine phosphorylation profiling of neuroblastoma cells (CLB-BAR,

2 CLB-GE and SK-N-AS) in the presence or absence of ceritinib (200 nM for 60 mins) was

3 undertaken by immunoaffinity purification with P-Tyr-100 or P-Ser/Thr-antibodies

4 accordingly (Cell Signaling Technology, Danvers, MA, USA). Cells were lysed in lysis buffer

5 (20 mM Hepes pH 8.0, 9M UREA, 1 mM sodium orthovanadate, 2.5 mM sodium

6 pyrophosphate and 1 mM β-glycerol-phosphate) and approximately 20 mg protein per sample

7 subjected to LC-MS/MS. This was performed as previously described (Sattu, Hochgrafe et al.

8 2013). Phosphorylation responses were quantified for each site (tyrosine, serine or threonine)

9 under analysis as log2 fold change (FC) values of ceritinib treated/untreated control counts. If

10 a specific site was measured in multiple peptides, its median log2 FC value was used for

11 analysis. Protein sites were considered differentially phosphorylated when their absolute log2

12 FC values were higher than 1.5. A gene set enrichment analysis (GSEA) was performed using

13 Fisher’s exact test and false discovery rate correction using the Benjamini Hochberg method

14 (Benjamini and Hochberg 1995). Reactome pathway information was downloaded from the

15 Molecular Signatures Database v5.2 (Subramanian, Tamayo et al. 2005). The differentially

16 phosphorylated proteins were mapped to the InWeb_InBioMap protein-protein interaction

17 (PPI) network (Li, Wernersson et al. 2017). For the analysis of related TRKs, a list of 40

18 different TRKs was downloaded from HGNC (Gray et al., 2015).

19

20

13

Downloaded from molecularcasestudies.cshlp.org on October 9, 2021 - Published by Cold Spring Harbor Laboratory Press

1 Results

2

3 Patient presentation

4 A 16-month-old boy presenting with neurologic symptoms at the pediatric emergency

5 unit was found to have metastatic neuroblastoma stage M (INRGSS) (Fig. 1). There was no

6 previous family history of neuroblastoma. SNP-microarray analysis of genomic DNA from

7 initial biopsy showed no MYCN-amplification but revealed several unfavorable segmental

8 alterations (Fig. 2A). COJEC chemotherapy induction was started according to HR-NBL-1

9 SIOPEN (Ladenstein, Potschger et al. 2017) and the patient received one A course (day 0;

10 vincristine, carboplatin, etoposide), one B course (day 10; vincristine, cisplatin) and a partial

11 C course (day 20; vincristine, etoposide, cyclophosphamide), before chemotherapy had to be

12 discontinued because of severe bone marrow toxicity with anemia, neutropenia and

13 thrombocytopenia. For the ensuing four months, the patient was continuously hospitalized and

14 suffered repeated episodes of septicemia, intestinal hemorrhage and typhlitis as well as

15 respiratory failure requiring intensive care. Because of the unusual severity of the toxicity,

16 and also because the patient displayed moderate radial dysplasia with deviant thumbs

17 bilaterally, suspicion of Fanconi anemia was raised. A clinical chromosome breakage

18 assessment was found positive. A more detailed genomic analysis then revealed inherited

19 mutations in the FANCA gene; one novel missense variant at genomic position

20 chr16:89831348 (NM_000135.2; c.2728C>T, p.L910F) predicted to be deleterious and

21 probably damaging by SIFT and PP2 respectively and one splice site mutation at the

22 acceptor site of intron 42 at genomic position chr16:89805118, (NM_000135.2; c.4261-

23 2A>C) (Fig. 2B, Table S1). The L910F variant is not present in either ClinVar or

24 HGMD and to best of our knowledge, this is the first report of a L910F variant in

25 association with FA. A third rare missense variant at chr16:89871709 (c.688G>A; p.V230I),

14

Downloaded from molecularcasestudies.cshlp.org on October 9, 2021 - Published by Cold Spring Harbor Laboratory Press

1 was detected in the patient although predicted to be benign by SIFT and PP2, indicating a

2 non-pathogenic function. This is supported by a recent study showing that the V230I variant

3 is localized to the nucleus and does not confer sensitivity to mitomycin C (Kimble, Lach et al.

4 2017). The fact that the child presents as a compound heterozygote for FANCA

5 (p.L910F/c.4261-2A>C) is a likely cause for the above described toxicity (Fig. 2B, Table S1).

6 Further use of chemotherapy, or irradiation, was advised against in this patient due to the

7 DNA repair defects in Fanconi anemia. Since initial genomic analysis also identified an ALK-

8 I1171T mutation in the patient (Fig. 2C, Table S1), ALK TKI therapy was considered and

9 experimentally investigated preclinically for this patient.

10

11 ALK-I1171T is a gain-of-function ALK mutation

12 Exome sequence analysis of a tumor biopsy sample revealed a heterozygous mutation

13 of ALK (chr2:29445213) in exon 22; (NM_004304.4; c.3512T>C) leading to a missense

14 mutation I117IT (Fig. 2C, Table S1). Sanger sequencing of tumor and patient’s

15 corresponding constitutional DNA indicated that the ALK variant was caused by a somatic

16 event. I1171 is located in the ALK kinase domain in close proximity to F1174, one of the

17 mutational hotspots of ALK in neuroblastoma that is localized in the αC-helix in the N-

18 terminal lobe of the kinase (Fig. 4A). Mutation of I1171 to either N/T or S has previously

19 been described in ALK TKI-treated patients with ALK fusion-positive NSCLC (in the EML4-

20 ALK and HIP1-ALK fusion proteins) (Katayama, Friboulet et al. 2014, Ou, Klempner et al.

21 2014, Toyokawa, Hirai et al. 2014), resulting in tumor resistance.

22 This is the first report of mutation of ALK-I1171T in neuroblastoma, although mutation

23 of ALK-I1171N has been reported (Mosse, Laudenslager et al. 2008, Bresler, Weiser et al.

24 2014). Intriguingly, the residue I1171 contributes to the regulatory spine (Kornev and Taylor

25 2015) (Fig. 4A), prompting us to experimentally validate its behavior in the context of the full

15

Downloaded from molecularcasestudies.cshlp.org on October 9, 2021 - Published by Cold Spring Harbor Laboratory Press

1 length ALK RTK. To evaluate the ALK-I1171T mutant we employed a PC12 cell culture

2 system for expression of either wild type or ALK-I1171T. Wild type ALK can be activated

3 with the ALKAL1 ligand (FAM150A, AUGβ) (Guan, Umapathy et al. 2015, Reshetnyak,

4 Murray et al. 2015) leading to extensive tyrosine phosphorylation of the receptor and

5 activation of downstream targets, such as ERK1/2 (Fig. 4B). In contrast to wild type ALK we

6 observed ligand-independent auto-trans-phosphorylation of the ALK-I1171T mutant together

7 with activation of the downstream target ERK1/2 (Fig. 4B).

8 We next investigated whether the ALK-I1171T mutant was capable of stimulating

9 neurite outgrowth in the PC12 cell line. PC12 cells are a clonal rat adrenal

10 pheochromocytoma cell line with enteric cell origin, which differentiate and extend neurites

11 upon extended ERK1/2 stimulation (Cowley, Paterson et al. 1994). We and others have

12 previously shown that activation of ALK triggers differentiation of PC12 cells into

13 sympathetic-like neurons, a process characterized by extension of neurites (Fig. 4C) (Motegi,

14 Fujimoto et al. 2004, Schonherr, Ruuth et al. 2011). Expression of wild type ALK mediates

15 only a low level of neurite outgrowth in this assay (Fig. 4C). In contrast, expression of the

16 ALK-I1171T mutant results in robust neurite outgrowth to a level similar to that observed for

17 the well characterized ALK-F1174L mutant employed as positive control.

18 Finally, we investigated whether the human ALK-I1171T mutant displays transforming

19 potential in NIH3T3 cells. Expression of either the positive control human ALK-F1174L or

20 the ALK-I1171T mutant led to formation of foci of transformed cells over the background

21 monolayer (Fig. 4D), whereas expression of the wild type human ALK receptor was unable to

22 mediate foci formation. Thus, ALK-I1171T exhibits intrinsic transforming activity in a focus

23 formation assay, in addition to ligand-independent activation of downstream targets and

24 neurite outgrowth.

25

16

Downloaded from molecularcasestudies.cshlp.org on October 9, 2021 - Published by Cold Spring Harbor Laboratory Press

1 Ceritinib abrogates growth in ALK-addicted neuroblastoma cell lines.

2 Ceritinib is a small molecule ALK TKI that has been extensively explored for ALK-

3 positive NSCLC and is FDA approved for clinical use in that setting. It has not yet been

4 reported as being employed clinically in the context of ALK mutations in neuroblastoma.

5 Previous analysis of the I1171T mutation in NSCLC (arising in response to sequential

6 crizotinib and alectinib treatment) noted that the I1171 mutation is relatively insensitive to

7 inhibition by crizotinib and alectinib but was still sensitive to other ALK TKIs such as

8 TAE684 and ceritinib (Katayama, Friboulet et al. 2014). We therefore assembled a panel of

9 neuroblastoma cell lines harboring either mutated or wildtype ALK in combination with other

10 genetic aberrations, CLB-GE, CLB-GA, CLB-BAR, CLB-PE, IMR32, Kelly, SK-N-AS, SK-

11 N-DZ and SK-N-BE (Table 1) to examine the effect of ceritinib on ALK activity and cell

12 proliferation in a neuroblastoma setting (Kohl, Gee et al. 1984, Combaret, Turc-Carel et al.

13 1995, Schleiermacher, Janoueix-Lerosey et al. 2003, Cazes, Louis-Brennetot et al. 2013,

14 Fransson, Hansson et al. 2015, Umapathy, Guan et al. 2017). In this analysis we also

15 compared the ability of ceritinib to inhibit proliferation of neuroblastoma cells with the well-

16 characterized clinical ALK TKI crizotinib (Christensen, Zou et al. 2007), treating cells with

17 increasing doses of either crizotinib or ceritinib. Proliferation of ALK-addicted cell lines such

18 as CLB-BAR, CLB-GE, CLB-GA and Kelly was inhibited in a dose-dependent manner by

19 both crizotinib and ceritinib (Table 1). Of the neuroblastoma cell lines tested, growth of CLB-

20 BAR and CLB-GE was inhibited in a manner similar to that previously shown for crizotinib

21 (Schonherr, Ruuth et al. 2012), although the observed IC50 values differ up to 2-fold. IC50

22 values were similar for both ceritinib and crizotinib in CLB-GA and Kelly cells with values of

23 approximately 110 nM and 330 nM, respectively (Table 1). Importantly, we did not observe

24 inhibition of proliferation in non-ALK-addicted cell lines such as CLB-PE, SK-N-AS SK-N-

25 BE, SK-N-DZ, which carry other driver mutations. IMR32 cells, which express wild type

17

Downloaded from molecularcasestudies.cshlp.org on October 9, 2021 - Published by Cold Spring Harbor Laboratory Press

1 ALK and respond to stimulation with ALKAL ligands (Guan, Umapathy et al. 2015), are

2 more sensitive to crizotinib than ceritinib, although IC50 values are high, more than 350 nM

3 (Table 1). Thus, these preclinical results show that while activity of the ALK-dependent

4 neuroblastoma cells tested here is inhibited by crizotinib, treatment with ceritinib has greater

5 efficacy.

6

7 Ceritinib effectively inhibits the crizotinib resistant ALK-I1171T mutant in PC12 cells.

8 To determine which ALK TKI would be suitable in this clinical case we examined the

9 ability of different ALK inhibitors to abrogate ALK-I1171T activity. As readout for ALK

10 activity we employed phosphorylation of ALK Y1604, which reflects ALK activation, in

11 PC12 cells (Chand, Yamazaki et al. 2013). PC12 cells expressing ALK-I1171T were treated

12 with serial dilution of ALK inhibitors crizotinib, ceritinib, lorlatinib and brigatinib and Y1604

13 phosphorylation levels were quantified. We observed that full length ALK-I1171T was more

14 resistant to crizotinib than other inhibitors as measured by Y1604 phosphorylation (Fig. 4E).

15 The IC50 of crizotinib for ALK-I1171T in this analysis was 193 ± 57 nM, approximately 11

16 times that of ceritinib for ALK-I1171T (Fig. 4F). The next generation ALK TKIs also showed

17 strong anti-ALK-I1171T activity. The observed IC50 values of ALK-I1171T for brigatinib and

18 lorlatinib were in a similar range (6.8 nM to 7.5 nM), approximately 1/28th of the crizotinib

19 IC50 for ALK-I1171T (Fig. 4F). Taken together, our results indicate that ceritinib as well as

20 the third generation TKIs lorlatinib and brigatinib represent experimentally well supported

21 choices for treatment of tumor(s) harboring the ALK-I1171T mutation.

22 To extend our analysis we examined the activity of ceritinib to inhibit a range of gain-

23 of-function ALK variants expressed in PC12 cells, employing ALK Y1604 phosphorylation

24 as readout (Fig. S2; includes all loading ALK control blots for this experiment). In this

25 analysis crizotinib abrogates wild type ALK phosphorylation with an IC50 of approximately

18

Downloaded from molecularcasestudies.cshlp.org on October 9, 2021 - Published by Cold Spring Harbor Laboratory Press

1 18 nM, which is similar to earlier reports (Schonherr, Ruuth et al. 2011, Siaw, Wan et al.

2 2016). In comparison, ceritinib blocks ALK Y1604 phosphorylation with a 3-fold lower IC50

3 level, of approximately 5 nM. The IC50 values for crizotinib inhibition of ALK Y1604

4 phosphorylation of the various ALK mutations tested here are similar to earlier reports,

5 highlighting a requirement for higher crizotinib dosage for effective inhibition of ALK-

6 I1171N (Table 2) (Schonherr, Ruuth et al. 2011, Guan, Tucker et al. 2016, Siaw, Wan et al.

7 2016). ALK-G1269A has only been described in EML4-ALK fusions arising in NSCLC

8 patients that have developed resistance to ALK TKI treatment (Doebele, Pilling et al.

9 2012), but is included here as reference in the context of full length ALK.

10 Our extensive examination of the inhibition profiles of both crizotinib and ceritinib on

11 the different ALK mutations reveals one important exception - ALK-I1171T. We observe that

12 inhibition of ALK-I1171T by ceritinib exhibits an IC50 value (17.4 ± 3.1, Fig. 4F) that is not

13 too far from that of wild type ALK (5.3 ± 0.2) (Table 2). This is in stark contrast to the ALK-

14 I1171N mutation at the same residue, which displays resistance to both ceritinib (46.6 ± 0.16)

15 and crizotinib (139.5 ± 13.8) when compared with wild type ALK (Table 2). In summary,

16 ceritinib was active in a range comparable to crizotinib as measured by inhibition of ALK

17 phosphorylation/activation in PC12 cells with one exception, the ALK-I1171T mutant which

18 was insensitive to crizotinib but efficiently inhibited by ceritinib.

19

20 Phosphoproteomic profiling of ceritinib in different neuroblastoma cell lines

21 Phosphoproteomic profiling has clinical value since it can be translated into prognostic

22 biomarkers, which can be employed to identify functional downstream targets as

23 combinatorial treatment target options together with ceritinib. Here we determined the

24 difference in the phosphoproteomic patterns upon treatment with ceritinib in ALK-addicted

25 (CLB-BAR and CLB-GE) neuroblastoma cell lines with that of a non-ALK-addicted (SK-N-

19

Downloaded from molecularcasestudies.cshlp.org on October 9, 2021 - Published by Cold Spring Harbor Laboratory Press

1 AS) neuroblastoma cell line (Fig. 5A). The CLB-BAR and CLB-GE cell lines harbor

2 activating ALK mutations, are ALK/MYCN amplified, lack p53/RAS mutations and display

3 high levels of NF1 (Umapathy, Guan et al. 2017). The SK-N-AS cell line harbors an

4 activating mutation in the NRAS gene and exhibits a down-regulated NF1 expression leading

5 to an activation of the RAS-MAPK pathway. SK-N-AS has a 11q deletion, is non-MYCN

6 amplified, has no p53/ALK mutations and undetectable levels of ALK protein (Umapathy,

7 Guan et al. 2017). SK-N-AS was employed as a control cell line for off-target effects of the

8 inhibitors employed, since ALK protein is undetectable in this cell line with no MYCN

9 amplification.

10 To identify sites with altered phosphorylation associated with ALK activity, we utilized

11 an immunoaffinity-coupled LC-MS/MS approach by treating neuroblastoma cell lines with

12 ceritinib. Prior to MS, lysates were examined for ALK-, ERK- and AKT-activity, which have

13 previously been shown to be reduced upon ALK TKI treatment for 1 hour (Fig. S3). We

14 observed reduced phosphorylation levels of ALK and ERK in response to ceritinib, validating

15 response to inhibitor treatment (Fig. S3). Phosphoproteomic analysis of neuroblastoma cell

16 lines (CLB-BAR, CLB-GE and SK-N-AS) identified 2,223 phosphothreonine sites, 2,583

17 phosphotyrosine sites and 5,013 phosphoserine residue sites in 3,345 different proteins (Fig.

18 5A). All previously observed 11 ALK phosphotyrosine sites were identified and showed

19 drastically reduced phosphorylation signals in both ALK-addicted neuroblastoma cell lines

20 (i.e. CLB-BAR and CLB-GE) after ceritinib treatment. The strongest dephosphorylation

21 signals were measured at ALK tyrosine sites 1096, 1278, 1282 and 1283 (Fig. 5B). The latter

22 three sites were previously reported as important residues in the ALK activation loop

23 (Donella-Deana, Marin et al. 2005, Rush, Moritz et al. 2005, Rikova, Guo et al. 2007, Tartari,

24 Gunby et al. 2008, Wang, Wu et al. 2010, Sattu, Hochgrafe et al. 2013, Guan, Yamazaki et al.

25 2017). As expected, no phosphorylation signal was observed in the control SK-N-AS cell line.

20

Downloaded from molecularcasestudies.cshlp.org on October 9, 2021 - Published by Cold Spring Harbor Laboratory Press

1 Furthermore, this strong dephosphorylation response was only observed for ALK and not for

2 any other RTK (Fig. S4). In CLB-BAR, 182 proteins were dephosphorylated and 52 proteins

3 were phosphorylated upon treatment with ceritinib (Fig. 5C). Similarly, in CLB-GE, 200

4 proteins were dephosphorylated and 120 proteins were phosphorylated (Fig. 5C). Treatment

5 with ceritinib in SK-N-AS revealed only 35 proteins that were dephosphorylated and 20

6 proteins that were phosphorylated. Few similarities were found between the RAS-addicted

7 SK-N-AS cell line and the ALK-addicted CLB-BAR/GE cell lines and, while phosphorylation

8 responses were specific between all cell lines with little overlap, 60 proteins were

9 dephosphorylated in both CLB-BAR and CLB-GE but not SK-N-AS (Fig. 5D and Table S2).

10 Assuming that these 60 proteins represent the most specific response to ceritinib, they were

11 employed for further analysis. To illustrate downstream phosphorylation targets of ALK

12 signaling activity that are inhibited upon treatment with ceritinib in ALK-addicted

13 neuroblastoma cells, we mapped the dephosphorylated proteins to a protein-protein

14 interaction network (Fig. 5E). This network analysis highlighted FRS2, IRS2, ERK1/2, AKT

15 family members, SHC family members, SOS1 and GAB1/2 as key components of the

16 underlying signaling pathways. Many of these proteins are known to be active in , NGF

17 and FGFR receptor signaling pathways, as confirmed by a gene set enrichment analysis (Fig.

18 5F). Similar results were found when independently focusing on the 182 and 200

19 dephosphorylated proteins of the CLB-BAR and CLB-GE cell lines respectively (Figs. S5 and

20 S6).

21

22 Patient response to ceritinib.

23 Based on the experimental data presented here that indicated a potentially poor response

24 to crizotinib, as well as evidence for FA prohibiting use of conventional chemotherapy or

25 irradiation, the patient was started on ceritinib monotherapy as soon as severe acute toxicity

21

Downloaded from molecularcasestudies.cshlp.org on October 9, 2021 - Published by Cold Spring Harbor Laboratory Press

1 had subsided (initiated 9.5 months after diagnosis, indicated by *; Fig. 3A). Initial dosage was

2 225 mg once daily (450 mg/m2 body surface area) for a period of 10.5 months, after which

3 dosage was increased to 540 mg/m2 body surface area. Hematological counts were stabilized

4 at normal levels (Fig. 3A). After the first 4 week cycle, elevated urinary catecholamines had

5 decreased to levels below the upper reference limit (Fig. 3B). Other than mild gastrointestinal

6 side effects, no toxicity was observed, and the child was able to lead a normal life (Kim,

7 Mehra et al. 2016). After 6.5 months of ceritinib treatment, the primary tumor had decreased

8 in size (-43.6% according to MRI with volumetric measurements, Fig. 1) and the remaining

9 CNS metastasis showed response by MRI (Fig. S1), although not as sensitive as CT scans

10 performed at diagnosis and after 21 months of treatment showing complete metastatic

11 remission (Fig. 1). The remaining primary was radically removed surgically together with the

12 left adrenal gland at 7.5 months from start of ceritinib treatment. The tumor was subjected to

13 immunohistochemical analysis with both proliferation and differentiation markers. The

14 treated tumor displayed a large proportion of calcification and non-malignant stroma, reduced

15 cell proliferation as measured by Ki-67 positivity (5% versus 27% before treatment) as well

16 as hallmarks of differentiation as assessed with NB84 and NFP (Fig. 6). The overall

17 morphology of the primary tumor indicated differentiation in response to treatment,

18 resembling a ganglioneuroblastoma, and was rich in schwannian stroma containing scattered

19 foci of tumor cells of varying degrees of maturation to ganglion cells. Complete clinical

20 evaluation with CT and MRI one year after surgery and 21 months after initiation of ceritinib

21 treatment showed no residual tumor in the abdomen, and complete resolution of metastases at

22 all sites (Fig. 1). MIBG-I123 scintigraphy showed normal findings, indicating completely

23 resolved tumor activity (Fig. 1B). After 34 months of continuous ceritinib treatment, the child

24 is in continous complete remission (Park, Bagatell et al. 2017).

22

Downloaded from molecularcasestudies.cshlp.org on October 9, 2021 - Published by Cold Spring Harbor Laboratory Press

1 Discussion

2 Here we report the case of a metastatic high-risk neuroblastoma that was treated

3 according to HR_NBL-1 SIOPEN protocol and developed severe toxicity including protracted

4 neutropenia, anemia and thrombocytopenia that required intensive clinical care for 4 months.

5 During this time genetic analyses revealed inherited biallelic sequence variants in FANCA.

6 This confirmed the initial suspicion of FA as likely cause of the observed toxicity and a

7 potential contributing factor to tumor development. Genomic analysis also detected a somatic

8 ALK-I1171T mutation that has not previously been described in neuroblastoma. This

9 presented a clinical option to treat the child, whose FA status precluded further chemotherapy

10 or irradiation, with targeted inhibition of ALK. Therefore an extensive preclinical

11 experimental investigation of the ALK-I1171T mutant was performed.

12 ALK-I1171T exhibits robust gain-of-function activity in several different systems, such

13 as triggering neurite outgrowth in PC12 cells, activation of validated downstream ALK targets

14 and transformation competence in NIH 3T3 cells. A critical part of this investigation was

15 defining the sensitivity of ALK-I1171T to available ALK TKIs, including crizotinib, ceritinib,

16 brigatinib and lorlatinib. Of these, the next-generation inhibitors ceritinib, brigatinib and

17 lorlatinib, which have been explored in preclinical neuroblastoma models (Guan, Tucker et al.

18 2016, Wood, Krytska et al. 2016), were effective in inhibition. Most importantly, crizotinib

19 was a very poor inhibitor of ALK-I1171T activity and does not efficiently cross the blood

20 brain barrier. This is in agreement with data from non-small cell lung cancer patients, where

21 resistance mutations in ALK fusions have been reported at I1171, although in these cases to

22 Aspargine and Serine, and not as yet to Threonine (Ou, Klempner et al. 2014, Ou, Greenbowe

23 et al. 2015). We also performed a preclinical LC-MS/MS analysis of ceritinib activity in

24 neuroblastoma confirming a strong dephosphorylation response at all tyrosine residues that

25 are known to be involved in the ALK activation loop, but not in any other RTK, indicating the

23

Downloaded from molecularcasestudies.cshlp.org on October 9, 2021 - Published by Cold Spring Harbor Laboratory Press

1 specificity of the response. The downstream signaling response included FRS2, ERK1/2,

2 AKT family members, SHC family members and GAB1/2 amoung others and was similar to

3 insulin, NGF and FGFR signaling pathways. Based on the collective preclinical information,

4 the child was included in the compassionate use individual patient program for ceritinib.

5 The patient responded strongly to ceritinib with minimal side effects allowing the child

6 to lead a normal life. To date, the patient is in complete clinical remission after continuous

7 ceritinib mono-therapy treatment for 34 months. It is unclear to what extent the efficacy of

8 ceritinib treatment may have been influenced by the initial chemotherapy treatment which

9 induced partial remission. One possibility that should be considered, is that the initial

10 chemotherapy treatment sensitized the tumor to the subsequent ALK TKI treatment. A

11 preclinical report investigating combination of crizotinib with commonly employed

12 chemotherapy showed synergistic cytotoxicity and increased in neuroblastoma cell

13 lines with ALK aberrations, supporting this senario (Krytska, Ryles et al. 2016). However, it

14 is important to note that at initiation of ceritinib treatment the patient displayed significant

15 active disease, as shown by persistant elevated urine catecholamine markers, metastatic

16 disease and remaining primary tumor tissue, which all responded strongly and rapidly to ALK

17 TKI therapy with finally complete clinical remission at all sites.

18 In conclusion, we have shown the importance of comprehensive genetic profiling

19 combined with preclinical investigation, which was conducted during a forced drug holiday of

20 a neuroblastoma patient later discovered to present with FA due to FANCA mutations and an

21 ALK mutation. A compassionate use protocol with ceritinib has shown a clear benefit for the

22 patient who is in complete clinical remission with clearance of all metastatic sites 34 months

23 from start of targeted ceritinib treatment. After the intial 7.5 months of treatment with

24 ceritinib the primary tumor had decreased in size and was removed surgically. The removed

25 tumor displayed a large amount of calcification and non–malignant stroma, reduced

24

Downloaded from molecularcasestudies.cshlp.org on October 9, 2021 - Published by Cold Spring Harbor Laboratory Press

1 proliferation index and hallmarks of differentiation. Here we show that mono-therapy with

2 next generation ALK TKIs can be effective against metastatic high-risk neuroblastoma

3 carrying an activating ALK mutation, confirming ceritinib as a viable therapeutic option for

4 ALK-positive neuroblastoma patients.

5

25

Downloaded from molecularcasestudies.cshlp.org on October 9, 2021 - Published by Cold Spring Harbor Laboratory Press

1 Additional information

2

3 Data deposition and Access

4 The detected DNA variants in FANCA and ALK have been submitted to ClinVar

5 (http://www.ncbi.nlm.nih.gov/clinvar/) and can be found under accession numbers

6 SCV000777897 - SCV000777901.

7

8 Ethics Statement

9 Treatment was initiated according HR-NBL-1 SIOPEN protocol (permit 02-294). A Clinical

10 Ethical Board meeting and an endorsed license application from the MPA license (according

11 to LVFS 2008:1) supported LDK378 compassionate use. Written informed consent from the

12 patient's guardians was obtained and the patient was included in the Novartis compassionate

13 use Program for ceritinib (CLDK378A2003M). Tumor analyses and linkage to clinical

14 information were performed according to the ethical permit 2009/1369-31/1. Written

15 informed consent was obtained from the patient's guardians allowing the scientific research

16 publication and are filed with the case notes.

17

18 Acknowledgements

19 This work has been supported by grants from the Swedish Cancer Society

20 (TMCAN2015/794; BHCAN15/775; RHP CAN15/391; PK CAN16/3625), the Swedish

21 Childhood Cancer Foundation (TM PR2016-0147; BH 2015-80, and 2014-150; RHP 2015-

22 96; PK PR2014-0084; JG 2016-0011; SF 15-0061; DT 12-009 and 12-002), the Swedish

23 Research Council (RHP 2015-04466; BH 521-2012-2831; TM 521-2014-3031; PK 2014-

24 3036); the Swedish Foundation for Strategic Research (RB13-0204, www.nnbcr.se) and the

25 Göran Gustafsson Foundation (RHP2016). D.T was supported by a PhD-student program

26

Downloaded from molecularcasestudies.cshlp.org on October 9, 2021 - Published by Cold Spring Harbor Laboratory Press

1 (NC2012-0026), J.G. was supported by a Postdoctoral Research Fellow Position (TJ2016-

2 0088) and S.F. was supported by a Research Assistant Fellowship (14-0064), by the Swedish

3 Childhood Cancer Foundation. The CLB-BAR, CLB-GE, CLB-GA, and CLB-PE cell lines

4 used in this study were kindly provided by Valérie Combaret under an MTA agreement with

5 the Center

6 Léon Bérard laboratory (Lyon, France). Novartis supplied ceritinib free of charge within the

7 compassionate use program “Ceritinib IPP for ALK-Positive IMT, ALCL and Neuroblastoma

8 Pediatric Patients”.

9

10

11

12

27

Downloaded from molecularcasestudies.cshlp.org on October 9, 2021 - Published by Cold Spring Harbor Laboratory Press

1 Figure Legends

2

3 Figure 1. Patient tumor imaging. Disease at initial diagnosis and after

4 in full clinical remission (A-D). (A) Whole body MIBG-I123 scintigraphy showing extensive

5 metastatic disease at diagnosis of high-risk neuroblastoma (upper panels). Left panel –

6 anterior view; Right panel – posterior view. Lower panels display head and neck region. (B)

7 MIBG-scintigraphy after 21 months of ceritinib therapy showing complete remission at all

8 metastatic sites. (C) Brain CT scans at diagnosis showing a CNS metastasis (43 mm x 46.7

9 mm) posterior to the right orbit. (D) CT scans after 21 months of ceritinib therapy showing

10 complete metastatic response. (E-H) Primary tumor immediately before and after

11 targeted therapy (E-H). (E) Magnetic resonance imaging (MRI) of primary abdominal

12 tumor (indicated by arrows and measurements inserted) immediately before start of

13 ceritinib targeted therapy (approximately 18.8 mL tumor volume), (F) MRI after 3

14 months therapy (approximately 13.8 mL tumor volume) and (G) MRI after 6 months

15 therapy immediately prior to radical surgery (approximately 10.6 mL tumor volume).

16 (H) Decrease of evaluable tumor volume after 3 and 6 months of targeted therapy.

17

18

19 Figure 2. Genomic analysis of the tumor sample. (A) Copy number profiling using

20 Affymetrix HD SNP-microrray shows a genomic profile with 11q-deletion and 17q-gain

21 indicative of high-risk neuroblastoma. (B) Two different likely pathogenic variants in the

22 FANCA gene were detected through exome sequencing and confirmed in constitutional DNA

23 of the patient. Read mapping is visualized in IGV. (C) Exome sequencing also detected a

24 novel ALK I1171T mutation in the tumor that was confirmed to be somatic through Sanger

28

Downloaded from molecularcasestudies.cshlp.org on October 9, 2021 - Published by Cold Spring Harbor Laboratory Press

1 sequencing. Electrophorogram of tumor in upper panel and from constitutional DNA in lower

2 panel.

3

4 Figure 3. Hematological counts and catecholamine metabolites in urine. (A) After

5 conventional high-risk neuroblastoma chemotherapy was started, the patient suffered severe

6 bone marrow toxicity with protracted anemia and thrombocytopenia requiring frequent

7 transfusions (each peak indicates post transfusion measurement). When hematological counts

8 stabilized, ceritinib treatment could be started (*). Hemoglobin and platelet counts remained

9 stable during TKI treatment, allowing for surgery (#) with radical removal of the

10 differentiated and calcified decreased primary tumor. (B) Catecholamine metabolites

11 (dopamine in black, homovanillic acid, HVA in red, and vanillylmandelic acid, VMA in

12 green) as neuroblastoma markers in the patient's urine (molar concentrations/creatinine

13 concentration). During ceritinib therapy (*), elevated urine catecholamine metabolites

14 returned to normal (below respective dashed line), indicating a biologically inactive residual

15 tumour. At surgery (#) normal levels had been reached.

16

17 Figure 4. Characterization of ALK-I1171T with cell culture systems. (A) Model of the

18 ALK kinase domain (PDB #3LCS) employing PyMol, which can be divided into the upper N-

19 terminal lobe and the lower C-terminal lobe by the hinge region and ceritinib binding/ATP

20 (red) binding pocket. Regulatory spine (R-spine) residues are shown in magenta. The spine is

21 anchored in the αF helix (D1311), and includes the DFG (F1271) and the HRD motif (H1247)

22 of the C-lobe. R-spine residues in the N-terminal lobe include the β4 strand (C1182) and the

23 residue mutated in this patient in the α-C-helix (I1171). Mutation of residue I1171 to

24 threonine in PyMol, results in a small shift, that potentially drives a dynamic allostery that

29

Downloaded from molecularcasestudies.cshlp.org on October 9, 2021 - Published by Cold Spring Harbor Laboratory Press

1 results in a gain-of-function ALK activity (upper insert:wild type ALK kinase domain; lower

2 insert: ALK-I1171T kinase domain). (B) ALK kinase activity and activation of downstream

3 signaling pathways were visualized by western blot with antibodies against pALK (Y1604)

4 and pERK1/2. Total ALK and pan-ERK were used as loading controls. Blots are

5 representative of three independent experiments. (C) Neurite outgrowth of PC12 cells as a

6 readout for ALK kinase activity was performed with wild type ALK and ALK variants in the

7 absence or presence of ALKAL1 ligand. Bars represent mean percentage ± STD of neurite-

8 carrying cells among GFP-positive cells from three independent experiments. (D)

9 Representative focus formation assays for NIH 3T3 cells transfected with wild type ALK,

10 ALK variants (ALK-I1171T and ALK-F1174L) or empty vector. (E) Inhibition profiling of

11 ALK TKIs on ALK-I1171T. PC12 cells expressing ALK-I1171T were treated with serial

12 dilution of ALK inhibitors as indicated. Phosphorylation of ALK was detected with pALK

13 (Y1604) antibody and total ALK was used as loading control. (F) IC50 values of different

14 ALK inhibitors were calculated with GraphPad Prism 6.0 by fitting data to a log (inhibitor

15 concentration) vs. normalized response (variable slope) equation and shown in the

16 accompanying table. Values represent average ± STD from three independent experiments.

17

18 Figure 5. Ceritinib phosphoproteomic profile in neuroblastoma cell lines. (A) Three

19 neuroblastoma cell lines (CLB-BAR, CLB-GE and SK-N-AS) were cultured in control

20 conditions or in the presence of ceritinib. Cells were harvested for tyrosine and

21 serine/threonine phosphoproteomic analysis after 60 mins. Pie chart indicates the number of

22 targeted sites. (B) Overview of all ALK tyrosine phosphorylation sites with log2 fold change

23 values (FC; treated/untreated) in CLB-BAR and CLB-GE. Note that no signal was measured

24 in SK-N-AS cells reflecting known absent expression of ALK. (C) Log2 FC values of all

25 analyzed phosphosites indicating the number of proteins found to contain phosphorylated

30

Downloaded from molecularcasestudies.cshlp.org on October 9, 2021 - Published by Cold Spring Harbor Laboratory Press

1 (red) or dephosphorylated (green) sites for the three cell lines analyzed. Dotted lines indicate

2 thresholds to determine differential phosphorylation. (D) Venn diagrams show the correlation

3 between phosphorylated (red) and dephosphorylated (green) proteins in the different cell

4 lines. Sixty proteins were found to be dephosphorylated upon ceritinib treatment in the ALK-

5 addicted CLB-BAR and CLB-GE lines but not in the SK-N-AS cell line. These proteins were

6 mapped to the InWeb_InBioMap protein protein interaction network (E). For visualization

7 purposes only direct interactions between the identified proteins are shown. Node sizes are

8 proportional to the number of connections while node colors indicate different log2 FC values

9 as indicated in the color scale legend on the top left. (F) Reactome gene set enrichment

10 analysis on all 60 proteins that were identified to be dephosphorylated. The 10 most

11 significantly enriched pathways are shown and ranked on FDR values.

12

13 Figure 6. Immunohistochemical analysis of tumor material. Tumor sample taken in

14 November 2014 was stroma-poor, comprising of small, round, primitive appearing cells

15 positive for the Ki-67 proliferation marker in up to 27%. Post ceritinib treatment resected

16 tumor displayed few Ki-67 positive cells, and increased expression of NB84 and NFP, that are

17 markers for differentiated neuroblastoma.

18

19

31

Downloaded from molecularcasestudies.cshlp.org on October 9, 2021 - Published by Cold Spring Harbor Laboratory Press

1 Supplementary Figure Legends

2

3 Figure S1. Patient tumor imaging. (A-B) CNS metastasis immediately before and after

4 targeted therapy. (A) MRI of CNS with metastasis indicated by arrows immediately before

5 start of targeted therapy. (B) MRI of CNS after 6 months of targeted therapy showing

6 decreased metastasis indicated by arrows. (C-D) Imaging showing metastatic bone

7 involvement at diagnosis and after completed targeted therapy. (C) 3D bone visualization of

8 CT scan at diagnosis showing metastatic bone involvement of the skull. (D) CT scan after

9 completed 21 months of targeted therapy with no remaining metastatic bone involvement.

10

11 Figure S2. Ceritinib blocks phosphorylation of mutant ALK variants expressed in PC12

12 cells. (A) PC12 cells were transfected with ALK-mutant variants or ALK-wild type as

13 indicated. 48 hours post-transfection cells were treated with serial diluted amounts of either

14 ceritinib or crizotinib. Whole cell lysates were blotted for pALK (Y1604) to measure ALK

15 activation/phosphorylation inhibition. (B) Western blot analysis of ALK expression, in PC12

16 whole cell lysate. (C) Inhibition profiles for ceritinib and crizotinib were determined with

17 GraphPad Prism 6.0 by fitting data from A, to a log (inhibitor concentration) vs. normalised

18 response (variable slope) equation. IC50 values are shown in Table 1. Values represent

19 average ± STD from at least two independent experiments.

20

21 Figure S3. Ceritinib inhibits ALK activity and proliferation of ALK addicted

22 neuroblastoma cell lines. (A) CLB-BAR (ALK-∆4-11), and CLB-GE (ALK-F1174F), both

23 ALK addicted cell lines and SK-N-AS (Ras-Q61K), a RAS addicted cell line, were treated

32

Downloaded from molecularcasestudies.cshlp.org on October 9, 2021 - Published by Cold Spring Harbor Laboratory Press

1 with increasing concentrations of ceritinib for 72 hours and cell viability assessed by resazurin

2 assay. Plotted values are means ± SE from growth curves from at least three independent

3 experiments preformed in triplicate. (B) CLB-BAR, CLB-GE and SK-N-AS were treated with

4 crizotinib (250 nM) and ceritinib (200 nM) for 1 hour. Non-treated cells was employed as a

5 negative control. Cell lysates were resolved by SDS/PAGE followed by immunoblotting as

6 indicated.

7

8 Figure S4. Ceritinib phosphorylation response of different tyrosine receptor kinases

9 (RTKs) in neuroblastoma cell lines. Log2 FC values of all analyzed phosphosites in

10 different RTKs and neuroblastoma cell lines as indicated. Median values are indicated by

11 horizontal lines.

12

13 Figure S5. Ceritinib phosphoproteomic profile in CLB-BAR. (A) Reactome gene set

14 enrichment analysis on all 182 proteins that were dephosphorylated upon ceritinib treatment.

15 The 10 most significantly enriched pathways are shown and ranked on FDR values. (B) All

16 dephosphorylated proteins were mapped to the InWeb_InBioMap protein protein interaction

17 network. For visualization purposes only direct interactions between the identified proteins

18 are shown. Node sizes are proportional to the number of connections while node colors

19 indicate different fold change values as indicated in the color scale legend on the top left.

20

21 Figure S6. Ceritinib phosphoproteomic profile in CLB-GE. (A) Reactome gene set

22 enrichment analysis on all 200 proteins that were dephosphorylated upon ceritinib treatment.

23 The 10 most significantly enriched pathways are shown and ranked on FDR values. (B) All

33

Downloaded from molecularcasestudies.cshlp.org on October 9, 2021 - Published by Cold Spring Harbor Laboratory Press

1 dephosphorylated proteins were mapped to the InWeb_InBioMap protein protein interaction

2 network. For visualization purposes only direct interactions between the identified proteins

3 are shown. Node sizes are proportional to the number of connections while node colors

4 indicate different fold change values as indicated in the color scale legend on the top left.

5

6

7

8

34

Downloaded from molecularcasestudies.cshlp.org on October 9, 2021 - Published by Cold Spring Harbor Laboratory Press

1 Table Legends

2

3 Table 1

4 Indicated neuroblastoma cell lines were treated with increasing doses of either ceritinib or

5 crizotinib for 72 hours and cell viability was assassed by resazurin assay. NA, non-amplified;

6 A - amplified; g-o-f ∆exon 4-11 – gain of function deletion of exons in the extracellular

7 domain of ALK; WT - wild type.

8 9

10 Table 2

11 IC50 values for inhibition of ALK Y1604 phosphorylation by ceritinib or crizotinib in PC12

12 cells. IC50 values were determined by quantification of Y1604 phosphorylation from the

13 immunoblots in Supplementary figure 1. Values represent mean ± SD from at least two

14 independent experiments.

15

35

Downloaded from molecularcasestudies.cshlp.org on October 9, 2021 - Published by Cold Spring Harbor Laboratory Press

1 Supplementary Table Legends

2

3 Table S1. FANCA and ALK mutation details. Variant specific information including

4 position, transcript-ID, coverage and presence in publically available databases.

5

6 Table S2. Log2 FC values of 64 dephosphorylated proteins in ALK expressing

7 neuroblastoma cell lines. Sixty-four proteins were dephosphorylated upon ceritinib treatment

8 in the CLB-BAR and CLB-GE but not the SK-N-AS cell lines with indication of log2 FC

9 values for the three cell lines.

10

36

Downloaded from molecularcasestudies.cshlp.org on October 9, 2021 - Published by Cold Spring Harbor Laboratory Press

1 References

2 3 4 (FDA) (26th of August, 2011). "U.F.a.D.A. FDA approves Xalkori with 5 companion diagnostic for a type of late-stage lung cancer." 6 http://www.fda.gov/NewsEvents/Newsroom/PressAnnouncements/ucm2698 7 56.htm. 8 (FDA) (26th of May, 2017). "FDA broadens ceritinib indication to previously 9 untreated ALK-positive metastatic NSCLC." 10 https://www.fda.gov/drugs/informationondrugs/approveddrugs/ucm560873.h 11 tm. 12 (FDA) (29th of April, 2014). "FDA approves Zykadia for late-stage lung 13 cancer." 14 https://www.accessdata.fda.gov/scripts/cder/daf/index.cfm?event=overview. 15 process&applno=205755. 16 Abbondanzo, S. L., H. J. Manz, R. S. Klappenbach and J. E. Gootenberg 17 (1986). "Hepatocellular carcinoma in an 11-year-old girl with Fanconi's 18 anemia. Report of a case and review of the literature." Am J Pediatr 19 Hematol Oncol 8(4): 334-337. 20 Benjamini, Y. and Y. Hochberg (1995). "Controlling the False Discovery Rate - 21 a Practical and Powerful Approach to Multiple Testing." Journal of the Royal 22 Statistical Society Series B-Methodological 57(1): 289-300. 23 Berrebi, D., M. N. Lebras, N. Belarbi, J. Couturier, S. Fattet, A. Faye, M. 24 Peuchmaur and P. de Lagausie (2006). "Bilateral adrenal neuroblastoma 25 and nephroblastoma occurring synchronously in a child with Fanconi's 26 anemia and VACTERL syndrome." J Pediatr Surg 41(1): e11-14. 27 Berry, T., W. Luther, N. Bhatnagar, Y. Jamin, E. Poon, T. Sanda, D. Pei, B. 28 Sharma, W. R. Vetharoy, A. Hallsworth, Z. Ahmad, K. Barker, L. Moreau, H. 29 Webber, W. Wang, Q. Liu, A. Perez-Atayde, S. Rodig, N. K. Cheung, F. 30 Raynaud, B. Hallberg, S. P. Robinson, N. S. Gray, A. D. Pearson, S. A. 31 Eccles, L. Chesler and R. E. George (2012). "The ALK(F1174L) mutation 32 potentiates the oncogenic activity of MYCN in neuroblastoma." Cancer Cell 33 22(1): 117-130. 34 Bissig, H., F. Staehelin, M. Tolnay, P. Avoledo, J. Richter, D. Betts, E. Bruder 35 and T. Kuhne (2002). "Co-occurrence of neuroblastoma and 36 nephroblastoma in an infant with Fanconi's anemia." Hum Pathol 33(10): 37 1047-1051. 38 Bresler, S. C., D. A. Weiser, P. J. Huwe, J. H. Park, K. Krytska, H. Ryles, M. 39 Laudenslager, E. F. Rappaport, A. C. Wood, P. W. McGrady, M. D. Hogarty, 40 W. B. London, R. Radhakrishnan, M. A. Lemmon and Y. P. Mosse (2014). 41 "ALK mutations confer differential oncogenic activation and sensitivity to 42 ALK inhibition therapy in neuroblastoma." Cancer Cell 26(5): 682-694. 43 Caren, H., F. Abel, P. Kogner and T. Martinsson (2008). "High incidence of 44 DNA mutations and gene amplifications of the ALK gene in advanced 45 sporadic neuroblastoma tumours." Biochem J 416(2): 153-159.

37

Downloaded from molecularcasestudies.cshlp.org on October 9, 2021 - Published by Cold Spring Harbor Laboratory Press

1 Cazes, A., L. Lopez-Delisle, K. Tsarovina, C. Pierre-Eugene, K. De Preter, M. 2 Peuchmaur, A. Nicolas, C. Provost, C. Louis-Brennetot, R. Daveau, C. 3 Kumps, I. Cascone, G. Schleiermacher, A. Prignon, F. Speleman, H. 4 Rohrer, O. Delattre and I. Janoueix-Lerosey (2014). "Activated Alk triggers 5 prolonged neurogenesis and Ret upregulation providing a therapeutic target 6 in ALK-mutated neuroblastoma." Oncotarget 5(9): 2688-2702. 7 Cazes, A., C. Louis-Brennetot, P. Mazot, F. Dingli, B. Lombard, V. Boeva, R. 8 Daveau, J. Cappo, V. Combaret, G. Schleiermacher, S. Jouannet, S. 9 Ferrand, G. Pierron, E. Barillot, D. Loew, M. Vigny, O. Delattre and I. 10 Janoueix-Lerosey (2013). "Characterization of rearrangements involving the 11 ALK gene reveals a novel truncated form associated with tumor 12 aggressiveness in neuroblastoma." Cancer Res 73(1): 195-204. 13 Chand, D., Y. Yamazaki, K. Ruuth, C. Schonherr, T. Martinsson, P. Kogner, E. 14 F. Attiyeh, J. Maris, O. Morozova, M. A. Marra, M. Ohira, A. Nakagawara, P. 15 E. Sandstrom, R. H. Palmer and B. Hallberg (2013). "Cell culture and 16 Drosophila model systems define three classes of anaplastic lymphoma 17 kinase mutations in neuroblastoma." Dis Model Mech. 18 Chen, Y., J. Takita, Y. L. Choi, M. Kato, M. Ohira, M. Sanada, L. Wang, M. 19 Soda, A. Kikuchi, T. Igarashi, A. Nakagawara, Y. Hayashi, H. Mano and S. 20 Ogawa (2008). "Oncogenic mutations of ALK kinase in neuroblastoma." 21 Nature 455(7215): 971-974. 22 Chia, P. L., P. Mitchell, A. Dobrovic and T. John (2014). "Prevalence and 23 natural history of ALK positive non-small-cell lung cancer and the clinical 24 impact of targeted therapy with ALK inhibitors." Clin Epidemiol 6: 423-432. 25 Christensen, J. G., H. Y. Zou, M. E. Arango, Q. Li, J. H. Lee, S. R. McDonnell, 26 S. Yamazaki, G. R. Alton, B. Mroczkowski and G. Los (2007). 27 "Cytoreductive antitumor activity of PF-2341066, a novel inhibitor of 28 anaplastic lymphoma kinase and c-Met, in experimental models of 29 anaplastic large-cell lymphoma." Mol Cancer Ther 6(12 Pt 1): 3314-3322. 30 Combaret, V., C. Turc-Carel, P. Thiesse, A. C. Rebillard, D. Frappaz, O. Haus, 31 T. Philip and M. C. Favrot (1995). "Sensitive detection of numerical and 32 structural aberrations of chromosome 1 in neuroblastoma by interphase 33 fluorescence in situ hybridization. Comparison with restriction fragment 34 length polymorphism and conventional cytogenetic analyses." Int J Cancer 35 61(2): 185-191. 36 Cowley, S., H. Paterson, P. Kemp and C. J. Marshall (1994). "Activation of 37 MAP kinase kinase is necessary and sufficient for PC12 differentiation and 38 for transformation of NIH 3T3 cells." Cell 77(6): 841-852. 39 De Brouwer, S., K. De Preter, C. Kumps, P. Zabrocki, M. Porcu, E. M. 40 Westerhout, A. Lakeman, J. Vandesompele, J. Hoebeeck, T. Van Maerken, 41 A. De Paepe, G. Laureys, J. H. Schulte, A. Schramm, C. Van Den Broecke, 42 J. Vermeulen, N. Van Roy, K. Beiske, M. Renard, R. Noguera, O. Delattre, I. 43 Janoueix-Lerosey, P. Kogner, T. Martinsson, A. Nakagawara, M. Ohira, H. 44 Caron, A. Eggert, J. Cools, R. Versteeg and F. Speleman (2010). "Meta- 45 analysis of neuroblastomas reveals a skewed ALK mutation spectrum in 46 tumors with MYCN amplification." Clin Cancer Res 16(17): 4353-4362.

38

Downloaded from molecularcasestudies.cshlp.org on October 9, 2021 - Published by Cold Spring Harbor Laboratory Press

1 Doebele, R. C., A. B. Pilling, D. L. Aisner, T. G. Kutateladze, A. T. Le, A. J. 2 Weickhardt, K. L. Kondo, D. J. Linderman, L. E. Heasley, W. A. Franklin, M. 3 Varella-Garcia and D. R. Camidge (2012). "Mechanisms of resistance to 4 crizotinib in patients with ALK gene rearranged non-small cell lung cancer." 5 Clin Cancer Res 18(5): 1472-1482. 6 Donella-Deana, A., O. Marin, L. Cesaro, R. H. Gunby, A. Ferrarese, A. M. 7 Coluccia, C. J. Tartari, L. Mologni, L. Scapozza, C. Gambacorti-Passerini 8 and L. A. Pinna (2005). "Unique substrate specificity of anaplastic 9 lymphoma kinase (ALK): development of phosphoacceptor peptides for the 10 assay of ALK activity." Biochemistry 44(23): 8533-8542. 11 Eleveld, T. F., D. A. Oldridge, V. Bernard, J. Koster, L. C. Daage, S. J. Diskin, 12 L. Schild, N. B. Bentahar, A. Bellini, M. Chicard, E. Lapouble, V. Combaret, 13 P. Legoix-Ne, J. Michon, T. J. Pugh, L. S. Hart, J. Rader, E. F. Attiyeh, J. S. 14 Wei, S. Zhang, A. Naranjo, J. M. Gastier-Foster, M. D. Hogarty, S. 15 Asgharzadeh, M. A. Smith, J. M. Guidry Auvil, T. B. Watkins, D. A. 16 Zwijnenburg, M. E. Ebus, P. van Sluis, A. Hakkert, E. van Wezel, C. E. van 17 der Schoot, E. M. Westerhout, J. H. Schulte, G. A. Tytgat, M. E. Dolman, I. 18 Janoueix-Lerosey, D. S. Gerhard, H. N. Caron, O. Delattre, J. Khan, R. 19 Versteeg, G. Schleiermacher, J. J. Molenaar and J. M. Maris (2015). 20 "Relapsed neuroblastomas show frequent RAS-MAPK pathway mutations." 21 Nat Genet 47(8): 864-871. 22 Fadeev, A., P. Mendoza-Garcia, U. Irion, J. Guan, K. Pfeifer, S. Wiessner, F. 23 Serluca, A. P. Singh, C. Nusslein-Volhard and R. H. Palmer (2018). 24 "ALKALs are in vivo ligands for ALK family receptor tyrosine kinases in the 25 neural crest and derived cells." Proc Natl Acad Sci U S A. 26 Fransson, S., M. Hansson, K. Ruuth, A. Djos, A. Berbegall, N. Javanmardi, J. 27 Abrahamsson, R. H. Palmer, R. Noguera, B. Hallberg, P. Kogner and T. 28 Martinsson (2015). "Intragenic anaplastic lymphoma kinase (ALK) 29 rearrangements: translocations as a novel mechanism of ALK activation in 30 neuroblastoma tumors." Genes Chromosomes Cancer 54(2): 99-109. 31 Fransson, S., M. Ostensson, A. Djos, N. Javanmardi, P. Kogner and T. 32 Martinsson (2016). "Estimation of copy number aberrations: Comparison of 33 exome sequencing data with SNP microarrays identifies homozygous 34 deletions of 19q13.2 and CIC in neuroblastoma." Int J Oncol 48(3): 1103- 35 1116. 36 George, R. E., T. Sanda, M. Hanna, S. Frohling, W. Luther, 2nd, J. Zhang, Y. 37 Ahn, W. Zhou, W. B. London, P. McGrady, L. Xue, S. Zozulya, V. E. Gregor, 38 T. R. Webb, N. S. Gray, D. G. Gilliland, L. Diller, H. Greulich, S. W. Morris, 39 M. Meyerson and A. T. Look (2008). "Activating mutations in ALK provide a 40 therapeutic target in neuroblastoma." Nature 455(7215): 975-978. 41 Guan, J., E. R. Tucker, H. Wan, D. Chand, L. S. Danielson, K. Ruuth, A. El 42 Wakil, B. Witek, Y. Jamin, G. Umapathy, S. P. Robinson, T. W. Johnson, T. 43 Smeal, T. Martinsson, L. Chesler, R. H. Palmer and B. Hallberg (2016). 44 "The ALK inhibitor PF-06463922 is effective as a single agent in 45 neuroblastoma driven by expression of ALK and MYCN." Dis Model Mech 46 9(9): 941-952.

39

Downloaded from molecularcasestudies.cshlp.org on October 9, 2021 - Published by Cold Spring Harbor Laboratory Press

1 Guan, J., G. Umapathy, Y. Yamazaki, G. Wolfstetter, P. Mendoza, K. Pfeifer, 2 A. Mohammed, F. Hugosson, H. Zhang, A. W. Hsu, R. Halenbeck, B. 3 Hallberg and R. H. Palmer (2015). "FAM150A and FAM150B are activating 4 ligands for anaplastic lymphoma kinase." Elife 4: e09811. 5 Guan, J., Y. Yamazaki, D. Chand, J. R. van Dijk, K. Ruuth, R. H. Palmer and 6 B. Hallberg (2017). "Novel Mechanisms of ALK Activation Revealed by 7 Analysis of the Y1278S Neuroblastoma Mutation." Cancers (Basel) 9(11). 8 Hallberg, B. and R. H. Palmer (2013). "Mechanistic insight into ALK receptor 9 tyrosine kinase in human cancer biology." Nat Rev Cancer 13(10): 685-700. 10 Hallberg, B. and R. H. Palmer (2016). "The role of the ALK receptor in cancer 11 biology." Ann Oncol 27 Suppl 3: iii4-iii15. 12 Heukamp, L. C., T. Thor, A. Schramm, K. De Preter, C. Kumps, B. De Wilde, 13 A. Odersky, M. Peifer, S. Lindner, A. Spruessel, F. Pattyn, P. Mestdagh, B. 14 Menten, S. Kuhfittig-Kulle, A. Kunkele, K. Konig, L. Meder, S. Chatterjee, R. 15 T. Ullrich, S. Schulte, J. Vandesompele, F. Speleman, R. Buttner, A. Eggert 16 and J. H. Schulte (2012). "Targeted expression of mutated ALK induces 17 neuroblastoma in transgenic mice." Sci Transl Med 4(141): 141ra191. 18 Infarinato, N. R., J. H. Park, K. Krytska, H. T. Ryles, R. Sano, K. M. Szigety, Y. 19 Li, H. Y. Zou, N. V. Lee, T. Smeal, M. A. Lemmon and Y. P. Mosse (2016). 20 "The ALK/ROS1 Inhibitor PF-06463922 Overcomes Primary Resistance to 21 Crizotinib in ALK-Driven Neuroblastoma." Cancer Discov 6(1): 96-107. 22 Iwahara, T., J. Fujimoto, D. Wen, R. Cupples, N. Bucay, T. Arakawa, S. Mori, 23 B. Ratzkin and T. Yamamoto (1997). "Molecular characterization of ALK, a 24 receptor tyrosine kinase expressed specifically in the nervous system." 25 Oncogene 14(4): 439-449. 26 Janoueix-Lerosey, I., D. Lequin, L. Brugieres, A. Ribeiro, L. de Pontual, V. 27 Combaret, V. Raynal, A. Puisieux, G. Schleiermacher, G. Pierron, D. 28 Valteau-Couanet, T. Frebourg, J. Michon, S. Lyonnet, J. Amiel and O. 29 Delattre (2008). "Somatic and germline activating mutations of the ALK 30 kinase receptor in neuroblastoma." Nature 455(7215): 967-970. 31 Johnson, T. W., P. F. Richardson, S. Bailey, A. Brooun, B. J. Burke, M. R. 32 Collins, J. J. Cui, J. G. Deal, Y. L. Deng, D. Dinh, L. D. Engstrom, M. He, J. 33 Hoffman, R. L. Hoffman, Q. Huang, R. S. Kania, J. C. Kath, H. Lam, J. L. 34 Lam, P. T. Le, L. Lingardo, W. Liu, M. McTigue, C. L. Palmer, N. W. Sach, 35 T. Smeal, G. L. Smith, A. E. Stewart, S. Timofeevski, H. Zhu, J. Zhu, H. Y. 36 Zou and M. P. Edwards (2014). "Discovery of (10R)-7-amino-12-fluoro- 37 2,10,16-trimethyl-15-oxo-10,15,16,17-tetrahydro-2H-8,4-(m 38 etheno)pyrazolo[4,3-h][2,5,11]-benzoxadiazacyclotetradecine-3-carbonitrile 39 (PF-06463922), a macrocyclic inhibitor of anaplastic lymphoma kinase 40 (ALK) and c-ros oncogene 1 (ROS1) with preclinical brain exposure and 41 broad-spectrum potency against ALK-resistant mutations." J Med Chem 42 57(11): 4720-4744. 43 Katayama, R., L. Friboulet, S. Koike, E. L. Lockerman, T. M. Khan, J. F. 44 Gainor, A. J. Iafrate, K. Takeuchi, M. Taiji, Y. Okuno, N. Fujita, J. A. 45 Engelman and A. T. Shaw (2014). "Two novel ALK mutations mediate

40

Downloaded from molecularcasestudies.cshlp.org on October 9, 2021 - Published by Cold Spring Harbor Laboratory Press

1 acquired resistance to the next-generation ALK inhibitor alectinib." Clin 2 Cancer Res 20(22): 5686-5696. 3 Katayama, R., T. M. Khan, C. Benes, E. Lifshits, H. Ebi, V. M. Rivera, W. C. 4 Shakespeare, A. J. Iafrate, J. A. Engelman and A. T. Shaw (2011). 5 "Therapeutic strategies to overcome crizotinib resistance in non-small cell 6 lung cancers harboring the fusion oncogene EML4-ALK." Proc Natl Acad 7 Sci U S A 108(18): 7535-7540. 8 Katayama, R., C. M. Lovly and A. T. Shaw (2015). "Therapeutic targeting of 9 anaplastic lymphoma kinase in lung cancer: a paradigm for precision cancer 10 medicine." Clin Cancer Res 21(10): 2227-2235. 11 Kim, D. W., R. Mehra, D. S. Tan, E. Felip, L. Q. Chow, D. R. Camidge, J. 12 Vansteenkiste, S. Sharma, T. De Pas, G. J. Riely, B. J. Solomon, J. Wolf, M. 13 Thomas, M. Schuler, G. Liu, A. Santoro, S. Sutradhar, S. Li, T. Szczudlo, A. 14 Yovine and A. T. Shaw (2016). "Activity and safety of ceritinib in patients 15 with ALK-rearranged non-small-cell lung cancer (ASCEND-1): updated 16 results from the multicentre, open-label, phase 1 trial." Lancet Oncol. 17 Kimble, D. C., F. P. Lach, S. Q. Gregg, F. X. Donovan, E. K. Flynn, A. Kamat, 18 A. Young, M. Vemulapalli, J. W. Thomas, J. C. Mullikin, A. D. Auerbach, A. 19 Smogorzewska and S. C. Chandrasekharappa (2017). "A comprehensive 20 approach to identification of pathogenic FANCA variants in Fanconi anemia 21 patients and their families." Hum Mutat. 22 Kohl, N. E., C. E. Gee and F. W. Alt (1984). "Activated expression of the N- 23 myc gene in human neuroblastomas and related tumors." Science 24 226(4680): 1335-1337. 25 Kopic, S., K. Eirich, B. Schuster, H. Hanenberg, R. Varon-Mateeva, O. 26 Rittinger, G. Schimpl, D. Schindler and N. Jones (2011). "Hepatoblastoma in 27 a 4-year-old girl with Fanconi anaemia." Acta Paediatr 100(5): 780-783. 28 Kornev, A. P. and S. S. Taylor (2015). "Dynamics-Driven Allostery in Protein 29 Kinases." Trends Biochem Sci 40(11): 628-647. 30 Krytska, K., H. T. Ryles, R. Sano, P. Raman, N. R. Infarinato, T. D. Hansel, M. 31 R. Makena, M. M. Song, C. P. Reynolds and Y. P. Mosse (2016). "Crizotinib 32 Synergizes with Chemotherapy in Preclinical Models of Neuroblastoma." 33 Clin Cancer Res 22(4): 948-960. 34 Kutler, D. I., A. D. Auerbach, J. Satagopan, P. F. Giampietro, S. D. Batish, A. 35 G. Huvos, A. Goberdhan, J. P. Shah and B. Singh (2003). "High incidence 36 of head and neck squamous cell carcinoma in patients with Fanconi 37 anemia." Arch Otolaryngol Head Neck Surg 129(1): 106-112. 38 Kutler, D. I., B. Singh, J. Satagopan, S. D. Batish, M. Berwick, P. F. 39 Giampietro, H. Hanenberg and A. D. Auerbach (2003). "A 20-year 40 perspective on the International Fanconi Anemia Registry (IFAR)." Blood 41 101(4): 1249-1256. 42 Kwak, E. L., Y. J. Bang, D. R. Camidge, A. T. Shaw, B. Solomon, R. G. Maki, 43 S. H. Ou, B. J. Dezube, P. A. Janne, D. B. Costa, M. Varella-Garcia, W. H. 44 Kim, T. J. Lynch, P. Fidias, H. Stubbs, J. A. Engelman, L. V. Sequist, W. 45 Tan, L. Gandhi, M. Mino-Kenudson, G. C. Wei, S. M. Shreeve, M. J. Ratain, 46 J. Settleman, J. G. Christensen, D. A. Haber, K. Wilner, R. Salgia, G. I.

41

Downloaded from molecularcasestudies.cshlp.org on October 9, 2021 - Published by Cold Spring Harbor Laboratory Press

1 Shapiro, J. W. Clark and A. J. Iafrate (2010). "Anaplastic lymphoma kinase 2 inhibition in non-small-cell lung cancer." N Engl J Med 363(18): 1693-1703. 3 Ladenstein, R., U. Potschger, A. D. J. Pearson, P. Brock, R. Luksch, V. Castel, 4 I. Yaniv, V. Papadakis, G. Laureys, J. Malis, W. Balwierz, E. Ruud, P. 5 Kogner, H. Schroeder, A. F. de Lacerda, M. Beck-Popovic, P. Bician, M. 6 Garami, T. Trahair, A. Canete, P. F. Ambros, K. Holmes, M. Gaze, G. 7 Schreier, A. Garaventa, G. Vassal, J. Michon, D. Valteau-Couanet and S. E. 8 N. Group (2017). "Busulfan and melphalan versus carboplatin, etoposide, 9 and melphalan as high-dose chemotherapy for high-risk neuroblastoma 10 (HR-NBL1/SIOPEN): an international, randomised, multi-arm, open-label, 11 phase 3 trial." Lancet Oncol 18(4): 500-514. 12 Li, T., R. Wernersson, R. B. Hansen, H. Horn, J. Mercer, G. Slodkowicz, C. T. 13 Workman, O. Rigina, K. Rapacki, H. H. Staerfeldt, S. Brunak, T. S. Jensen 14 and K. Lage (2017). "A scored human protein-protein interaction network to 15 catalyze genomic interpretation." Nat Methods 14(1): 61-64. 16 Malric, A., A. S. Defachelles, T. Leblanc, B. Lescoeur, B. Lacour, M. 17 Peuchmaur, C. A. Maurage, G. Pierron, D. Guillemot, C. D. d'Enghien, J. 18 Soulier, D. Stoppa-Lyonnet and F. Bourdeaut (2015). "Fanconi anemia and 19 solid malignancies in childhood: a national retrospective study." Pediatr 20 Blood Cancer 62(3): 463-470. 21 Mamrak, N. E., A. Shimamura and N. G. Howlett (2017). "Recent discoveries 22 in the molecular pathogenesis of the inherited bone marrow failure 23 syndrome Fanconi anemia." Blood Rev 31(3): 93-99. 24 Marsilje, T. H., W. Pei, B. Chen, W. Lu, T. Uno, Y. Jin, T. Jiang, S. Kim, N. Li, 25 M. Warmuth, Y. Sarkisova, F. Sun, A. Steffy, A. C. Pferdekamper, A. G. Li, 26 S. B. Joseph, Y. Kim, B. Liu, T. Tuntland, X. Cui, N. S. Gray, R. Steensma, 27 Y. Wan, J. Jiang, G. Chopiuk, J. Li, W. P. Gordon, W. Richmond, K. 28 Johnson, J. Chang, T. Groessl, Y. Q. He, A. Phimister, A. Aycinena, C. C. 29 Lee, B. Bursulaya, D. S. Karanewsky, H. M. Seidel, J. L. Harris and P. Y. 30 Michellys (2013). "Synthesis, structure-activity relationships, and in vivo 31 efficacy of the novel potent and selective anaplastic lymphoma kinase (ALK) 32 inhibitor 5-chloro-N2-(2-isopropoxy-5-methyl-4-(piperidin-4-yl)phenyl)-N4-(2- 33 (isopropylsulf onyl)phenyl)pyrimidine-2,4-diamine (LDK378) currently in 34 phase 1 and phase 2 clinical trials." J Med Chem 56(14): 5675-5690. 35 Martinsson, T., T. Eriksson, J. Abrahamsson, H. Caren, M. Hansson, P. 36 Kogner, S. Kamaraj, C. Schonherr, J. Weinmar, K. Ruuth, R. H. Palmer and 37 B. Hallberg (2011). "Appearance of the novel activating F1174S ALK 38 mutation in neuroblastoma correlates with aggressive tumor progression 39 and unresponsiveness to therapy." Cancer Res 71(1): 98-105. 40 Mo, E. S., Q. N. Cheng, A. V. Reshetnyak, J. Schlessinger and S. Nicoli 41 (2017). "Alk and Ltk ligands are essential for iridophore development in 42 zebrafish mediated by the receptor tyrosine kinase Ltk." Proceedings of the 43 National Academy of Sciences of the United States of America 114(45): 44 12027-12032. 45 Moog-Lutz, C., J. Degoutin, J. Y. Gouzi, Y. Frobert, N. Brunet-de Carvalho, J. 46 Bureau, C. Creminon and M. Vigny (2005). "Activation and inhibition of

42

Downloaded from molecularcasestudies.cshlp.org on October 9, 2021 - Published by Cold Spring Harbor Laboratory Press

1 anaplastic lymphoma kinase receptor tyrosine kinase by monoclonal 2 antibodies and absence of agonist activity of ." J Biol Chem 3 280(28): 26039-26048. 4 Moore, N. F., A. M. Azarova, N. Bhatnagar, K. N. Ross, L. E. Drake, S. 5 Frumm, Q. S. Liu, A. L. Christie, T. Sanda, L. Chesler, A. L. Kung, N. S. 6 Gray, K. Stegmaier and R. E. George (2014). "Molecular rationale for the 7 use of PI3K/AKT/mTOR pathway inhibitors in combination with crizotinib in 8 ALK-mutated neuroblastoma." Oncotarget 5(18): 8737-8749. 9 Morris, S. W., M. N. Kirstein, M. B. Valentine, K. G. Dittmer, D. N. Shapiro, D. 10 L. Saltman and A. T. Look (1994). "Fusion of a kinase gene, ALK, to a 11 nucleolar protein gene, NPM, in non-Hodgkin's lymphoma." Science 12 263(5151): 1281-1284. 13 Morris, S. W., C. Naeve, P. Mathew, P. L. James, M. N. Kirstein, X. Cui and D. 14 P. Witte (1997). "ALK, the chromosome 2 gene locus altered by the t(2;5) in 15 non-Hodgkin's lymphoma, encodes a novel neural receptor tyrosine kinase 16 that is highly related to leukocyte tyrosine kinase (LTK)." Oncogene 14(18): 17 2175-2188. 18 Mosse, Y. P., M. Laudenslager, L. Longo, K. A. Cole, A. Wood, E. F. Attiyeh, 19 M. J. Laquaglia, R. Sennett, J. E. Lynch, P. Perri, G. Laureys, F. Speleman, 20 C. Kim, C. Hou, H. Hakonarson, A. Torkamani, N. J. Schork, G. M. Brodeur, 21 G. P. Tonini, E. Rappaport, M. Devoto and J. M. Maris (2008). "Identification 22 of ALK as a major familial neuroblastoma predisposition gene." Nature 23 455(7215): 930-935. 24 Mosse, Y. P., M. S. Lim, S. D. Voss, K. Wilner, K. Ruffner, J. Laliberte, D. 25 Rolland, F. M. Balis, J. M. Maris, B. J. Weigel, A. M. Ingle, C. Ahern, P. C. 26 Adamson and S. M. Blaney (2013). "Safety and activity of crizotinib for 27 paediatric patients with refractory solid tumours or anaplastic large-cell 28 lymphoma: a Children's Oncology Group phase 1 consortium study." Lancet 29 Oncol 14(6): 472-480. 30 Mosse, Y. P., S. D. Voss, M. S. Lim, D. Rolland, C. G. Minard, E. Fox, P. 31 Adamson, K. Wilner, S. M. Blaney and B. J. Weigel (2017). "Targeting ALK 32 With Crizotinib in Pediatric Anaplastic Large Cell Lymphoma and 33 Inflammatory Myofibroblastic Tumor: A Children's Oncology Group Study." J 34 Clin Oncol 35(28): 3215-3221. 35 Motegi, A., J. Fujimoto, M. Kotani, H. Sakuraba and T. Yamamoto (2004). 36 "ALK receptor tyrosine kinase promotes cell growth and neurite outgrowth." 37 J Cell Sci 117(Pt 15): 3319-3329. 38 Neveling, K., D. Endt, H. Hoehn and D. Schindler (2009). "Genotype- 39 phenotype correlations in Fanconi anemia." Mutat Res 668(1-2): 73-91. 40 Okubo, J., J. Takita, Y. Chen, K. Oki, R. Nishimura, M. Kato, M. Sanada, M. 41 Hiwatari, Y. Hayashi, T. Igarashi and S. Ogawa (2012). "Aberrant activation 42 of ALK kinase by a novel truncated form ALK protein in neuroblastoma." 43 Oncogene 31(44): 4667-4676. 44 Ou, S. H., J. Greenbowe, Z. U. Khan, M. C. Azada, J. S. Ross, P. J. Stevens, 45 S. M. Ali, V. A. Miller and B. Gitlitz (2015). "I1171 missense mutation 46 (particularly I1171N) is a common resistance mutation in ALK-positive

43

Downloaded from molecularcasestudies.cshlp.org on October 9, 2021 - Published by Cold Spring Harbor Laboratory Press

1 NSCLC patients who have progressive disease while on alectinib and is 2 sensitive to ceritinib." Lung Cancer 88(2): 231-234. 3 Ou, S. H., S. J. Klempner, J. R. Greenbowe, M. Azada, A. B. Schrock, S. M. 4 Ali, J. S. Ross, P. J. Stephens and V. A. Miller (2014). "Identification of a 5 novel HIP1-ALK fusion variant in Non-Small-Cell Lung Cancer (NSCLC) and 6 discovery of ALK I1171 (I1171N/S) mutations in two ALK-rearranged 7 NSCLC patients with resistance to Alectinib." J Thorac Oncol 9(12): 1821- 8 1825. 9 Park, J. R., R. Bagatell, S. L. Cohn, A. D. Pearson, J. G. Villablanca, F. 10 Berthold, S. Burchill, A. Boubaker, K. McHugh, J. G. Nuchtern, W. B. 11 London, N. L. Seibel, O. W. Lindwasser, J. M. Maris, P. Brock, G. 12 Schleiermacher, R. Ladenstein, K. K. Matthay and D. Valteau-Couanet 13 (2017). "Revisions to the International Neuroblastoma Response Criteria: A 14 Consensus Statement From the National Cancer Institute Clinical Trials 15 Planning Meeting." J Clin Oncol 35(22): 2580-2587. 16 Reshetnyak, A. V., P. B. Murray, X. Shi, E. S. Mo, J. Mohanty, F. Tome, H. 17 Bai, M. Gunel, I. Lax and J. Schlessinger (2015). "Augmentor alpha and 18 beta (FAM150) are ligands of the receptor tyrosine kinases ALK and LTK: 19 Hierarchy and specificity of ligand-receptor interactions." Proc Natl Acad Sci 20 U S A 112(52): 15862-15867. 21 Rikova, K., A. Guo, Q. Zeng, A. Possemato, J. Yu, H. Haack, J. Nardone, K. 22 Lee, C. Reeves, Y. Li, Y. Hu, Z. Tan, M. Stokes, L. Sullivan, J. Mitchell, R. 23 Wetzel, J. Macneill, J. M. Ren, J. Yuan, C. E. Bakalarski, J. Villen, J. M. 24 Kornhauser, B. Smith, D. Li, X. Zhou, S. P. Gygi, T. L. Gu, R. D. 25 Polakiewicz, J. Rush and M. J. Comb (2007). "Global survey of 26 phosphotyrosine signaling identifies oncogenic kinases in lung cancer." Cell 27 131(6): 1190-1203. 28 Robinson, J. T., H. Thorvaldsdottir, W. Winckler, M. Guttman, E. S. Lander, G. 29 Getz and J. P. Mesirov (2011). "Integrative genomics viewer." Nat 30 Biotechnol 29(1): 24-26. 31 Rush, J., A. Moritz, K. A. Lee, A. Guo, V. L. Goss, E. J. Spek, H. Zhang, X. M. 32 Zha, R. D. Polakiewicz and M. J. Comb (2005). "Immunoaffinity profiling of 33 tyrosine phosphorylation in cancer cells." Nat Biotechnol 23(1): 94-101. 34 Sakamoto, H., T. Tsukaguchi, S. Hiroshima, T. Kodama, T. Kobayashi, T. A. 35 Fukami, N. Oikawa, T. Tsukuda, N. Ishii and Y. Aoki (2011). "CH5424802, a 36 Selective ALK Inhibitor Capable of Blocking the Resistant Gatekeeper 37 Mutant." Cancer Cell 19(5): 679-690. 38 Sattu, K., F. Hochgrafe, J. Wu, G. Umapathy, C. Schonherr, K. Ruuth, D. 39 Chand, B. Witek, J. Fuchs, P. K. Li, F. Hugosson, R. J. Daly, R. H. Palmer 40 and B. Hallberg (2013). "Phosphoproteomic analysis of ALK downstream 41 signaling pathways identifies STAT3 as a functional target of activated ALK 42 in neuroblastoma cells." Febs J. 43 Schleiermacher, G., I. Janoueix-Lerosey, V. Combaret, J. Derre, J. Couturier, 44 A. Aurias and O. Delattre (2003). "Combined 24-color karyotyping and 45 comparative genomic hybridization analysis indicates predominant

44

Downloaded from molecularcasestudies.cshlp.org on October 9, 2021 - Published by Cold Spring Harbor Laboratory Press

1 rearrangements of early replicating chromosome regions in neuroblastoma." 2 Cancer Genet Cytogenet 141(1): 32-42. 3 Schleiermacher, G., N. Javanmardi, V. Bernard, Q. Leroy, J. Cappo, T. Rio 4 Frio, G. Pierron, E. Lapouble, V. Combaret, F. Speleman, B. de Wilde, A. 5 Djos, I. Ora, F. Hedborg, C. Trager, B. M. Holmqvist, J. Abrahamsson, M. 6 Peuchmaur, J. Michon, I. Janoueix-Lerosey, P. Kogner, O. Delattre and T. 7 Martinsson (2014). "Emergence of new ALK mutations at relapse of 8 neuroblastoma." J Clin Oncol 32(25): 2727-2734. 9 Schonherr, C., K. Ruuth, S. Kamaraj, C. L. Wang, H. L. Yang, V. Combaret, A. 10 Djos, T. Martinsson, J. G. Christensen, R. H. Palmer and B. Hallberg (2012). 11 "Anaplastic Lymphoma Kinase (ALK) regulates initiation of transcription of 12 MYCN in neuroblastoma cells." Oncogene 31(50): 5193-5200. 13 Schonherr, C., K. Ruuth, Y. Yamazaki, T. Eriksson, J. Christensen, R. H. 14 Palmer and B. Hallberg (2011). "Activating ALK mutations found in 15 neuroblastoma are inhibited by Crizotinib and NVP-TAE684." Biochem J 16 440(3): 405-413. 17 Shaw, A. T., D. W. Kim, R. Mehra, D. S. Tan, E. Felip, L. Q. Chow, D. R. 18 Camidge, J. Vansteenkiste, S. Sharma, T. De Pas, G. J. Riely, B. J. 19 Solomon, J. Wolf, M. Thomas, M. Schuler, G. Liu, A. Santoro, Y. Y. Lau, M. 20 Goldwasser, A. L. Boral and J. A. Engelman (2014). "Ceritinib in ALK- 21 rearranged non-small-cell lung cancer." N Engl J Med 370(13): 1189-1197. 22 Shaw, A. T., D. W. Kim, K. Nakagawa, T. Seto, L. Crino, M. J. Ahn, T. De Pas, 23 B. Besse, B. J. Solomon, F. Blackhall, Y. L. Wu, M. Thomas, K. J. O'Byrne, 24 D. Moro-Sibilot, D. R. Camidge, T. Mok, V. Hirsh, G. J. Riely, S. Iyer, V. 25 Tassell, A. Polli, K. D. Wilner and P. A. Janne (2013). "Crizotinib versus 26 chemotherapy in advanced ALK-positive lung cancer." N Engl J Med 27 368(25): 2385-2394. 28 Siaw, J. T., H. Wan, K. Pfeifer, V. M. Rivera, J. Guan, R. H. Palmer and B. 29 Hallberg (2016). "Brigatinib, an anaplastic lymphoma kinase inhibitor, 30 abrogates activity and growth in ALK-positive neuroblastoma cells, 31 Drosophila and mice." Oncotarget 7(20): 29011-29022. 32 Subramanian, A., P. Tamayo, V. K. Mootha, S. Mukherjee, B. L. Ebert, M. A. 33 Gillette, A. Paulovich, S. L. Pomeroy, T. R. Golub, E. S. Lander and J. P. 34 Mesirov (2005). "Gene set enrichment analysis: a knowledge-based 35 approach for interpreting genome-wide expression profiles." Proc Natl Acad 36 Sci U S A 102(43): 15545-15550. 37 Tartari, C. J., R. H. Gunby, A. M. Coluccia, R. Sottocornola, B. Cimbro, L. 38 Scapozza, A. Donella-Deana, L. A. Pinna and C. Gambacorti-Passerini 39 (2008). "Characterization of some molecular mechanisms governing 40 autoactivation of the catalytic domain of the anaplastic lymphoma kinase." J 41 Biol Chem 283(7): 3743-3750. 42 Toyokawa, G., F. Hirai, E. Inamasu, T. Yoshida, K. Nosaki, T. Takenaka, M. 43 Yamaguchi, T. Seto, M. Takenoyama and Y. Ichinose (2014). "Secondary 44 mutations at I1171 in the ALK gene confer resistance to both Crizotinib and 45 Alectinib." J Thorac Oncol 9(12): e86-87.

45

Downloaded from molecularcasestudies.cshlp.org on October 9, 2021 - Published by Cold Spring Harbor Laboratory Press

1 Ueda, T., Y. Nakata, N. Yamasaki, H. Oda, K. Sentani, A. Kanai, N. Onishi, K. 2 Ikeda, Y. Sera, Z. I. Honda, K. Tanaka, M. Sata, S. Ogawa, W. Yasui, H. 3 Saya, J. Takita and H. Honda (2016). "ALK(R1275Q) perturbs extracellular 4 matrix, enhances cell invasion and leads to the development of 5 neuroblastoma in cooperation with MYCN." Oncogene 35(34): 4447-4458. 6 Umapathy, G., A. El Wakil, B. Witek, L. Chesler, L. Danielson, X. Deng, N. S. 7 Gray, M. Johansson, S. Kvarnbrink, K. Ruuth, C. Schonherr, R. H. Palmer 8 and B. Hallberg (2014). "The kinase ALK stimulates the kinase ERK5 to 9 promote the expression of the oncogene MYCN in neuroblastoma." Sci 10 Signal 7(349): ra102. 11 Umapathy, G., J. Guan, D. E. Gustafsson, N. Javanmardi, D. Cervantes- 12 Madrid, A. Djos, T. Martinsson, R. H. Palmer and B. Hallberg (2017). "MEK 13 inhibitor does not prevent the growth of anaplastic lymphoma 14 kinase (ALK)-addicted neuroblastomas." Sci Signal 10(507). 15 Walsh, M. F., V. Y. Chang, W. K. Kohlmann, H. S. Scott, C. Cunniff, F. 16 Bourdeaut, J. J. Molenaar, C. C. Porter, J. T. Sandlund, S. E. Plon, L. L. 17 Wang and S. A. Savage (2017). "Recommendations for Childhood Cancer 18 Screening and Surveillance in DNA Repair Disorders." Clin Cancer Res 19 23(11): e23-e31. 20 Wang, P., F. Wu, J. Zhang, T. McMullen, L. C. Young, R. J. Ingham, L. Li and 21 R. Lai (2010). "Serine phosphorylation of NPM-ALK, which is dependent on 22 the auto-activation of the kinase activation loop, contributes to its oncogenic 23 potential." Carcinogenesis. 24 Wood, A. C., K. Krytska, H. T. Ryles, N. R. Infarinato, R. Sano, T. D. Hansel, 25 L. S. Hart, F. J. King, T. R. Smith, E. Ainscow, K. B. Grandinetti, T. 26 Tuntland, S. Kim, G. Caponigro, Y. Q. He, S. Krupa, N. Li, J. L. Harris and 27 Y. P. Mosse (2016). "Dual ALK and CDK4/6 Inhibition Demonstrates 28 Synergy against Neuroblastoma." Clin Cancer Res. 29 Zhang, H., L. I. Pao, A. Zhou, A. D. Brace, R. Halenbeck, A. W. Hsu, T. L. 30 Bray, K. Hestir, E. Bosch, E. Lee, G. Wang, H. Liu, B. R. Wong, W. M. 31 Kavanaugh and L. T. Williams (2014). "Deorphanization of the human 32 leukocyte tyrosine kinase (LTK) receptor by a signaling screen of the 33 extracellular proteome." Proc Natl Acad Sci U S A 111(44): 15741-15745. 34 Zhu, S., J. S. Lee, F. Guo, J. Shin, A. R. Perez-Atayde, J. L. Kutok, S. J. 35 Rodig, D. S. Neuberg, D. Helman, H. Feng, R. A. Stewart, W. Wang, R. E. 36 George, J. P. Kanki and A. T. Look (2012). "Activated ALK collaborates with 37 MYCN in neuroblastoma pathogenesis." Cancer Cell 21(3): 362-373. 38

46

Downloaded from molecularcasestudies.cshlp.org on October 9, 2021 - Published by Cold Spring Harbor Laboratory Press

TABLE 1

Cell line ALK status MYCN IC50 nM (ceritinib) IC50 nM (crizotinib) status CLB-GE F1174V, A A 131,3 ± 24 233,7 ± 18 CLB-BAR g-o-f ∆exon 4-12 A 36,1 ± 6.2 54,9 ± 20 KELLY F1174L, A A 328,9 ± 36 333,3 ± 78 CLB-GAR R1275Q NA 102,3 ± 16 119,3 ± 45 IMR32 Wt A ˃ 500 348,7 ± 45 SK-N-BE Wt, NA A ˃ 1500 ˃ 1000 SK-N-DZ Wt A ˃ 800 ˃ 4000 SKNAS Wt, NA NA ˃ 3000 353,0 ± 27 CLB-PE Wt A ˃ 15000 ˃ 4000

Downloaded from molecularcasestudies.cshlp.org on October 9, 2021 - Published by Cold Spring Harbor Laboratory Press

TABLE 2

ALK mutation Ceritinib Normalised to Crizotinib Normalised to Fold (IC50 nM) IC50 of WT (IC50 nM) IC50 of WT change WT 5.3 ± 0.2 1.0 17.8 ± 0.2 1 3.4 G1128A 19.8 ± 3.5 3.7 22.2 ± 3.4 1.2 1.1 I1171N 46.6 ± 16 8.8 139.5 ± 13.8 7.8 3.0 F1174L 16.9 ± 8 3.2 29.9 ± 4.8 1.7 1.8 R1192P 26.1 ± 1.2 4.9 28.1 ± 7.5 1.6 1.1 F1245C 24.6 ± 6.4 4.6 37.5 ± 7.7 2.1 1.5 G1269A 28.9 ± 3.6 5.5 113.5 ± 8.5 6.4 3.9 R1275Q 10.8 ± 3.5 2.0 30.3 ± 0.9 1.7 2.8 Y1278S 20.5 ± 0.1 3.9 69.5 ± 0.03 3.9 3.4 Downloaded from molecularcasestudies.cshlp.org on October 9, 2021 - Published by Cold Spring Harbor Laboratory Press

Figure1 A B

C D

E F G H FigureDownloaded 2 from molecularcasestudies.cshlp.org on October 9, 2021 - Published by Cold Spring Harbor Laboratory Press Figure 3Downloaded from molecularcasestudies.cshlp.org on October 9, 2021 - Published by Cold Spring Harbor Laboratory Press

A

B Downloaded from molecularcasestudies.cshlp.org on October 9, 2021 - Published by Cold Spring Harbor Laboratory Press Figure 4

A B

pcDNA3 + + - - - - ALK-WT - - + + - - ALK-I1171T - - - - + - ALK-F1174L - - - - - + ALKAL1 - + - + - -

pALK

ALK

pERK1/2

panERK

C D

pcDNA3 ALK-WT

pcDNA3 + + - - - - ALK-WT - - + + - - ALK-I1171T - - - - + - ALK-F1174L - - - - - + ALK-I1171T ALK-F1174L ALKAL1 - + - + - -

E F

Inhibitor (nM): 0 5 10 25 50 100 250 500 1000

pALK crizotinib ALK

pALK ceritinib ALK

pALK lorlatinib ALK ALK TKI IC50 nM crizotinib 193 ± 57 pALK brigatinib ceritinib 17.4 ± 3.1 ALK lorlatinib 6.8 ± 1.3

brigatinib 7.5 ± 0.4 Downloaded from molecularcasestudies.cshlp.org on October 9, 2021 - Published by Cold Spring Harbor Laboratory Press Figure 5

A B Log2 FC CLB-BAR Ceritinib (LDK-378) 3,345 proteins Site CLB-BAR CLB-GE CLB-GE Y1078 -1.98 -2.56 S SK-N-AS Y1092 -1.52 -1.87 5,013 Y1096 -3.49 -3.56 P Y1131 -1.74 -1.82 2,223 2,583 Y1278 -3.63 -2.79 Y1282 -3.42 -2.94 T Y Phosphoproteomics Y1283 -2.98 -1.93 Y1401 -0.41 0h 1h Y1507 -0.82 -1.47 Y1584 -1.49 -2.15 Y1586 -1.70 -2.15 Y1604 -1.36 -2.15 C E Log2 FC >4.5 SHC3 3.5-4.5 NEK9 BICD2

2.5-3.5 SHC2 EIF4B 5 1-5-2.5 FNBP1L TNKS1BP1 <1.5 ALK RPS6 AP2B1

GAB1

20 proteins 20 52 proteins 52 120 proteins 120 AKT1 FRS2 MAPK1 SOS1 PDCD11

AKT3 0 PTPN11 MAPK3 CCT2 GAB2 SLC35E1 BCR

Log2 FC treated/untreated FC Log2 MAP1A HSF1 IRS2

-5 SLC35B3 ERF

35 proteins 35 200 proteins 200 182 proteins 182 RBM4 PDCD4

F CLB-GE CLB-BAR SK-N-AS Reactome GSEA (60 dephosphorylated proteins)

INSULIN_RECEPTOR_SIGNALLING_CASCADE SIGNALING_BY_INSULIN_RECEPTOR D SK-N-AS SK-N-AS NGF_SIGNALLING_VIA_TRKA_FROM_THE_PLASMA_MEMBRANE SIGNALING_BY_FGFR_IN_DISEASE SHC_MEDIATED_SIGNALLING 17 26 SIGNALING_SIGNAL_ATTENUATION 1 1 3 2 SHC_RELATED_EVENTS 1 4 SIGNALLING_TO_ERKS 44 6 112 115 60 134 SIGNALLING_BY_NGF PI3K_CASCADE

CLB-BAR CLB-GE CLB-BAR CLB-GE 0 2 4 6 8 -log10 (FDR) Figure 6 Downloaded from molecularcasestudies.cshlp.org

Ki-67 NFP NB84 Nov 2014 Nov onOctober9,2021-Publishedby Dec 2015 Dec Cold SpringHarborLaboratoryPress

Downloaded from molecularcasestudies.cshlp.org on October 9, 2021 - Published by Cold Spring Harbor Laboratory Press

Clinical response of the novel activating ALK-I1171T mutation in neuroblastoma to the ALK inhibitor ceritinib.

Jikui Guan, Susanne Fransson, Joachim Tetteh T Siaw, et al.

Cold Spring Harb Mol Case Stud published online June 15, 2018 Access the most recent version at doi:10.1101/mcs.a002550

Supplementary http://molecularcasestudies.cshlp.org/content/suppl/2018/06/15/mcs.a00255 Material 0.DC1

Published online June 15, 2018 in advance of the full issue.

Accepted Peer-reviewed and accepted for publication but not copyedited or typeset; Manuscript accepted manuscript is likely to differ from the final, published version. Published onlineJune 15, 2018in advance of the full issue.

Creative This article is distributed under the terms of the Commons http://creativecommons.org/licenses/by-nc/4.0/, which permits reuse and License redistribution, except for commercial purposes, provided that the original author and source are credited. Email Alerting Receive free email alerts when new articles cite this article - sign up in the box at Service the top right corner of the article or click here.

Published by Cold Spring Harbor Laboratory Press