27
molecular tumor boards Successful Clinical Sequencing by Molecular Tumor Board in an Elderly Patient With Refractory S ´ ezary Syndrome Yasuki Hijikata, MD, PhD 1 ; Kazuaki Yokoyama, MD, PhD 2 ; Nozomi Yokoyama, MT 3 ; Yasuo Matsubara, MD, PhD 1 ; Eigo Shimizu 4 ; Makoto Nakashima, PhD 5 ; Makoto Yamagishi, PhD 5 ; Yasunori Ota, MD, PhD 7 ; Lay Ahyoung Lim, MD, PhD 1 ; Rui Yamaguchi, PhD 4 ; Mika Ito 6 ; Yukihisa Tanaka, MT 7 ; Tamami Denda, MS 7 ; Kenzaburo Tani, MD 1 ; Hiroshi Yotsuyanagi, MD 1 ; Seiya Imoto, PhD 8 ; Satoru Miyano, PhD 4 ; Kaoru Uchimaru, MD, PhD 5 ; and Arinobu Tojo, MD, PhD 2,8 INTRODUCTION ezary syndrome (SS), a rare, aggressive, leukemic variant of cutaneous T-cell lymphoma (CTCL), com- prises the malignant clonal proliferation of T lym- phocytes, and is prone to skin involvement. 1 Most patients with SS are elderly, with erythroderma present on . 80% of the body. SS is rarely curable and has a poor prognosis; the median survival of patients with stage IV SS is approximately 2 years. 2 Recent SS genomic proling with next-generation sequencing (NGS) demonstrated impaired normal signaling, which suggests the possibility of new treatment targets. 3 NGS-based genomic testing for cancer is becoming more widespread as a clinical tool for accurate di- agnosis and proper treatment in clinical oncology. 4 However, the use of various NGS techniques to guide cancer therapy has created challenges in an- alyzing large volumes of genomic data and reporting results to patients and caregivers. 5 To resolve this, we organized a clinical sequencing team called the mo- lecular tumor board (MTB) at the Institute of Medical Science, The University of Tokyo. Clinical sequencing is associated with several potential challenges in analysis, interpretation, and drug development for rare cancers such as SS. Here, we report the use of clinical sequencing to achieve a complete response in an elderly patient with SS refractory to standard treatment. CASE REPORT A 75-year-old male was diagnosed with SS (T4N3M0B1b; stage IVA2) in 2015. He had received various treatments in multiple hospitals, including pred- nisone, psoralen and ultraviolet A, extracorporeal photopheresis, bexarotene, romidepsin, total skin electron beam therapy, brentuximab vedotin, in- terferon alfa-2b, methotrexate, and pembrolizumab, but skin lesions did not respond adequately. Severe pneumonia also occurred during methotrexate therapy and was resolved by intensive care. The patient was admitted to our hospital in November 2016. Gener- alized erythroderma, clinical stage IIIA, was observed. He received mogalizumab, low-dose VP-16, and local radiation therapy. Each resulted in temporal clinical responses, but with limited effects. Our study protocol (No. 26-112-270402/28-17-0708; Appendix) was approved by our institutional review board and was in accordance with the Declaration of Helsinki. After obtaining written informed consent, whole-exome sequencing (WES) and RNA sequencing were performed on skin tumor biopsy samples on February 1, 2017, for comparisons with normal tissue, followed by analysis by our hospital curators (Appendix Figs A1 and A2). Specically, from WES, 77 coding variants were identied (Appendix Table A1). Among these, the MTB inferred only one driver mutation in TP53 (c.560-1G.A, splicing mutation, Appendix Table A1) from which no actionable molecular targets were identi ed. However, from RNA sequencing, actionable molecular targets were identi ed. We regarded the following gene upregulations as directly targetable: CCR4 (mogalizumab), LCK (dasatinib), CD274 as PD-L1 (PD-L1 inhibitors, pembrolizumab), CTLA4 (ipilimumab), IL2RA as CD25 (denileukin dif- titox), TNFRSF8 as CD30 (brentuximab vedotin), and nuclear factor-κB (NF-κB) signaling pathway (Table 1). Of note, we also inferred lenalidomide as a potential therapeutic option on the basis of the following two reasons: The tumor was enriched in pathways that were all indirectly targetable by lenalidomide (eg, Kyoto Encyclopedia of Genes and Genomes [KEGG]-NF-κB signaling [Appendix Fig A7], KEGG-cytokine-cytokine receptor interaction [Appendix Fig A9], and KEGG-natural killer [NK]-cellmediated cytotoxicity 6 [Appendix Fig A8], and KEGG-tumor necrosis factor [TNF] signal- ing [Appendix Fig A10] on the basis of the KEGG DRUG Database, 7 Therapeutic Target Database 8 ), and documented efcacy of lenalidomide was reported in patients with CTCL with overall response rates of 28% in a phase II trial of monotherapy for refractory SS 9 and a phase III trial of maintenance therapy. 10 Mogalizumab, pembrolizumab, and brentuximab vedotin had already been used during the patients protracted clinical course. Denileukin diftitox was excluded because the latest immunohistochemical ASSOCIATED CONTENT Appendix Author affiliations and support information (if applicable) appear at the end of this article. Accepted on December 14, 2019 and published at ascopubs.org/journal/ po on May 15, 2020: DOI https://doi.org/10. 1200/PO.19.00254 534 Downloaded from ascopubs.org by Tokyo Daigaku Ikagaku Kenkyujo on June 1, 2020 from 157.082.096.062 Copyright © 2020 American Society of Clinical Oncology. All rights reserved.

Successful Clinical Sequencing by Molecular ......more widespread as a clinical tool for accurate di-agnosis and proper treatment in clinical oncology.4 However, the use of various

  • Upload
    others

  • View
    1

  • Download
    0

Embed Size (px)

Citation preview

  • molecular

    tumor

    boards

    Successful Clinical Sequencing by MolecularTumor Board in an Elderly Patient With RefractorySézary SyndromeYasuki Hijikata, MD, PhD1; Kazuaki Yokoyama, MD, PhD2; Nozomi Yokoyama, MT3; Yasuo Matsubara, MD, PhD1; Eigo Shimizu4;Makoto Nakashima, PhD5; Makoto Yamagishi, PhD5; Yasunori Ota, MD, PhD7; Lay Ahyoung Lim, MD, PhD1; Rui Yamaguchi, PhD4;Mika Ito6; Yukihisa Tanaka, MT7; Tamami Denda, MS7; Kenzaburo Tani, MD1; Hiroshi Yotsuyanagi, MD1; Seiya Imoto, PhD8;Satoru Miyano, PhD4; Kaoru Uchimaru, MD, PhD5; and Arinobu Tojo, MD, PhD2,8

    INTRODUCTION

    Sézary syndrome (SS), a rare, aggressive, leukemicvariant of cutaneous T-cell lymphoma (CTCL), com-prises the malignant clonal proliferation of T lym-phocytes, and is prone to skin involvement.1 Mostpatients with SS are elderly, with erythroderma presenton . 80% of the body. SS is rarely curable and hasa poor prognosis; the median survival of patients withstage IV SS is approximately 2 years.2 Recent SSgenomic profiling with next-generation sequencing(NGS) demonstrated impaired normal signaling, whichsuggests the possibility of new treatment targets.3

    NGS-based genomic testing for cancer is becomingmore widespread as a clinical tool for accurate di-agnosis and proper treatment in clinical oncology.4

    However, the use of various NGS techniques toguide cancer therapy has created challenges in an-alyzing large volumes of genomic data and reportingresults to patients and caregivers.5 To resolve this, weorganized a clinical sequencing team called the mo-lecular tumor board (MTB) at the Institute of MedicalScience, The University of Tokyo. Clinical sequencingis associated with several potential challenges inanalysis, interpretation, and drug development for rarecancers such as SS. Here, we report the use of clinicalsequencing to achieve a complete response in anelderly patient with SS refractory to standard treatment.

    CASE REPORT

    A 75-year-old male was diagnosed with SS (T4N3M0B1b;stage IVA2) in 2015. He had received varioustreatments in multiple hospitals, including pred-nisone, psoralen and ultraviolet A, extracorporealphotopheresis, bexarotene, romidepsin, total skinelectron beam therapy, brentuximab vedotin, in-terferon alfa-2b, methotrexate, and pembrolizumab,but skin lesions did not respond adequately. Severepneumonia also occurred duringmethotrexate therapyand was resolved by intensive care. The patient wasadmitted to our hospital in November 2016. Gener-alized erythroderma, clinical stage IIIA, was observed.He received mogalizumab, low-dose VP-16, and local

    radiation therapy. Each resulted in temporal clinicalresponses, but with limited effects.

    Our study protocol (No. 26-112-270402/28-17-0708;Appendix) was approved by our institutional reviewboard and was in accordance with the Declarationof Helsinki. After obtaining written informed consent,whole-exome sequencing (WES) and RNA sequencingwere performed on skin tumor biopsy samples onFebruary 1, 2017, for comparisons with normal tissue,followed by analysis by our hospital curators (AppendixFigs A1 and A2). Specifically, from WES, 77 codingvariants were identified (Appendix Table A1). Amongthese, the MTB inferred only one driver mutation inTP53 (c.560-1G.A, splicing mutation, AppendixTable A1) from which no actionable molecular targetswere identified. However, from RNA sequencing,actionable molecular targets were identified. Weregarded the following gene upregulations as directlytargetable: CCR4 (mogalizumab), LCK (dasatinib),CD274 as PD-L1 (PD-L1 inhibitors, pembrolizumab),CTLA4 (ipilimumab), IL2RA as CD25 (denileukin dif-titox), TNFRSF8 as CD30 (brentuximab vedotin), andnuclear factor-κB (NF-κB) signaling pathway (Table 1).Of note, we also inferred lenalidomide as a potentialtherapeutic option on the basis of the following tworeasons: The tumor was enriched in pathways that wereall indirectly targetable by lenalidomide (eg, KyotoEncyclopedia of Genes and Genomes [KEGG]-NF-κBsignaling [Appendix Fig A7], KEGG-cytokine-cytokinereceptor interaction [Appendix Fig A9], andKEGG-naturalkiller [NK]-cell–mediated cytotoxicity6 [Appendix Fig A8],and KEGG-tumor necrosis factor [TNF] signal-ing [Appendix Fig A10] on the basis of the KEGGDRUGDatabase,7 Therapeutic Target Database8), anddocumented efficacy of lenalidomide was reported inpatients with CTCL with overall response rates of28% in a phase II trial of monotherapy for refractorySS9 and a phase III trial of maintenance therapy.10

    Mogalizumab, pembrolizumab, and brentuximabvedotin had already been used during the patient’sprotracted clinical course. Denileukin diftitox wasexcluded because the latest immunohistochemical

    ASSOCIATEDCONTENT

    Appendix

    Author affiliationsand supportinformation (ifapplicable) appear atthe end of thisarticle.

    Accepted onDecember 14, 2019and published atascopubs.org/journal/po on May 15, 2020:DOI https://doi.org/10.1200/PO.19.00254

    534

    Downloaded from ascopubs.org by Tokyo Daigaku Ikagaku Kenkyujo on June 1, 2020 from 157.082.096.062Copyright © 2020 American Society of Clinical Oncology. All rights reserved.

    http://ascopubs.org/journal/pohttp://ascopubs.org/journal/pohttp://ascopubs.org/doi/full/10.1200/PO.19.00254http://ascopubs.org/doi/full/10.1200/PO.19.00254

  • analysis of the patient’s lesions showed that they wereCD25−. The two remaining candidates were ipilimumaband lenalidomide. The patient was reluctant to receiveipilimumab because its mechanism of action is similar tothat of pembrolizumab. Therefore, we focused on lenali-domide as a therapeutic option.

    To identify any potential predictive signatures of responseto lenalidomide therapy in this patient, we investigatedwhether there could be any overlapping lenalidomide-related gene signatures in our in-house, independentRNA sequencing data set (Appendix Figs A2-A5). Spe-cifically, we focused on available data from patients withadult T-cell leukemia/lymphoma (ATL), another mature/peripheral T-cell neoplasm in which lenalidomide dem-onstrated clinical efficacy with an overall response rate of42% in a phase II study (ATLL-002 [ClinicialTrials.govidentifier: NCT01724177]). Gene expression data fromtumors of two patients with ATL before lenalidomidetherapy were available for this purpose; one patient (uniquepatient number [UPN] 1) had an objective response tolenalidomide therapy, whereas the other patient (UPN2)was primarily refractory to lenalidomide (the patient char-acteristics are listed in Appendix Table A2). To identifymolecular signatures associated with lenalidomide re-sponse and to reduce bias derived from different dis-eases, such as ATL, we extracted differentially expressedgenes from the tumor of UPN1 (responsive ATL case)compared with expression levels in the tumor from UPN2(refractory ATL case). Overall, we found 1,250 and 1,197genes upregulated in CTCL and the current ATL case,respectively.

    KEGG pathway enrichment on the basis of the DAVIDannotation tool revealed that 73 and 88 pathways wereenriched in CTCL and ATL, respectively (Appendix Fig A3;Appendix Table A3). Among common pathways (n = 52),we identified HTLV-1 and lenalidomide target signatures(Appendix Fig A3, A7-A10; Appendix Table A3). Gene setenrichment analysis (GSEA) of panels of curated gene setsrelated to lenalidomide confirmed the enrichment of asignature predictive of response to lenalidomide in both

    CTCL and ATL. In contrast, neither signature of resistancewas enriched in CTCL and ATL (Table 2). On the basis ofthese predictive signatures and documented efficacy inpatients with CTCL,9,10 we chose lenalidomide.

    Beginning on September 11, 2017, lenalidomide was orallyadministered at a daily dose of 25 mg for 21 days of a28-day cycle while the patient had clinical stage IIB disease(Fig 1A). After the first course of treatment, all skin tumorsdisappeared except for one on his sternal region (Fig 1B). Inaddition, a transient flare reaction (TFR) that presented aserythema around skin tumors was observed during the firstcourse (Fig 1C). Aspiration pneumonia developed at theend of the first course, and thus, the dosage of the secondcourse was reduced to 10 mg daily; it was subsequentlyincreased by 5 mg every cycle to a maximum of 20 mgdaily. We extended the interval course from 1 to 2 weeksaccording to the patient’s status. Although new lesionsdisappeared after three courses of treatment, a 9-cmsternal tumor remained, but it was smaller than its pre-vious size. It was later irradiated with 20 Gy in December2017. The fifth course of lenalidomide was completed onFebruary 3, 2017, and all tumors, including sternal lesions,disappeared completely (Fig 1D).

    Upon immunohistochemical analysis of tumor specimensbefore and after lenalidomide treatment, a remarkablenumber of CD8+ cells and few CD25+ and Foxp3+ cells wereobserved in each skin tumor (Appendix Table A4; AppendixFig A6). Especially, the number of CD8+ cells after the firstcourse increased compared with that before the treatmentbut decreased in the treatment-resistant lesion after thethird course of lenalidomide.

    DISCUSSION

    To our knowledge, this report is the first of successfulclinical results in a patient with refractory SS treated withlenalidomide, which was selected on the basis of clinicalsequencing. Advances in clinical sequencing have enabledthe discovery of actionable alterations that yield clinicalbenefit.11 Patients with advanced-stage SS have an un-favorable prognosis and an unmet clinical need for effective

    TABLE 1. Actionable Alterations With DrugUpregulated Gene Drug Reason of the Exclusion

    CCR4 Mogalizumab Drug resistance

    LCK Dasatinib Lack of clinical evidence

    PDL1 PD-L1 inhibitor Drug resistance

    CTLA4 Ipilimumab Lack of consent

    CD25 Denileukin diftitox Lack of expression by immunohistochemistry

    CD30 Brentuximab vedotin Drug resistance

    Activated signaling pathway

    NF-κB Lenalidomide MTB recommendation

    NOTE. See also Table 2. Candidate genes and drugs selected by the MTB proposed the activated NF-κB pathway on the basis of gene set enrichmentanalysis results. The attending physician chose lenalidomide (off-label use) from this report.Abbreviations: MTB, molecular tumor board; NF-κB, nuclear factor-κB.

    Clinical Sequencing in a Patient With Sézary Syndrome

    JCO Precision Oncology 535

    Downloaded from ascopubs.org by Tokyo Daigaku Ikagaku Kenkyujo on June 1, 2020 from 157.082.096.062Copyright © 2020 American Society of Clinical Oncology. All rights reserved.

  • treatment. Overall survival has not been shown to benefitfrom aggressive intervention with cytotoxic agents becauseof serious treatment-associated toxicities, including pro-longed myelosuppression. An increased understanding ofthe pathogenesis of SS, with the identification of new tar-gets, has led to new therapeutic approaches with biologicagents to reduce treatment-related toxicities and improveoutcomes.1

    The rationale for lenalidomide selection by the MTB wasbased on five factors. First, lenalidomide binds the cullin4-RING-E3 ubiquitin ligase cereblon complex and de-grades lymphoid transcription factors IKZF1 and IKZF3,leading to a decrease in NF-κB.12 Second, activated tumornecrosis factor signaling, known as a lenalidomide-targetpathway in the KEGG DRUG database, was detected in thetumor, which is also considered targetable by lenalidomideon the basis of our in-house data, UPN1. Third, a phase IItrial of lenalidomide monotherapy for advanced refractoryCTCL reported reliable and meaningful clinical benefits; inthat trial, lenalidomide-induced TFR was correlated with

    good clinical response in patients with SS, which suggeststhat mechanisms that underlie this reaction might repre-sent specific anticancer immune responses.9 Indeed, in thecurrent patient, immune response to cancer was consid-ered to be induced by lenalidomide as shown by the ap-pearance of TFR (Fig 1C). Immunohistochemical stainingshowed that CD8+ cells in the tumor after the appearance ofTFR were increased after lenalidomide treatment. Fourth,lenalidomide exerts immunomodulatory effects, such asNK- and T-cell activation and the induction of T-helper1 cytokine production and cytotoxic activity; it also altersthe tumor microenvironment through its anti-angiogenic,antiproliferative, and pro-apoptotic properties.13 These char-acteristics provided the rationale for using this agent inour elderly patient with SS with susceptibility to infection.Actually, NK-cell–mediated cytotoxicity and cytokine-cytokine receptor interactions were enriched in the tu-mor before lenalidomide treatment. In addition, the fre-quencies of circulating NK cells and CD3+CD8+ T cellsincreased after the administration of lenalidomide compared

    TABLE 2. Lenalidomide-Related Gene Sets and Annotations

    Lenalidomide-Related Gene Sets and Annotations Current Patient

    A Lenalidomide-Responsive Patient

    With ATL

    Gene SetGeneSize Signature Reference NES P FDR q NES P FDR q

    HINATA_NFKB_IMMU_INF 17 Lenalidomide-responsive/target pathway

    TTD8, KEGG DRUG/pathway7

    1.88 .004 0.008 1.55 .027 0.013

    TIAN_TNF_SIGNALING_VIA_NFKB 25 Lenalidomide-responsive/target pathway

    TTD8, KEGG DRUG/pathway7

    1.67 .005 0.025 1.36 .029 0.046

    KEGG_NF-kappa_B_signaling_pathway 102 Lenalidomide-responsive/target pathway

    TTD8, KEGG DRUG/pathway7

    1.93 .000 0.005 1.38 .017 0.033

    KEGG_CYTOKINE_CYTOKINE_RECEPTOR_INTERACTION

    249 Lenalidomide-responsive/target pathway

    TTD8, KEGG DRUG/pathway7

    1.75 .000 0.005 1.58 .000 0.009

    KEGG_NATURAL_KILLER_CELL_MEDIATED_CYTOTOXICITY

    130 Lenalidomide-responsive/target pathway

    TTD8, KEGG DRUG/pathway7

    1.73 .000 0.005 1.45 .003 0.018

    KEGG_APOPTOSIS 86 Lenalidomide-responsive/target pathway

    TTD, KEGG DRUG/pathway

    1.44 .004 0.023 1.18 .118 0.114

    Aue_CLL_lenalidomide_target_Th1immunity_TableS1

    155 Lenalidomide-responsive/target pathway

    Aue17 1.93 .000 0.009 1.94 .000 0

    Bhutani_myeloma_lenalidomide_target_genes_TableS2

    147 Lenalidomide-responsive/target pathway

    Bhutani18 1.22 .035 0.104 1.60 .000 0.009

    E2F3_UP.V1_UP 186 Lenalidomide resistance Neri19 1.23 .029 0.284 NE

    KRAS.600.LUNG.BREAST_UP.V1_UP 271 Lenalidomide resistance Neri19 1.23 .320 0.220 1.42 0.062

    KRAS.KIDNEY_UP.V1_UP 139 Lenalidomide resistance Neri19 NE NE

    KRAS.LUNG_UP.V1_UP 136 Lenalidomide resistance Neri19 1.44 .005 0.206 1.00 .452 0.486

    RAF_UP.V1_DN 183 Lenalidomide resistance Neri19 NE 1.12 .181 0.358

    MEK_UP.V1_DN 182 Lenalidomide resistance Neri19 NE 1.03 .388 0.445

    RPS14_DN.V1_DN 179 Lenalidomide resistance Neri19 1.12 .109 0.333 NE

    AKT_UP_MTOR_DN.V1_DN 173 Lenalidomide resistance Neri19 NE 1.12 .217 0.334

    STK33_DN 256 Lenalidomide resistance Neri19 NE 1.33 .004 0.103

    Abbreviations: ATL, adult T-cell leukemia/lymphoma; FDR, false discovery rate; KEGG, Kyoto Encyclopedia of Genes and Genomes; NE, not enriched; NES,normalized enrichment score; TTD, Therapeutic Target Database.

    Hijikata et al

    536 © 2020 by American Society of Clinical Oncology

    Downloaded from ascopubs.org by Tokyo Daigaku Ikagaku Kenkyujo on June 1, 2020 from 157.082.096.062Copyright © 2020 American Society of Clinical Oncology. All rights reserved.

  • with those at first treatment (Appendix Fig A11). Incontrast, the frequency of effector regulatory T cells didnot increase (Appendix Fig A11). These findings weresimilar to those described in the immunohistochemistry(Appendix Fig A6). These immunologic changes mighthave induced anticancer effects9 and prevented severeinfection.14 Finally, the Japanese national health in-surance system has already approved lenalidomide for thetreatment of myelodysplastic syndrome, refractory/relapsemultiple myeloma, and ATL.

    Lenalidomide did not adequately treat the patient’s bulkysternal mass. Acquired lenalidomide resistance is con-sidered to be caused by reduced levels of tumor cellcereblon protein because of epigenetic modifications ofthe cereblon promotor region, cereblon gene mutations or

    chromosomal deletions, or acquired cereblon pathway mu-tations and detects in other components of the E3 ligasecomplex.15 Another possible reason is that the delivery oflenalidomide to the tumor was hindered by the abundantsurrounding interstitial tissue and the immunosuppressivetumor microenvironment.16

    In the near future, clinical sequencing will support clinicaldecisions with regard to appropriate treatments for patientswith rare and refractory cancers. In addition, clinical se-quencing on the basis of a combination of several NGStechniques might be particularly useful in identifying ap-propriate treatment options. This case report is the first toour knowledge of the use of clinical sequencing to iden-tify a successful treatment strategy for a patient withrefractory SS.

    AFFILIATIONS1Department of General Medicine, IMSUT Hospital, Institute of MedicalScience, The University of Tokyo, Tokyo, Japan2Department of Hematology/Oncology, IMSUT Hospital, Institute ofMedical Science, The University of Tokyo, Tokyo, Japan3Department of Applied Genomics, Research Hospital, Institute ofMedical Science, The University of Tokyo, Tokyo, Japan4Laboratory of DNA Information Analysis, Human Genome Center,Institute of Medical Science, The University of Tokyo, Tokyo, Japan5Laboratory of Tumor Cell Biology, Department of Computational Biologyand Medical Sciences, Graduate School of Frontier Sciences, TheUniversity of Tokyo, Tokyo, Japan6Division of Molecular Therapy, Advanced Clinical Research Center,Institute of Medical Science, The University of Tokyo, Tokyo, Japan

    7Department of Diagnostic Pathology, IMSUT Hospital, Institute ofMedical Science, The University of Tokyo, Tokyo, Japan8Division of Health Medical Data Science, Health Intelligence Center,Institute of Medical Science, The University of Tokyo, Tokyo, Japan

    CORRESPONDING AUTHORArinobu Tojo, MD, PhD, Institute of Medical Science, The University ofTokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo 108-8639, Japan; e-mail:[email protected].

    EQUAL CONTRIBUTIONY.H. and K.Y. contributed equally as co-first authors.K.Y. and A.T. contributed equally as senior authors.

    A B

    DC

    FIG 1. Changes in the patient’s skin lesions during lenalidomide treatment. (A) The skin lesions on the trunk andarms before lenalidomide treatment. (B) After the first treatment course, almost all skin lesions except for that onthe patient’s sternal region disappeared. (C) Documented transient flare reaction presented as erythema aroundskin lesions. (D) All lesions, including the sternal lesion, disappeared after the fifth cycle.

    Clinical Sequencing in a Patient With Sézary Syndrome

    JCO Precision Oncology 537

    Downloaded from ascopubs.org by Tokyo Daigaku Ikagaku Kenkyujo on June 1, 2020 from 157.082.096.062Copyright © 2020 American Society of Clinical Oncology. All rights reserved.

    mailto:[email protected]

  • SUPPORTSupported in part by the Center of Innovation Program of the JapanScience and Technology Agency and Ministry of Education, Culture,Sports, Science and Technology Innovative Area 15H05912.

    AUTHOR CONTRIBUTIONSConception and design: Kazuaki Yokoyama, Arinobu TojoFinancial support: Kenzaburo Tani, Satoru MiyanoAdministrative support: Hiroshi YotsuyanagiProvision of study material or patients: Makoto Nakashima, Yasunori Ota,Kaoru UchimaruCollection and assembly of data: Yasuki Hijikata, Kazuaki Yokoyama,Makoto Yamagishi, Lay Ahyoung Lim, Kaoru UchimaruData analysis and interpretation: Yasuki Hijikata, Kazuaki Yokoyama,Nozomi Yokoyama, Yasuo Matsubara, Eigo Shimizu, Makoto Nakashima,Yasunori Ota, Rui Yamaguchi, Mika Ito, Yukihisa Tanaka, TamamiDenda, Kenzaburo Tani, Hiroshi Yotsuyanagi, Seiya Imoto, SatoruMiyanoManuscript writing: All authorsFinal approval of manuscript: All authorsAccountable for all aspects of the work: All authors

    DATA AVAILABILITY STATEMENTBoth sequence data and materials described in this article are availableupon request to the corresponding author K.Y. ([email protected]), but the request must include a description of the researchproposal.

    AUTHORS’ DISCLOSURES OF POTENTIAL CONFLICTS OFINTERESTThe following represents disclosure information provided by authors ofthis manuscript. All relationships are considered compensated unlessotherwise noted. Relationships are self-held unless noted. I = Immediate

    Family Member, Inst = My Institution. Relationships may not relate to thesubject matter of this manuscript. For more information about ASCO’sconflict of interest policy, please refer to www.asco.org/rwc or ascopubs.org/po/author-center.Open Payments is a public database containing information reported bycompanies about payments made to US-licensed physicians (OpenPayments).

    Rui YamaguchiHonoraria: Janssen Pharmaceuticals, Chugai Pharma

    Kenzaburo TaniStock and Other Ownership Interests: Shinnihonseiyaku Co Ltd, PrecisionTherapeutics, Oncolys BioPharmaConsulting or Advisory Role: Precision TherapeuticsResearch Funding: Shinnihonseiyaku Co Ltd, Neopharma Japan,Takara Bio

    Kaoru UchimaruHonoraria: CelgeneResearch Funding: Daiichi Sankyo, Celgene, NEC

    Arinobu TojoHonoraria: Otsuka PharmaceuticalConsulting or Advisory Role: SysmexSpeakers’ Bureau: Novartis, Otsuka Pharmaceutical, Bristol-MyersSquibbResearch Funding: Chugai Pharma, Torii Pharmaceutical, KM Biologics

    No other potential conflicts of interest were reported.

    ACKNOWLEDGMENTWe thank all members of the study team as well as the patients and theirfamilies.

    REFERENCES1. Agar NS, Wedgeworth E, Crichton S, et al: Survival outcomes and prognostic factors in mycosis fungoides/Sézary syndrome: Validation of the revised In-

    ternational Society for Cutaneous Lymphomas/European Organisation for Research and Treatment of Cancer staging proposal. J Clin Oncol 28:4730-4739,2010

    2. Wilcox RA: Cutaneous T-cell lymphoma: 2017 update on diagnosis, risk-stratification, and management. Am J Hematol 92:1085-1102, 2017

    3. Wang L, Ni X, Covington KR, et al: Genomic profiling of Sézary syndrome identifies alterations of key T cell signaling and differentiation genes. Nat Genet47:1426-1434, 2015

    4. Wakai T, Prasoon P, Hirose Y, et al: Next-generation sequencing-based clinical sequencing: Toward precision medicine in solid tumors. Int J Clin Oncol24:115-122, 2019

    5. Patel NM, Michelini VV, Snell JM, et al: Enhancing next-generation sequencing-guided cancer care through cognitive computing. Oncologist 23:179-185, 2018

    6. KEGG: Kyoto Encyclopedia of Genes and Genomes. http://www.kegg.jp or http://www.genome.jp/kegg

    7. Kyoto Encyclopedia of Genes and Genomes: KEGG DRUG: Lenalidomide. https://www.kegg.jp/dbget-bin/www_bget?D04687+-us

    8. Innovative Drug Research and Bioinformatics Group: Therapeutic Target Database. http://db.idrblab.net/ttd/data/drug/details/d0q5nx

    9. Querfeld C, Rosen ST, Guitart J, et al: Results of an open-label multicenter phase 2 trial of lenalidomidemonotherapy in refractory mycosis fungoides and Sézarysyndrome. Blood 123:1159-1166, 2014

    10. Bagot M, Hasan B, Whittaker S, et al: A phase III study of lenalidomide maintenance after debulking therapy in patients with advanced cutaneous T-celllymphoma - EORTC 21081 (NCT01098656): Results and lessons learned for future trial designs. Eur J Dermatol 27:286-294, 2017

    11. Damodaran S, Berger MF, Roychowdhury S: Clinical tumor sequencing: Opportunities and challenges for precision cancer medicine. Am Soc Clin Oncol EducBook 35:e175-e182, 2015

    12. Kritharis A, Coyle M, Sharma J, et al: Lenalidomide in non-Hodgkin lymphoma: Biological perspectives and therapeutic opportunities. Blood 125:2471-2476,2015

    13. Kotla V, Goel S, Nischal S, et al: Mechanism of action of lenalidomide in hematological malignancies. J Hematol Oncol 2:36, 2009

    14. Lenartić M, Jelenčić V, Zafirova B, et al: NKG2D promotes B1a cell development and protection against bacterial infection. J Immunol 198:1531-1542, 2017

    15. Franssen LE, Nijhof IS, Couto S, et al: Cereblon loss and up-regulation of c-Myc are associated with lenalidomide resistance in multiple myeloma patients.Haematologica 103:e368-e371, 2018

    16. Sriraman SK, Aryasomayajula B, Torchilin VP: Barriers to drug delivery in solid tumors. Tissue Barriers 2:e29528, 2014

    17. Aue G, Sun C, Liu D, et al: Activation of Th1 immunity within the tumor microenvironment is associated with clinical response to lenalidomide in chroniclymphocytic leukemia. J Immunol 201:1967-1974, 2018

    Hijikata et al

    538 © 2020 by American Society of Clinical Oncology

    Downloaded from ascopubs.org by Tokyo Daigaku Ikagaku Kenkyujo on June 1, 2020 from 157.082.096.062Copyright © 2020 American Society of Clinical Oncology. All rights reserved.

    http://www.asco.org/rwchttps://ascopubs.org/po/author-centerhttps://ascopubs.org/po/author-centerhttps://openpaymentsdata.cms.gov/https://openpaymentsdata.cms.gov/http://www.kegg.jp or http://www.genome.jp/kegghttps://www.kegg.jp/dbget-bin/www_bget?D04687+-ushttp://db.idrblab.net/ttd/data/drug/details/d0q5nx

  • 18. Bhutani M, Zhang Q, Friend R, et al: Investigation of a gene signature to predict a gene signature to predict response to immunomodulatory derivatives forpatients with multiple myeloma: An exploratory, retrospective study using microarray datasets from prospective clinical trials. Lancet Haematol 4:e443-e451,2017

    19. Neri P, Gratton K, Ren L, et al: RNA-sequencing of paired pre-treatment and relapse samples reveals differentially expressed genes associated withlenalidomide resistance in multiple myeloma. Blood 122:530, 2013

    n n n

    Clinical Sequencing in a Patient With Sézary Syndrome

    JCO Precision Oncology 539

    Downloaded from ascopubs.org by Tokyo Daigaku Ikagaku Kenkyujo on June 1, 2020 from 157.082.096.062Copyright © 2020 American Society of Clinical Oncology. All rights reserved.

  • APPENDIX

    Materials and Methods

    Patients and samples. Our study protocol was approved by ourinstitutional research ethics committee and was in accordance with theDeclaration of Helsinki. Three patients were recruited for our study, allof whom provided written informed consent. The characteristics of tworeference patients with adult T-cell leukemia/lymphoma (ATL) arelisted in Table A2. The following samples were collected: (1) the tumor(cutaneous T-cell lymphoma [CTCL]), patient’s skin tumor samplebefore lenalidomide therapy, and control (CTCL), patient’s tumor-freenormal skin lesion, which had been histologically confirmed, and (2)the tumor (ATL), tumor-rich peripheral blood mononuclear cells(PBMNCs) before lenalidomide therapy in a patient with ATL(uniquepatient number 1 [UPN1]) for whom clinical partial response wasobserved under lenalidomide therapy, and control (ATL), tumor-richPBMNCs before lenalidomide therapy in another patient with ATL(UPN2) for whom no objective response had been observed withlenalidomide therapy.

    Extraction of DNA and RNA. DNA was extracted using the GentraPuregene Blood Kit and the QIAamp Circulating Nucleic Acid Kit(QIAGEN, Hilden, Germany). DNA was quantified using the QubitdsDNA HS Assay Kit (Life Technologies, Carlsbad, CA). Total RNA wasextracted using the RNeasy Mini Kit (QIAGEN).

    Whole-exome sequencing. To identify driver mutations, we per-formed whole-exome sequencing (WES). Libraries were prepared from200 ng of paired DNA (tumor/control) using SureSelectXT Human AllExon V5 (Agilent Technologies, Santa Clara, CA). WES was performedon the basis of 2× 100-base pair (bp) paired-end reads on the NextSeq500 platform with NextSeq 500/550 Kit v2 chemistry (Illumina, SanDiego, CA). All procedures were performed according to the manu-facturers’ protocols.

    Somatic variant calling and subsequent primary filtering ofWES data. We used human genome build 19 as the referencesequence for our analysis. Primary processing of sequencing data wasperformed using our in-house software. Single nucleotide variantswere identified using Genomon2 (http://genomon.hgc.jp/exome/en/)with default parameters. All variant calls were annotated withANNOVAR (using an in-house annotation database resource). TheIntegrative Genomics Viewer (IGV) version 2.3.57 (https://software.broadinstitute.org/software/igv/download) was used to visualize andinspect the read alignments and variant calls. For our analysis, thedatabases used to annotate variants included the following: RefSeq(http://www.ncbi.nlm.nih.gov/RefSeq/), the 1000 Genomes Projectas of August 2015 (http://www.internationalgenome.org/data),dbSNP131 (http://www.ncbi.nlm.nih.gov/projects/SNP/), ToMMoversion 1 (https://ijgvd.megabank.tohoku.ac.jp/), the Human GeneticVariation Database as of October 2013 (http://www.hgvd.genome.med.kyoto-u.ac.jp/), ClinVar as of June 2015 (https://www.ncbi.nlm.nih.gov/clinvar/), the Human Gene Mutation Database Professional asof March 2015 (http://www.hgmd.cf.ac.uk/ac/index.php), the cBio-Portal for Cancer Genomics as of September 2015 (http://cbioportal.org), the TumorPortal as of September 2015 (http://www.tumorportal.org/), the Catalogue of Somatic Mutations in Cancer (COSMIC) version70 (http://cancer.sanger.ac.uk/cosmic), and the International CancerGenome Consortium Data Portal (https://dcc.icgc.org/). The compu-tational algorithms Sorting Intolerant From Tolerant (http://sift-dna.org)and PolyPhen-2 (http://genetics.bwh.harvard.edu/pph2/) were used topredict whether mutations were damaging. Synonymous or noncodingvariants were excluded, and single nucleotide polymorphisms (SNPs;minor allele frequency . 1%) reported to be present in either of theaforementioned SNP databases were automatically excluded, unlessthey were recurrently (three or more times) found in the COSMICdatabase. Copy number variation analysis was performed using CNVkitversion 0.8.2 software (E. Talevich, University of Southern California,San Francisco, San Francisco, CA), and the data were filtered on thebasis of the following parameters: log2 ≥ 0.6 or log2 ≤ −1 and read

    depth . 10. Upon somatic mutation calling followed by a primaryfiltering step, the list of variants was stored in a variant calling file format(.vcf). Gene-level copy number alterations, as a log2 ratio of tumor tonormal, were also stored in text format (.log2).

    Identification of putative driver mutations fromWES data. Weclassified variants into three tiers. We regarded tier 1 mutations asdefinitive driver mutations. Briefly, tier 1 included definitive drivermutations with abundant evidence in the literature and/or cancermutation databases with a variant frequency (VAF) of ≥ 5%. Tier2 included mutations with a little or no evidence in the literature and/ordatabases, namely, uncertain/unclassified significance, with VAF≥ 5%. Tier 3 included all remaining variants. The selection of mutationcriteria for tier 1 was as follows: all changes in the amino acid codingregions of annotated exons that were frequently (at least three times)reported in hematopoietic tumor samples in the COSMIC database(version 71) or all predictive, highly deleterious variants (in consensussplice site regions, frameshift mutations, any premature predictedtruncation mutation, or large in-frame insertions/deletions) in non-Hodgkin lymphoma or CTCL-associated genes, according to previousstudies (Ungewickell A, et al: Nat Genet 47:1056-1060, 2015; Choi J,et al: Nat Genet 47:1011-1019, 2015; Lohr JG, et al: Proc Natl AcadSci U S A 109:3879-3884, 2012). Curated tier 1-2 somatic mutationsare listed in Table A1.

    RNA SequencingWe used the following RNA samples: the tumor (CTCL), patient’s skintumor sample before lenalidomide therapy; control (CTCL), and pa-tient’s tumor-free normal skin lesion; the tumor (ATL) and tumor-richPBMNCs in lenalidomide-responsive ATL (UPN1); and the control(ATL) and tumor-rich PBMNCs in lenalidomide-primary refractory ATL(UPN2). cDNA libraries for next-generation sequencing were con-structed from 100 ng of total RNA using TruSeq RNA Access LibraryPrep Kit (Illumina). Each paired end–indexed library was sequenced toa length of 75 nucleotides per pair (2 × 75 bp) using a NextSeq in-strument. Sequence reads were processed by our in-house Genomon-RNA/Genomon-expression pipeline (Shiraishi Y, et al: PLoS One 9:e114263, 2014; http://genomon.hgc.jp/rna/). IGV version 2.3.57 wasused to visualize the mapped sequence. For gene expression, frag-ments per kilobase of gene model per million mapped read (FPKM)values were calculated and used.

    To investigate any changes in gene expression in a certain enrichedpathway involved in the clinical features of the current patient withCTCL, as well as the reference patients with ATL, we used the onlineDAVID tool version 6.8 (https://david.ncifcrf.gov/) and gene set en-richment analysis (GSEA) with FPKMmetrics at the gene level (tumor vcontrol). For DAVID-based pathway analysis, upregulated genes rel-ative to expression in the control (fold-change ≥ 2 and with a cutoffvalue of FPKM ≥ 10) were stored in comma-separated value format(.csv) and inputted into the web-based interface of DAVID. Theseoutputs were reviewed, and significantly enriched Kyoto Encyclopediaof Genes and Genomes (KEGG) pathways are listed in Table A3. Thefour significant lenalidomide-target pathways were also visualizedusing Pathview (https://pathview.uncc.edu/) and displayed in FiguresA7-A10. For GSEA, a stand-alone application was used (Broad In-stitute, Cambridge, MA; http://broadinstitute.org/gsea/index.jsp) withthe MSIGDB library version 7.1 as well as in-house–created gene setsthat were based on a false discovery rate threshold of q , 0.05.

    ImmunohistochemistryFormalin-fixed paraffin embedded tissue sections of 3-μm thick-ness were deparaffinized in xylene and rehydrated in a series ofgraded alcohols and heated in an antigen retrieval solution at pH9.0 (Vector Laboratories, Burlingame, CA) for 10 minutes at 120°C.Endogenous peroxidase was blocked by incubation with 3% hy-drogen peroxidase for 5 minutes at room temperature. Next,sections were incubated with the following primary antibodies for30 minutes at 37°C: CD4 (clone EPR6855, dilution 1:250; Abcam,Cambridge, United Kingdom), CD8 (clone SP16, dilution 1:100;

    Hijikata et al

    540 © 2020 by American Society of Clinical Oncology

    Downloaded from ascopubs.org by Tokyo Daigaku Ikagaku Kenkyujo on June 1, 2020 from 157.082.096.062Copyright © 2020 American Society of Clinical Oncology. All rights reserved.

    http://genomon.hgc.jp/exome/en/https://software.broadinstitute.org/software/igv/downloadhttps://software.broadinstitute.org/software/igv/downloadhttp://www.ncbi.nlm.nih.gov/RefSeq/http://www.internationalgenome.org/datahttp://www.ncbi.nlm.nih.gov/projects/SNP/https://ijgvd.megabank.tohoku.ac.jp/http://www.hgvd.genome.med.kyoto-u.ac.jp/http://www.hgvd.genome.med.kyoto-u.ac.jp/https://www.ncbi.nlm.nih.gov/clinvar/https://www.ncbi.nlm.nih.gov/clinvar/http://www.hgmd.cf.ac.uk/ac/index.phphttp://cbioportal.orghttp://cbioportal.orghttp://www.tumorportal.org/http://www.tumorportal.org/http://cancer.sanger.ac.uk/cosmichttps://dcc.icgc.org/http://sift-dna.orghttp://genetics.bwh.harvard.edu/pph2/http://genomon.hgc.jp/rna/https://david.ncifcrf.gov/https://pathview.uncc.edu/http://broadinstitute.org/gsea/index.jsp

  • Abcam), CD25 (clone 305, dilution 1:80; Leica Biosystems,Newcastle, United Kingdom), and Foxp3 (clone SP97, dilution 1:80; Abcam). After washing, sections were treated with secondaryantibody (EnVision+ Dual Link System-HRP; Dako, Glostrup,Denmark) for 30 minutes at 37°C. To visualize antigen-antibody

    complex, EnVision+ DAB kit (Dako) was used, and then sectionswere counterstained in hematoxylin. Normal lymph node wasplaced on the same slide glass as a positive control, and negativecontrols were used in phosphate-buffered saline instead of theprimary antibodies.

    Tumor DNAskin tumor

    Control DNAnormal tumor

    Somatic mutation call byGenomon pipeline

    Copy number alterationcall by CNVkit pipeline

    1. Variants

    .vcf file

    2. Copy number alterations

    .log2 file

    Annotation by ANNOVARwith in-house medicalinformatics pipeline

    Curation of candidategenes

    MTBActionable gene alteration

    WES on NextSeq(Illumina)

    FIG A1. Flowchart of whole-exome sequencing. MTB,molecular tumor board; vcf, variant calling file; WES, wholeexome sequencing.

    Clinical Sequencing in a Patient With Sézary Syndrome

    JCO Precision Oncology 541

    Downloaded from ascopubs.org by Tokyo Daigaku Ikagaku Kenkyujo on June 1, 2020 from 157.082.096.062Copyright © 2020 American Society of Clinical Oncology. All rights reserved.

  • Curation of candidate genes

    Identification of lenalidomidetarget signature

    Prediction of lenalidomideResponse in current patient

    with CTCL

    MTBActionable gene alteration

    Tumor RNA-seqlenalidomide-responsive ATL (UPN1) PBMNC

    Control RNA-seqlenalidomide-refractory ATL (UPN2) PBMNC

    Tumor RNAskin tumor

    Control RNAnormal tumor

    Present CTCL case

    RNA-seq on NextSeq(Illumina)

    Transcript assembly/quantification (FPKM)by Genomon RNA

    Differential gene expression Fold change ≥ 2 Tumor (or control) FPKM ≥10 .dge file

    Exploring overlapping lenalidomide targetSignature in in-house RNA-seq data:A lenalidomide-responsive patient with ATL (Fig A3)

    Functional–annotation/pathway analysis

    GSEA DAVID

    FIG A2. Flow of overall RNA sequencing. ATL, adult T-cell leukemia/lymphoma; CTCL, cutaneous T-cell lymphoma;.dge, digital gene expression; FPKM, fragments per kilobase of gene model per million mapped read; GSEA, gene setenrichment analysis; Len, lenalidomide; MTB, molecular tumor board; PBMNC, peripheral blood mononuclear cell;RNA-seq, RNA sequencing; UPN, unique patient number.

    Hijikata et al

    542 © 2020 by American Society of Clinical Oncology

    Downloaded from ascopubs.org by Tokyo Daigaku Ikagaku Kenkyujo on June 1, 2020 from 157.082.096.062Copyright © 2020 American Society of Clinical Oncology. All rights reserved.

  • RNA-seq: tumor v control

    Differential gene expression,upregulated in tumor (v control):

    1,250 genes (patient with CTCL) 1,197 genes (patient with ATL)

    Pathway analysis by DAVID (KEGG pathway)

    Curation of overlapping KEGG pathways

    1. Lenalidomide target pathway in KEGG-DRUG/pathway

    KEGG_NF-kappa_B_signaling_pathway

    KEGG_CYTOKINE_CYTOKINE_RECEPTOR_INTERACTION

    KEGG_NATURAL_KILLER_CELL_MEDIATED_CYTOTOXICITY

    KEGG_APOPTOSIS

    2. HTLV-1 signature

    KEGG_HTLV-1 infection

    Identification of HTLV-1 + lenalidomide target pathway Confirmation of lenalidomide target signature by GSEA analysis

    Current patient with CTCL

    73 52 88

    Patient with ATL

    FIG A3. Flow chart of overlapping lenalidomide (Len) target signaturediscovery. ATL, adult T-cell leukemia/lymphoma; CTCL, cutaneousT-cell lymphoma; GSEA, gene set enrichment analysis; KEGG, KyotoEncyclopedia of Genes and Genomes; RNA-seq, RNA sequencing.

    Clinical Sequencing in a Patient With Sézary Syndrome

    JCO Precision Oncology 543

    Downloaded from ascopubs.org by Tokyo Daigaku Ikagaku Kenkyujo on June 1, 2020 from 157.082.096.062Copyright © 2020 American Society of Clinical Oncology. All rights reserved.

  • Rank

    ed L

    ist M

    etric

    (Sig

    nal2

    Noi

    se)

    Enric

    hmen

    tSc

    ore

    –5.0–2.50.02.55.0

    0.00.10.20.30.40.5

    0 5,000 10,000 15,000 20,000 25,000

    Rank in Ordered Data Set

    Enrichment plot: GEORG ETALJOURNALOFIMMUNOLOGYTABLES2_LENALIDOMIDE-CLL_ALL_DAY8TODAY0

    Zero cross at 7660

    ‘TUMOR’ (positively correlated)

    ‘NORMAL’ (negatively correlated)

    Enrichment plot: KEGG-NF-KAPPAB SIGNALING PATHWAY

    Rank

    ed L

    ist M

    etric

    (Sig

    nal2

    Noi

    se)

    Enric

    hmen

    tSc

    ore

    –5.0–2.50.02.55.0

    0.00.10.20.30.40.50.6

    0 5,000 10,000 15,000 20,000 25,000

    Rank in Ordered Data Set

    Zero cross at 7660

    ‘TUMOR’ (positively correlated)

    ‘NORMAL’ (negatively correlated)

    Enrichment plot:HINATA_NFKB_IMMU_INF

    Rank

    ed L

    ist M

    etric

    (Sig

    nal2

    Noi

    se)

    Enric

    hmen

    tSc

    ore

    –5.0–2.50.02.55.0

    0.00.10.20.30.40.5

    0.70.6

    0.8

    0 5,000

    Zero cross at 7660

    ‘TUMOR’ positively correlated)

    ‘NORMAL’ (negatively correlated)

    10,000 15,000 20,000 25,000

    Rank in Ordered Data Set

    Rank

    ed L

    ist M

    etric

    (Sig

    nal2

    Noi

    se)

    Enric

    hmen

    tsc

    ore

    –5.0–2.50.02.55.0

    0.450.400.350.300.250.200.150.100.050.00

    –0.05–0.10

    0 5,000 10,000 15,000 20,000 25,000

    Rank in Ordered Data Set

    Enrichment plot: KEGG_APOPTOSIS

    Zero cross at 7660

    ‘TUMOR’ (positively correlated)

    ‘NORMAL’ (negatively correlated)

    Zero cross at 7660

    ‘TUMOR’ (positively correlated)

    ‘NORMAL’ (negatively correlated)

    Rank

    ed L

    ist M

    etric

    (Sig

    nal2

    Noi

    se)

    Enric

    hmen

    tSc

    ore

    –5.0–2.50.02.55.0

    –0.1–0.2

    0.00.10.20.3

    0 5,000 10,000 15,000 20,000 25,000

    Rank in Ordered Data Set

    Enrichment plot:LANCETHEMATOLOGYTABLES2_ALL

    Enrichment profile Hits Ranking metric scores Enrichment profile Hits Ranking metric scores

    Rank

    ed L

    ist M

    etric

    (Sig

    nal2

    Noi

    se)

    Enric

    hmen

    tSc

    ore

    –5.0–2.50.02.55.0

    0.00.10.20.30.4

    0.60.5

    0.7

    0 5,000 10,000 15,000 20,000 25,000

    Rank in Ordered Data Set

    Enrichment plot:TIAN_TNF_SIGNALING_VIA_NFKB

    Zero cross at 7660

    ‘TUMOR’ (positively correlated)

    ‘NORMAL’ (negatively correlated)

    Zero cross at 7660

    ‘TUMOR’ (positively correlated)

    ‘NORMAL’ (negatively correlated)

    Rank

    ed L

    ist M

    etric

    (Sig

    nal2

    Noi

    se)

    Enric

    hmen

    tSc

    ore

    –5.0–2.50.02.55.0

    0.0–0.1–0.2

    0.10.20.30.40.5

    0 5,000 10,000 15,000 20,000 25,000

    Rank in Ordered Data Set

    Enrichment plot:KEGG_CYTOKINE_CYTOKINE_

    RECEPTOR_INTERACTION

    Zero cross at 7660

    ‘TUMOR’ positively correlated)

    Enrichment plot:KEGG_NATURAL_KILLER_

    CELL_MEDIATED_CYTOTOXICITY

    ‘NORMAL’ (negatively correlated)

    Rank

    ed L

    ist M

    etric

    (Sig

    nal2

    Noi

    se)

    Enric

    hmen

    tSc

    ore

    –5.0–2.50.02.55.0

    0.0–0.1

    0.10.20.30.40.5

    0 5,000 10,000 15,000 20,000 25,000

    Rank in Ordered Data SetEnrichment profile Hits Ranking metric scores Enrichment profile Hits Ranking metric scores Enrichment profile Hits Ranking metric scores

    Enrichment profile Hits Ranking metric scores Enrichment profile Hits Ranking metric scores Enrichment profile Hits Ranking metric scores

    FIG A4. Lenalidomide-target signature enriched in cutaneous T-cell lymphoma (CTCL) on the basis of gene set enrichment analysis (GSEA; related toTable 1). Enrichment plots and heat maps from GSEA in the patient. GSEA of the tumor disclosed significant enrichment of nuclear factor-κB (NF-κB)pathways, including that of a custom gene set, GEORG ETALJOURNALOFIMMUNOLOGYTABLES2_LENALIDOMIDE-CLL_ALL_DAY8TODAY0, on the basisof the report and Supplementary Table 2 by Aue et al17 as follows: NATURAL_KILLER_CELL_MEDIATED_CYTOTOXICITY from the MSIGDB library,CYTOKINE_CYTOKINE_RECEPTOR_INTERACTION from the MSIGDB library, TIAN_TNF_SIGNALING_VIA_NFKB and APOPTOSIS from the MSIGDBlibrary, and a custom gene set, LANCETHEMATOLOGYTABLES2_ALL, on the basis of the report and Supplementary Table 2 by Bhutani et al18. Red indicatesthat gene expression increased compared with that in control. Blue indicates that gene expression decreased compared with that in control. KEGG, KyotoEncyclopedia of Genes and Genomes.

    Hijikata et al

    544 © 2020 by American Society of Clinical Oncology

    Downloaded from ascopubs.org by Tokyo Daigaku Ikagaku Kenkyujo on June 1, 2020 from 157.082.096.062Copyright © 2020 American Society of Clinical Oncology. All rights reserved.

  • Enrichment plot:KEGG_CYTOKINE_CYTOKINE_

    RECEPTOR_INTERACTION

    Rank

    ed L

    ist M

    etric

    (Sig

    nal2

    Noi

    se)

    Enric

    hmen

    tSc

    ore

    –5.0–2.50.02.55.0

    0.00.10.20.30.4

    0.60.5

    0 5,000 10,000 15,000 20,000 25,000

    Rank in Ordered Data Set

    Enrichment plot: GEORG ETALJOURNALOFIMMUNOLOGYTABLES2_LENALIDO MIDE-CLL_ALL_DAY8TODAY0

    Zero cross at 8016

    ‘TUMOR’ (positively correlated)

    ‘NORMAL’ (negatively correlated)

    Enrichment plot:LANCETHEMATOLOGYTABLES2_ALL

    Rank

    ed L

    ist M

    etric

    (Sig

    nal2

    Noi

    se)

    Enric

    hmen

    tSc

    ore

    –5.0–2.50.02.55.0

    0.0–0.1

    0.10.20.30.40.5

    0 5,000 10,000 15,000 20,000 25,000

    Rank in Ordered Data Set

    Zero cross at 8016

    ‘TUMOR’ (positively correlated)

    ‘NORMAL’ (negatively correlated)

    Rank

    ed L

    ist M

    etric

    (Sig

    nal2

    Noi

    se)

    Enric

    hmen

    tSc

    ore

    –5.0–2.50.02.55.0

    0.0–0.1

    0.10.20.30.40.5

    0 5,000

    Zero cross at 8016

    ‘TUMOR’ (positively correlated)

    ‘NORMAL’ (negatively correlated)

    10,000 15,000 20,000 25,000

    Rank in Ordered Data SetEnrichment profile Hits Ranking metric scores Enrichment profile Hits Ranking metric scores Enrichment profile Hits Ranking metric scores

    Rank

    ed L

    ist M

    etric

    (Sig

    nal2

    Noi

    se)

    Enric

    hmen

    tSc

    ore

    –5.0–2.50.02.55.0

    0.0–0.1

    0.10.20.30.4

    0.60.5

    0 5,000 10,000 15,000 20,000 25,000

    Rank in Ordered Data Set

    Enrichment plot:TIAN_TNF_SIGNALING_VIA_NFKB

    Zero cross at 8016

    ‘TUMOR’ (positively correlated)

    ‘NORMAL’ (negatively correlated)

    Zero cross at 8016

    ‘TUMOR’ (positively correlated)

    ‘NORMAL’ (negatively correlated)

    Rank

    ed L

    ist M

    etric

    (Sig

    nal2

    Noi

    se)

    Enric

    hmen

    tSc

    ore

    –5.0–2.50.02.55.0

    0.00–0.05–0.10

    0.100.05

    0.150.200.250.300.350.400.45

    0 5,000 10,000 15,000 20,000 25,000

    Rank in Ordered Data Set

    Enrichment plot:KEGG_APOPTOSIS

    Enrichment profile Hits Ranking metric scores Enrichment profile Hits Ranking metric scores

    Rank

    ed L

    ist M

    etric

    (Sig

    nal2

    Noi

    se)

    Enric

    hmen

    tSc

    ore

    –5.0–2.50.02.55.0

    0.00.10.20.30.4

    0.60.5

    0.7

    0 5,000 10,000 15,000 20,000 25,000

    Rank in Ordered Data Set

    Enrichment plot:HINATA_NFKB_IMMU_INF

    Zero cross at 8016

    ‘TUMOR’ (positively correlated)

    ‘NORMAL’ (negatively correlated)

    Zero cross at 8016

    ‘TUMOR’ (positively correlated)

    ‘NORMAL’ (negatively correlated)

    Rank

    ed L

    ist M

    etric

    (Sig

    nal2

    Noi

    se)

    Enric

    hmen

    tSc

    ore

    –5.0–2.50.02.55.0

    0.0–0.1

    0.10.20.30.40.5

    0 5,000 10,000 15,000 20,000 25,000

    Rank in Ordered Data Set

    Enrichment plot:KEGG_NATURAL_KILLER_CELL_

    MEDIATED_CYTOTOXICITY

    Zero cross at 8016

    ‘TUMOR’ (positively correlated)

    Enrichment plot:KEGG-NF-KAPPA B SIGNALING PATHWAY

    ‘NORMAL’ (negatively correlated)

    Rank

    ed L

    ist M

    etric

    (Sig

    nal2

    Noi

    se)

    Enric

    hmen

    tSc

    ore

    –5.0–2.50.02.55.0

    0.0–0.1

    0.10.20.30.40.5

    0 5,000 10,000 15,000 20,000 25,000

    Rank in Ordered Data SetEnrichment profile Hits Ranking metric scores Enrichment profile Hits Ranking metric scores Enrichment profile Hits Ranking metric scores

    FIG A5. Lenalidomide-target signature enriched in adult T-cell leukemia/lymphoma (ATL) on the basis of gene set enrichment analysis (GSEA; related toTable 1). Enrichment plots and heat maps from GSEA of ATL. In relation to Fig A4, GSEA of the tumor disclosed significant enrichment of nuclear factor-κB(NF-κB) pathways, including the gene sets of NATURAL_KILLER_CELL_MEDIATED_CYTOTOXICITY, CYTOKINE_CYTOKINE_RECEPTOR_INTERACTION,TIAN_TNF_SIGNALING_VIA_NFKB, APOPTOSIS and custom gene sets on the basis of the report and Appendix Table A2 by Aue et al17 and Bhutani et al18.Red indicates that gene expression increased compared with that in the control. Blue indicates that gene expression decreased compared with that in thecontrol. KEGG, Kyoto Encyclopedia of Genes and Genomes.

    Clinical Sequencing in a Patient With Sézary Syndrome

    JCO Precision Oncology 545

    Downloaded from ascopubs.org by Tokyo Daigaku Ikagaku Kenkyujo on June 1, 2020 from 157.082.096.062Copyright © 2020 American Society of Clinical Oncology. All rights reserved.

  • B C

    E F

    A

    D

    IHG

    FIG A6. Histologic and immunohistochemical analysis of tumor specimens before and after the lenalidomidetreatment. Hematoxylin and eosin staining and anti-CD8 and anti-Foxp3 antibody staining of tumor cells andlymphocytes (A, D, and G) before the lenalidomide treatment, (B, E, and H) in a tumor showing good response afterthe first course, and (C, F, and I) in a tumor showing a poor response after three courses of lenalidomide. (D and E) Aremarkable number of CD8+ cells are shown, whereas (G, H, and I) few Foxp3+ cells were observed in each tumor.The number of CD8+ cells after the first course of lenalidomide was increased compared with that (E) beforetreatment but was decreased in the poorly responding lesion (F) after the third course of lenalidomide.

    Hijikata et al

    546 © 2020 by American Society of Clinical Oncology

    Downloaded from ascopubs.org by Tokyo Daigaku Ikagaku Kenkyujo on June 1, 2020 from 157.082.096.062Copyright © 2020 American Society of Clinical Oncology. All rights reserved.

  • TCRLck

    Zap

    SykBLNK

    LAT PLC1

    PLC2Btk

    LynBCR

    IL-1R

    TNF-R1TNF

    IL-1

    Antigen

    MHC/antigen

    EDA-A1

    EDA-A2

    LBPLPS(G-)

    MD-2

    CD40L

    RANKL

    LTALTB

    LIGHT

    BAFF

    EDA-A2 XEDAR

    BAFF-R

    LT-R

    TRAF2

    TRAF2TRAF3

    TRAF6cIAF1/2

    +u

    TIRAP

    TRAF6

    TRAF6

    TRAF6

    TRAF2/5TRADD

    CYLD

    TAK1TAB

    Btk

    IB

    (Y42)

    IB

    p50

    p50

    p65

    p50

    IB Degradation

    NEMO

    SUMOylation

    DNA

    Shuttling

    DNA damage

    PARP1

    –1 0 1

    –1 0 1

    ATMNEMO

    NEMOPIDDRIP1

    c-IAP1/2

    Bcl-XL

    TRAF1/2

    p100

    IL8

    IL-1 TNF-

    IB

    MIP-2

    uPA

    Survival

    Survival

    Activation ofnoncanonical pathway

    Survival

    Positive feedback

    Negative feedback

    Inflammation

    Inflammation

    Survival

    Lymphoid tissuehoming

    B-cell development,B-cell survival

    Myelpoiesis andB-cell lymphopoiesis

    Lymphocyte adhesion,T-cell costimulation

    (H/R)

    Bcl-XL(NGF)

    Bcl-XL

    A1/Bf1-1

    (UV)

    MIP-1

    (TNF-)VCAM-1

    A20

    COX2

    (TNF-, IL-1, LPS)

    (Antigen, TNF-)

    c-FLIP

    Bcl-2 gadd45

    A1/Bf1-1

    XIAP

    PIASyUbs9

    (S32, S36)+p

    p65

    p65

    CK2UV, HER2

    Atypical pathways

    Pervanadate,H/R,H2O2, EGF, CNTF, NGF

    +p

    +p

    +p +p

    NF-B

    NF-B

    NF-B

    NF-B

    p100RelB

    p52DNA

    DNA

    DNA

    Degradation

    Degradation

    BLC ELC

    SLC

    SDF-1

    BAFF

    ICAM

    (LT/LIGHT)

    (S/T (PEST))

    IKKIKK

    NEMO

    CARMA

    PKC

    +p+pCa2+

    Ca2+

    DAG

    DAG

    IP3

    IP3 PKC8

    Bcl10ELKS

    MALT1

    Canonical pathway

    RIG-I–like receptorsignaling pathway

    Ubiquitin-mediatedproteolysis

    Proteasome

    BCRsignaling pathway

    TCRsignaling pathway

    Genotoxic agent

    Calcium signalingpathway

    cIAP1/2

    Apoptosis

    Auto-ubiquitination

    Auto-ubiquitination

    Toll-like receptorsignaling pathway

    Osteoclastdifferentiation

    Cytokine-cytokinereceptor interaction

    Data on KEGG graphrendered by Pathview

    +u

    -u

    +u

    +p

    +u

    +u

    +p

    +p

    RIP1

    TRAF3

    RIG-I

    TRAF6

    TRIM25

    TRAF2/6

    IRAK1/4

    IRAK1/4

    TRAF6TRAM

    MyD88

    MyD88

    TRIFRIP1

    TRAF6TRAF2

    NIK

    NIK

    cIAP1/2

    (S866, S870)

    Partialdegradation

    IKK

    Noncanonical pathway

    TRAF5

    TRAF2

    TRAF2TRAF3

    TRAF3TRAF6

    TRAF3

    RANK

    TLR4

    CD14

    CD40

    Virus

    XEDAR

    EDAR EDARADD

    TCRLck

    Zap

    SykBLNK

    LAT PLC1

    PLC2BtkLyn

    BCR

    IL-1R

    TNF-R1TNF

    IL-1

    Antigen

    MHC/antigen

    EDA-A1

    EDA-A2

    LBPLPS(G-)

    MD-2

    CD40L

    RANKL

    LTALTB

    LIGHT

    BAFF

    EDA-A2 XEDAR

    BAFF-R

    LT-R

    TRAF2

    TRAF2TRAF3

    TRAF6cIAP1/2

    +u

    TIRAP

    TRAF6

    TRAF6

    TRAF6

    TRAF2/5TRADD

    CYLD

    TAK1TAB

    Btk

    IBa

    (Y42)

    IkB

    p50

    p50

    p65

    p50

    IB Degradation

    NEMO

    SUMOylation

    DNA

    Shuttling

    DNA damage

    PARP1ATM

    NEMO

    NEMOPIDDRIP1

    c-IAP1/2

    Bcl-XL

    TRAF1/2

    p100

    IL8

    IL-1 TNF-

    IB

    MIP-2

    uPASurvival

    Survival

    Activation ofnoncanonical pathway

    Survival

    Positive feedback

    Negative feedback

    Inflammation

    Inflammation

    Survival

    Lymphoid tissuehoming

    B-cell development,B-cell survival

    Myelpoiesis andB-cell lymphopoiesis

    Lymphocyte adhesion,T-cell costimulation

    (H/R)

    Bcl-XL(NGF)

    Bcl-XL

    A1/Bf1-1

    (UV)

    MIP-1

    (TNF-)VCAM-1

    A20

    COX2

    (TNF-, IL-1, LPS)

    (Antigen, TNF-)

    c-FLIP

    Bcl-2 gadd45

    A1/Bf1-1

    XIAP

    PIASyUbs9

    (S32, S36)+p

    p65

    p65

    CK2UV, HER2

    Atypical pathways

    Pervanadate,H/R,H2O2, EGF, CNTF, NGF

    +p

    +p

    +p +p

    NF-B

    NF-B

    NF-B

    p100RelB

    p52 DNA

    DNA

    DNA

    Degradation

    Degradation

    BLC ELC

    SLC

    SDF-1

    BAFF

    ICAM

    (LT/LIGHT)

    (S/T (PEST))

    IKKIKKNEMO

    CARMA

    PKC

    +p+pCa2+

    Ca2+

    DAG

    DAG

    IP3

    IP3 PKC8

    Bcl10ELKS

    MALT1

    Canonical pathway

    RIG-I–like receptorsignaling pathway

    Ubiquitin-mediatedproteolysis

    Proteasome

    BCRsignaling pathway

    TCRsignaling pathway

    Genotoxic agent

    Calcium signalingpathway

    cIAP1/2

    Apoptosis

    Auto-ubiquitination

    Auto-ubiquitination

    Toll-like receptorsignaling pathway

    Osteoclastdifferentiation

    Cytokine-cytokinereceptor interaction

    Data on KEGG graphrendered by Pathview

    -u

    +u

    +u

    +p

    +u

    +u

    +p

    +p

    RIP1

    TRAF3

    RIG-I

    TRAF6

    TRIM25

    TRAF2/6

    IRAK1/4

    IRAK1/4

    TRAF6TRAM

    MyD88

    MyD88

    TRIFRIP1

    TRAF6TRAF2

    NIK

    NIK

    cIAP1/2

    (S866, S870)

    Partialdegradation

    IKK

    Noncanonical pathway

    TRAF5

    TRAF2

    TRAF2TRAF3

    TRAF3TRAF6

    TRAF3

    RANK

    TLR4

    CD14

    CD40

    Virus

    XEDAR

    EDAR EDARADD

    NF-B Signaling Pathway

    NF-B Signaling Pathway

    A

    B

    FIG A7. Lenalidomide target pathway: nuclear factor-κB (NF-κB) signaling pathway. (A) Cutaneous T-cell lym-phoma. (B) Adult T-cell leukemia/lymphoma. Green gradient colors indicate downregulation of the gene, and redcolors indicate upregulation of the gene relative to the control sample. BCR, B-cell receptor; H/R, hypoxia/re-oxygenation; IL, interleukin; KEGG, Kyoto Encyclopedia of Genes and Genomes; LPS, lipopolysaccharide; MHC,major histocompatibility complex; TCR, T-cell receptor; TNF-α, tumor necrosis factor-α; UV, ultraviolet.

    Clinical Sequencing in a Patient With Sézary Syndrome

    JCO Precision Oncology 547

    Downloaded from ascopubs.org by Tokyo Daigaku Ikagaku Kenkyujo on June 1, 2020 from 157.082.096.062Copyright © 2020 American Society of Clinical Oncology. All rights reserved.

  • HumanTarget cell

    (MHCclass I)

    Normal cell

    Stressed ortumor cell

    Apoptosis

    Infectedcell

    ?

    DNA

    CASP3 Perforin

    TRAILR

    CD48

    IFNyR

    FAS FASL

    FASL

    Cytokine-cytokinereceptor interaction

    JAK-STATsignaling pathway

    Cytotoxic granulesmovement and exocytosis

    Perforin

    Granzyme

    TRAIL

    IFNsR

    2B4

    NKG2D DAP-10

    SAP

    EAT-2

    LAT

    +p

    +p

    +p

    +p

    +p

    +p

    Ca2+

    DAG

    NK-cell activation

    IP3 -p

    -p

    -p

    +p

    +p

    +p

    Fyn

    Syk

    ZAP70

    Lck

    DAP-12

    ITGAL

    CD94NKG2A/B

    KIR2DL

    KIR3DL2

    KIR3DL1HLA-B

    HLA-A3

    HLA-G1

    HLA-B46

    HLA-C

    HLA-E

    ICAM1/2

    MICA

    MICB

    ULPP1-3

    HA

    ?

    ?

    ?

    SHP-2

    SHP-1

    Activating receptors

    ITGB2

    KIR2DS

    NKG2C/E

    NKp44

    NKp46

    FcR1

    CD3FcRIII

    Antigen

    AntibodyIgG

    NKp30

    CD94 SLP-76

    LATPLC

    PI3K

    Shc Grb2

    PLC

    Fyn

    Sos Ras Raf

    ERK1/2

    MULT1

    Rae-1

    H-60

    ?

    m157Antigen

    IgGAntibody

    NKG2DL

    Cytokine release

    TNF-

    GM-CSF

    IFN-DNA

    DNA

    NFATCaN

    PKC

    Calcium signalingpathway

    NKR-P1C

    NKp46

    FcRIII

    Ly49H

    CD94NKG2C

    Ly49D

    ITGAL

    NKG2A/BCD94

    Activating receptors

    NK cellInhibitory receptors

    Ly49I

    Ly49G2

    Ly49C

    Ly49AH-2Db

    (MHCclass I)

    Target cell

    H-2Dd

    H-2Dk

    H-2Kb

    H-2Kd

    Qa-1

    ICAM1/2

    Inhibitory receptors

    ITGB2

    (MCMV)

    ?

    MEK1/2MAPK signalingpathway

    PAK1

    Vav

    Pyk-2

    Rac

    3BP2

    IFNs

    Bid

    Granzyme

    Data on KEGG graphrendered by Pathview

    HumanTarget cell

    (MHCclass I)

    Normal cell

    Stressed ortumor cell

    Apoptosis

    Infectedcell

    ?

    DNA

    CASP3 Perforin

    Perforin

    TRAILR

    CD48

    IFNR

    FAS FASL

    FASL

    Cytokine-cytokinereceptor interaction

    JAK-STATsignaling pathway

    Cytotoxic granulesmovement and exocytosis

    Perforin

    Granzyme

    TRAIL

    IFNsR

    2B4

    NKG2D DAP-10

    SAP

    EAT-2

    LAT

    +p

    +p

    +p

    +p

    +p

    +p

    Ca2+

    DAG

    NK-cell activation

    IP3 -p

    -p

    -p

    +p

    +p

    +p

    Fyn

    Syk

    ZAP70

    Lck

    DAP-12

    ITGAL

    CD94NKG2A/B

    KIR2DL

    KIR3DL2

    KIR3DL1HLA-B

    HLA-A3

    HLA-G1

    HLA-B46

    HLA-C

    HLA-E

    ICAM1/2

    MICA

    MICB

    ULPP1-3

    HA

    ?

    ?

    ?

    SHP-2

    SHP-1

    Activating receptors

    ITGB2

    KIR2DS

    NKG2C/E

    NKp44

    NKp46

    FcR1

    CD3FcRIII

    Antigen

    Antibody

    IgG

    NKp30

    CD94 SLP-76

    LATPLC

    PI3K

    Shc Grb2

    PLC

    Fyn

    Sos Ras Raf

    ERK1/2

    MULT1

    Rae-1

    H-60

    ?

    m157Antigen

    IgGAntibody

    NKG2DL

    Cytokine release

    TNF-

    GM-CSF

    IFN-

    TRAIL

    TRAIL

    DNA

    DNA

    NFATCaN

    PKC

    Calcium signalingpathway

    NKR-P1C

    NKp46

    FcRIII

    Ly49H

    CD94NKG2C

    Ly49D

    ITGAL

    NKG2A/BCD94

    Activating receptors

    NK cellInhibitory receptors

    Ly49I

    Ly49G2

    Ly49C

    Ly49AH-2Db

    (MHCclass I)

    Target cell

    H-2Dd

    H-2Dk

    H-2Kb

    H-2Kd

    Qa-1

    Inhibitory receptors

    ITGB2

    (MCMV)

    ?

    MEK1/2MAPK signalingpathway

    PAK1

    Vav

    Pyk-2

    Rac

    3BP2

    IFNs

    Bid

    Granzyme

    Data on KEGG graphrendered by Pathview

    ICAM1/2

    PI(3,4,5)P3

    Perforin

    PI(3,4,5)P3

    NK-Cell–Mediated Cytotoxicity

    NK-Cell–Mediated Cytotoxicity

    –1 0 1

    –1 0 1

    A

    B

    FIG A8. Lenalidomide target pathway: natural killer (NK)-cell–mediated cytotoxicity. (A) Cutaneous T-cell lymphoma. (B)Adult T-cell leukemia/lymphoma. Green gradient colors indicate downregulation of the gene, and red colors indicateupregulation of the gene relative to the control sample. JAK-STAT, Janus kinase–signal transducers and activators oftranscription; KEGG, Kyoto Encyclopedia of genes and genomes; MAPK, mitogen-activated protein kinase; MCMV, murinecytomegalovirus; MHC, major histocompatibility complex.

    Hijikata et al

    548 © 2020 by American Society of Clinical Oncology

    Downloaded from ascopubs.org by Tokyo Daigaku Ikagaku Kenkyujo on June 1, 2020 from 157.082.096.062Copyright © 2020 American Society of Clinical Oncology. All rights reserved.

  • CC subfamily

    Chemokines

    Cytokine-Cytokine Receptor Interaction

    CXC subfamily

    The class I helical cytokines

    -Chain utilizing IL6/12-like Interferon family

    IL1-like cytokines

    IL17-like cytokinesThe class

    II helical cytokinesIL10/28-like

    Human/murine

    Human

    Prolactin family

    IL4-like

    C subfamily

    CX3C subfamily

    Nonclassified

    NO signaltransduction

    TNF family TGF- family

    CCL1

    CCL25

    CCL19

    CCL21

    CCL3L1

    CCL4CCL4L1

    CCL4L2

    CCL17

    CCL22

    CCL5

    CCL3

    CCL8

    CCL9

    CCL14

    CCL16

    CCL23

    CCL15

    CCL13

    CCL6

    CCL7

    CCL2

    CCL12

    CCL11

    CCL24

    CCL27

    CCL28

    CCL18

    CCL20

    Data on KEGG graphrendered by Pathview

    Data on KEGG graphrendered by Pathview

    CCR6

    CCR10

    CCR3

    CCR2

    CCR11

    CCR3

    CCR1

    CCR2

    CCR11

    CCR3

    CCR5

    CCR4

    CCR7

    CCR11

    CCR9

    CCR8 CXCL1

    CXCL5

    CXCL6

    CXCL8

    CXCL2

    CXCL3

    CXCL7

    CXCL4

    CXCL9

    CXCL10

    CXCL11

    CXCL13

    CXCL12

    CXCL16

    CXCL14

    CXCR6

    CXCR7

    CXCR4

    CXCR5

    CXCR3

    CXCR2

    CXCR1

    IL2RA

    IL2

    IL4

    IL7

    IL9

    IL15

    IL21

    IL3

    IL5

    IL4

    IL13

    EPO

    GH1

    GH2

    PRL

    TPO

    CSF3

    LEPIL29

    IL28BIL28A

    IL26

    IL22

    IL24

    IL20

    IL19

    IL10

    OSM

    OSM

    LIF

    CTF1

    CNTF

    CLCF1

    IL31

    IL27A

    IL23A

    IL12

    IL11

    IL6IL6R

    IL6ST

    IL11RA

    IL12RB1

    IL23RIL12RB1

    IL27RA

    IL31RA

    IL6ST

    OSMR

    CNTFRLIFR

    IL6ST

    ?LIFR

    IL6ST

    LIFRIL6ST

    OSMRIL6ST

    IL12RB2

    IL6ST

    IL10RAIL10RB

    IL20RAIL20RB

    IL22RA1IL20RB

    IL22RA1IL10RB

    IL20RAIL10RB

    IL28RAIL10RB

    CSF1

    IL34

    IL32

    IL16

    IL17E

    IL17D

    IL17C

    IL17B

    IL17F

    IL17A

    IL18

    IL1F7

    IL1F9

    IL1F10

    IL1F8

    IL1F6

    IL1F5

    IL1RN

    IL1R1

    IL1R2

    IL1RL2IL1RAP

    IL18RAP

    IL1RAP

    IL17RC

    IL17RB

    IL17RE

    IL17RAIL17RB

    CD4

    CSF1R

    ?

    ?

    IL17RA

    ST2

    IL18R1

    IL1RAPIL1B

    IL1A

    IFNG

    IFNT1

    IFNK

    IFNW1

    IFNB1

    IFNATNF

    LTA

    LTB

    LIGHT

    FASLG

    VEGI

    TRAIL

    ?

    EDA-A1

    EDA-A2EDA

    EDA

    NGF

    RANKL

    TWEAK

    CD70

    CD30L

    CD40LG

    41BBL

    OX-40L

    GITRL

    APRIL

    BAFF

    ?

    ?

    DCR3

    FAS

    HVEM

    LTBR

    TNFR2

    TNFR1TGFB1

    TGFB2

    TGFB3

    GDF15

    GDF2

    BMP10

    BMP3

    GDF10

    GDF11

    MSTN

    INHBA

    INHBBGDF1

    GDF3

    NODAL

    GDF9

    INHBE

    AMH

    AMHR2

    AMHR2ACVR1

    ACVR1B

    ACVR2B

    ACVR2B

    ACVR2A

    ACVR2B

    ACVR2A

    ACVR2B

    ACVR2A

    ACVRL1BMPR2

    ACVRL1

    ACVRL1

    TGFBR2

    BMP2

    BMP4

    GDF5

    GDF6

    GDF7

    BMP15

    BMP5

    BMP6

    BMP7

    BMP8

    TGFBR2

    TGFBR2TGFBR1

    ACVR2A

    ?

    TGFBR1

    TGFBR1

    ACVR1B

    ACVR1B

    ACVR1C

    ACVR1CACVR2A

    BMPR2

    BMPR2BMPR1B

    BMPR1B

    BMPR1B

    BMPR1A

    BMPR1A

    BMPR1A

    BMPR2

    BMPR2BMPR1B

    BMPR1B

    BMPR1B

    BMPR1A

    BMPR1A

    BMPR1A

    BMPR2

    BMPR2

    ACVR2B

    ACVR2A

    ACVR2B

    ACVR2A

    ACVR1

    ACVR1

    ACVR1

    ACVR2B

    ACVR2A

    ACVR2B

    ACVR2A

    ACVR2A

    ACVR2B

    ?

    ?

    BMPR1A

    INHBC

    INHA

    DR3

    DR4

    DR5

    DCR1

    DCR2

    DR6

    EDAR

    XEDAR

    NGFR

    RANK

    FN14

    CD27

    CD30

    CD40

    41BB

    Ox40

    GITR

    BCMA

    TACI

    BAFFR

    TROY

    RELT

    OPG

    IFNAR1IFNAR2

    IFNGR1IFNGR2

    IL33

    TSLP

    CSF2

    IL2RBIL2RG

    IL2RBIL2RG

    IL4R

    IL7R

    IL9R

    IL15RA

    IL2RG

    IL2RG

    IL2RG

    IL2RB

    IL21RIL2RG

    IL7RTSLPR

    CSF2RBIL3RA

    CSF2RB

    CSF2RB

    IL4R

    IL4R

    IL4R

    IL13RA1

    IL13RA2

    IL13RA1

    IL13RA2

    EPOR

    GHR

    PRLR

    MPL

    CSF3R

    LEPR

    CSF2RA

    IL5RA

    IL2RG

    CXCL15

    CXCL17

    XCL1XCR1

    ?

    ?

    ?

    CX3CR1

    XCL2

    CX3CL1

    CXCL4L1

    ?

    CCL26

    CC subfamilyChemokines

    Cytokine-Cytokine Receptor Interaction

    CXC subfamilyThe class I helical cytokines

    -Chain utilizing IL6/12-like Interferon family

    IL1-like cytokines

    IL17-like cytokines

    The class II helical cytokinesIL10/28-like

    Human/murine

    Human

    Prolactin family

    IL4-like

    C subfamily

    CX3C subfamily

    Nonclassified

    NO signaltransduction

    TNF family TGF- family–1 0 1

    CCL1

    CCL25

    CCL19

    CCL21

    CCL3L1

    CCL4CCL4L1

    CCL4L2

    CCL17

    CCL22

    CCL5

    CCL3

    CCL8

    CCL9

    CCL14

    CCL16

    CCL123

    CCL15

    CCL13

    CCL6

    CCL7

    CCL2

    CCL12

    CCL11

    CCL24

    CCL27

    CCL28

    CCL18

    CCL20 CCR6

    CCR10

    CCR3

    CCR2

    CCR11

    CCR3

    CCR1

    CCR2

    CCR11

    CCR3

    CCR5

    CCR4

    CCR7

    CCR11

    CCR9

    CCR8 CXCL1

    CXCL5CXCL6

    CXCL8

    CXCL2

    CXCL3

    CXCL7

    CXCL4

    CXCL9

    CXCL10

    CXCL11

    CXCL13

    CXCL12

    CXCL16

    CXCL14

    CXCR6

    CXCR7

    CXCR4

    CXCR5

    CXCR3

    CXCR2

    CXCR1

    IL2RA

    IL2

    IL4

    IL7

    IL9

    IL15

    IL21

    IL3

    IL5

    IL4

    IL13

    EPO

    GH1

    GH2

    PRL

    TPO

    CSF3

    LEPIL29

    IL28BIL28A

    IL26

    IL22

    IL24

    IL20

    IL19

    IL10

    OSM

    OSM

    LIF

    CTF1

    CNTF

    CLCF1

    IL31

    IL27A

    IL23A

    IL12

    IL11

    IL6 IL6RIL6ST

    IL11RA

    IL12RB1

    IL23RIL12RB1

    IL27RA

    IL31RA

    IL6ST

    OSMR

    CNTFRLIFR

    IL6ST

    ?LIFR

    IL6ST

    LIFRIL6ST

    OSMRIL6ST

    IL12RB2

    IL6ST

    IL10RAIL10RB

    IL20RAIL20RB

    IL22RA1IL20RB

    IL22RA1IL10RB

    IL20RAIL10RB

    IL28RAIL10RB

    CSF1

    IL34

    IL32

    IL16

    IL17E

    IL17D

    IL17C

    IL17B

    IL17F

    IL17A

    IL18

    IL1F7

    IL1F9IL1F10

    IL1F8

    IL1F6

    IL1F5

    IL1RN

    IL1R1

    IL1R2

    IL1RL2IL1RAP

    IL18RAP

    IL1RAP

    IL17RC

    IL17RB

    IL17RE

    IL17RAIL17RB

    CD4

    CSF1R

    ?

    ?

    IL17RA

    ST2

    IL18R1

    IL1RAPIL1B

    IL1A

    IFNG

    IFNT1

    IFNK

    IFNW1

    IFNB1

    IFNATNF

    LTA

    LTB

    LIGHT

    FASLG

    VEGI

    TRAIL

    ?

    EDA-A1

    EDA-A2EDA

    EDA

    NGF

    RANKL

    TWEAK

    CD70

    CD30L

    CD40LG

    41BBL

    OX-40L

    GITRL

    APRIL

    BAFF

    ?

    ?

    DCR3

    FAS

    HVEM

    LTBR

    TNFR2

    TNFR1TGFB1

    TGFB2

    TGFB3

    GDF15

    GDF2

    BMP10

    BMP3

    GDF10

    GDF11

    MSTN

    INHBA

    INHBBGDF1

    GDF3

    NODAL

    GDF9

    INHBE

    AMH

    AMHR2

    AMHR2ACVR1

    ACVR1B

    ACVR2B

    ACVR2B

    ACVR2A

    ACVR2B

    ACVR2A

    ACVR2B

    ACVR2A

    ACVRL1BMPR2

    ACVRL1

    ACVRL1

    TGFBR2

    BMP2

    BMP4

    GDF5

    GDF6

    GDF7

    BMP15

    BMP5

    BMP6

    BMP7

    BMP8

    TGFBR2

    TGFBR2TGFBR1

    ACVR2A

    ?

    TGFBR1

    TGFBR1

    ACVR1B

    ACVR1B

    ACVR1C

    ACVR1CACVR2A

    BMPR2

    BMPR2BMPR1B

    BMPR1B

    BMPR1B

    BMPR1A

    BMPR1A

    BMPR1A

    BMPR2

    BMPR2BMPR1B

    BMPR1B

    BMPR1B

    BMPR1A

    BMPR1A

    BMPR1A

    BMPR2

    BMPR2

    ACVR2B

    ACVR2A

    ACVR2A

    ACVR2A

    ACVR1

    ACVR1

    ACVR1

    ACVR2B

    ACVR2A

    ACVR2B

    ACVR2A

    ACVR2A

    ACVR2B

    ?

    ?

    BMPR1A

    INHBC

    INHA

    DR3

    DR4

    DR5

    DCR1

    DCR2

    DR6

    EDAR

    XEDAR

    NGFR

    RANK

    FN14

    CD27

    CD30

    CD40

    41BB

    Ox40

    GITR

    BCMA

    TACI

    BAFFR

    TROY

    RELT

    OPG

    IFNAR1IFNAR2

    IFNGR1IFNGR2

    IL33

    TSLP

    CSF2

    IL2RBIL2RG

    IL2RBIL2RG

    IL4R

    IL7R

    IL9R

    IL15RA

    IL2RG

    IL2RG

    IL2RG

    IL2RB

    IL21RIL2RG

    IL7RTSLPR

    CSF2RBIL3RA

    CSF2RB

    CSF2RB

    IL4R

    IL4R

    IL4R

    IL13RA1

    IL13RA2

    IL13RA1

    IL13RA2

    EPOR

    GHR

    PRLR

    MPL

    CSF3R

    LEPR

    CSF2RA

    IL5RA

    IL2RG

    CXCL15

    CXCL17

    XCL1XCR1

    ?

    ?

    ?

    CX3CR1

    XCL2

    CX3CL1

    CXCL4L1

    ?

    CCL26

    –1 0 1

    A

    B

    FIG A9. Lenalidomide target pathway: cytokine-cytokine receptor interaction. (A) Cutaneous T-cell lymphoma. (B) Adult T-cellleukemia/lymphoma. Green gradient colors indicate downregulation of the gene, and red colors indicate upregulation of thegene relative to the control sample. IFN, interferon; IL, interleukin; KEGG, Kyoto Encyclopedia of genes and genomes; TGF-β,transforming growth factor-β; TNF, tumor necrosis factor.

    Clinical Sequencing in a Patient With Sézary Syndrome

    JCO Precision Oncology 549

    Downloaded from ascopubs.org by Tokyo Daigaku Ikagaku Kenkyujo on June 1, 2020 from 157.082.096.062Copyright © 2020 American Society of Clinical Oncology. All rights reserved.

  • TNF Signaling Pathway

    Data on KEGG graphrendered by Pathview

    cIAP1/2

    ASK1

    TAB1/2/3TAK1

    NIKNF-B

    Tpl2

    IKK

    IKK NF-B

    PGAM5LMLKL

    PI3K

    TRAF1

    TNF/LTA

    TNF

    TRAF2TRAF3

    NIK

    cIAP1/2

    AIP1

    RIP

    JNK

    MKK3

    IKKs

    NF-B

    IB

    p38

    c-Jun

    IRF1

    Apoptosis

    DNAIFN

    Cell survival

    Degradation

    Necroptosis

    DNA

    DNA

    Akt+p

    +p

    +p

    +p

    -p

    +p

    +p

    +p

    +p

    +p

    +p

    +p

    +p

    +u

    +p +p

    +p

    +p

    RIP3RIP1

    Deubiquitination

    Necrosome

    (Limited expression)

    PGAM5SDrp1

    NF-Bsignaling pathway

    IB ubiquitinationand degradation

    Ubiquitin-mediatedproteolysis

    PI3K-Aktsignaling pathway

    MEK1

    MKK3/6

    MSK1/2

    AP-1

    c/EBP

    CREB

    ERK1/2

    p38

    JNK1/2

    ITCH c-FLIP

    CASP3Apoptosis

    Apoptosis

    –1 0 1

    –1 0 1

    Ccl20Ccl5Ccl2

    Cxcl1

    Cxcl5

    Csf1

    Fas

    IL1b

    Bcl3

    Fos

    Mmp14Mmp9Mmp3

    Vegfc

    Nod2

    Icam1 Sele Vcam1

    Edn1

    PRRs

    Cell adhesion

    Ptgs2

    Synthesis of inflammatory mediators

    Vascular effects

    Jun JunB

    Ifi47

    Transcription factors

    Remodeling of extracellular matrix

    Intracellular signaling (negative)

    Intracellular signaling (positive)

    Nfkbia Socs3

    IL6 IL15 Lif Tnf

    Traf1Tnfaip3

    Csf2

    IL18R1

    Leukocyte activation

    Surface receptors

    Inflammatory cytokines

    Cxcl2

    Cxcl10 Cx3cl1

    Cxcl3

    Leukocyte recruitment

    CASP7CASP10

    CASP8

    FADD

    MKK4/7

    MAPKsignaling pathway

    cIAP1/2 degradation

    IBIKK

    TRAF2/5

    RIP1TRADD

    SODD

    Jag1

    TNF Signaling Pathway

    Data on KEGG graphrendered by Pathview

    cIAP1/2

    ASK1

    TAB1/2/3TAK1

    NIKNF-B

    Tpl2

    IKK

    IKK NF-B

    PGAM5LDrp1

    PI3K

    TRAF1

    TNF/LTA

    TNF

    TRAF2

    TRAF3

    NIK

    cIAP1/2

    AIP1

    RIP

    JNK

    MKK3

    IKKs

    NF-B

    IB

    p38

    c-Jun

    IRF1

    Apoptosis

    DNAIFN

    Cell survival

    Degradation

    Necroptosis

    DNA

    DNA

    Akt+p

    +p

    +p

    +p

    -p

    +p

    +p

    +p

    +p

    +p

    +p

    +p

    +p

    +u

    +p +p

    +p

    +p

    RIP3 MLKLRIP1

    Deubiquitination

    Necrosome

    (Limited expression)

    PGAM5S

    NF-Bsignaling pathway

    IB ubiquitinationand degradation

    Ubiquitin-mediatedproteolysis

    PI3K-Aktsignaling pathway

    MEK1

    MKK3/6

    MSK1/2

    AP-1

    c/EBP

    CREB

    ERK1/2

    p38

    JNK1/2

    ITCH c-FLIP

    CASP3Apoptosis

    ApoptosisCcl20Ccl5Ccl2

    Cxcl1

    Cxcl5

    Csf1

    Fas

    IL1b

    Bcl3

    Fos

    Mmp14Mmp9Mmp3

    Vegfc

    Nod2

    Icam1 Sele Vcam1

    Edn1

    PRRs

    Cell adhesion

    Ptgs2

    Synthesis of inflammatory mediators

    Vascular effects

    Jun JunB

    Ifi47

    Transcription factors

    Remodeling of extracellular matrix

    Intracellular signaling (negative)

    Intracellular signaling (positive)

    Nfkbia Socs3

    IL6 IL15 Lif Tnf

    Traf1Tnfaip3

    Csf2

    IL18R1

    Leukocyte activation

    Surface receptors

    Inflammatory cytokines

    Cxcl2

    Cxcl10 Cx3cl1

    Cxcl3

    Leukocyte recruitment

    CASP7CASP10

    CASP8

    FADD

    MKK4/7

    MAPKsignaling pathway

    cIAP1/2 degradation

    IBIKK

    TRAF2/5

    RIP1TRADD

    SODD

    Jag1

    TNFR1

    TNFR2

    TNFR1

    TNFR2

    A

    B

    FIG A10. Lenalidomide target pathway: tumor necrosis factor (TNF) signaling pathway. (A) Cutaneous T-cell lymphoma. (B)Adult T-cell leukemia/lymphoma. Green gradient colors indicate downregulation of the gene, and red colors indicateupregulation of the gene relative to the control sample. KEGG, Kyoto Encyclopedia of genes and genomes; MAPK, mitogen-activated protein kinase; NF-κB, nuclear factor-κB; PRR, pattern recognition receptor.

    Hijikata et al

    550 © 2020 by American Society of Clinical Oncology

    Downloaded from ascopubs.org by Tokyo Daigaku Ikagaku Kenkyujo on June 1, 2020 from 157.082.096.062Copyright © 2020 American Society of Clinical Oncology. All rights reserved.

  • B

    7 Nov 2017 12 Jan 2018 7 Feb 2018

    Perc

    ent

    CD3–CD16+CD56+/lym

    CD3+CD8+

    CD3+CD4+CD45RA–Foxp3+high

    0

    10

    20

    30

    40

    50

    60

    70

    16 Oct 2017

    A

    SSC-W, FSC-W subset102 103 104 105

    CD235a/PI

    0

    0

    102 103 104 1050 102 103 104 1050

    102 103 104 1050 102 103 104 1050

    50K

    100K

    150K

    200K

    250KFS

    C56.6

    PI–CD235a–250K

    SSC

    0

    0

    105

    104

    103

    102

    0

    105

    104

    103

    102

    0

    105

    104

    103

    102

    0

    105

    104

    103

    102

    0

    105

    104

    103

    102

    CD14

    73.3

    CD3–CD19– cells CD16

    CD56

    90.6

    CD14– cells CD3

    CD19

    44.3

    50.9

    4.75

    CD3+ T cells CD8

    CD4

    30.4

    63

    CD4+ T cells (Foxp3)

    CD45

    RA0.746 0.0403

    6.0780.4 12.7

    Doublet free

    CD3+ T cells

    CD235a–PI– cells CD14– cells CD3–CD19– cells

    CD4+ T cells

    50K 100K 150K 200K

    FIG A11. Immunophenotypes of peripheral blood mononuclear cells (PBMNCs) from the patient during lenalidomide treatment. PBMNCs were stainedfor CD3, CD4, CD8, CD14, CD16, CD19, CD45RA, CD235a/PI, and Foxp3. (A) Lymphocytes (lym) were identified based on light scatter characteristics(low forward and low side scatter) and negative for CD14 and CD235/PI. Natural killer (NK) cells were defined as CD3−CD16+CD56+ after gating for CD19−

    lymphocytes. Effector regulatory T (Treg) cells were defined as CD4+CD45RA−Foxp3+high. (B) The frequencies of NK cells and ratio of CD3+CD8+ T cellsincreased after lenalidomide treatment, which was not accompanied by an increase in effector Treg cells. FSC-W, forward scatter pulse width; SSC-W,side scatter pulse width.

    Clinical Sequencing in a Patient With Sézary Syndrome

    JCO Precision Oncology 551

    Downloaded from ascopubs.org by Tokyo Daigaku Ikagaku Kenkyujo on June 1, 2020 from 157.082.096.062Copyright © 2020 American Society of Clinical Oncology. All rights reserved.

  • TABLEA1

    .Cod

    ingVa

    riantsIden

    tified

    From

    Who

    le-ExomeSe

    quen

    cing

    Tier

    Grad

    ing

    (driv

    er)

    Chrom

    Positio

    nRe

    fALT

    Depth

    (tumor)

    Depth

    (normal)

    VAF

    −Log

    10

    Pa

    Gene

    AminoAc

    idCh

    ange

    COSM

    IC70

    IDSIFT

    PolyPh

    en-2

    MutationTaster

    117

    7578

    290

    CT

    9014

    80.56

    725

    .1TP

    53splicing

    COSM

    O41

    3986

    9—

    —D

    2(VUS)

    1740

    3121

    26TC

    TGGCAC

    T14

    813

    40.07

    43.0

    KCNH4

    p.P99

    3fs

    ——

    ——

    2(VUS)

    1753

    0074

    53G

    T89

    630.07

    91.4

    TOM1L

    1p.R24

    7L—

    DD

    D

    2(VUS)

    418

    7539

    038

    GT

    164

    133

    0.10

    44.4

    FAT1

    p.T2

    901K

    —T

    PD

    2(VUS)

    1870

    8042

    1C

    T12

    398

    0.12

    23.8

    LAMA1

    p.A33

    T—

    TD

    D

    2(VUS)

    312

    6194

    576

    CG

    4352

    0.14

    02.2

    ZXDC

    p.G45

    R—

    DB

    N

    2(VUS)

    1772

    8690

    18G

    A43

    530.14

    02.2

    FDXR

    p.R18

    W—

    DD

    N

    2(VUS)

    1832

    1506

    5C

    T57

    520.14

    02.2

    MYO

    M1

    p.A53

    T—

    TD

    D

    2(VUS)

    119

    9519

    97C

    G49

    840.14

    33.2

    MINOS1

    p.Q11

    2E—

    ——

    2(VUS)

    296

    8011

    04C

    A97

    790.14

    43.5

    ASTL

    p.D77

    Y—

    DD

    N

    2(VUS)

    1328

    6113

    93G

    T12

    511

    30.14

    45.2

    FLT3

    p.P41

    3Q—

    TD

    D

    2(VUS)

    216

    6854

    673

    GA

    102

    117

    0.14

    74.1

    SCN1A

    p.P14

    51S

    —D

    DD

    2(VUS)

    1395

    3638

    07CGCGGCGGC

    GGCGGCGGCGGCA

    C60

    670.15

    03.1

    SOX2

    1p.15

    9_16

    6del

    ——

    ——

    2(VUS)

    412

    1957

    868

    GA

    9981

    0.15

    23.0

    NDNF

    p.R42

    0X—

    T—

    D

    2(VUS)

    316

    4732

    921

    CA

    149

    163

    0.15

    47.8

    SIp.C13

    30F

    —T

    PN

    2(VUS)

    1779

    6619

    73G

    A17

    316

    10.16

    28.5

    HGS

    p.R33

    2QCOSM

    3388

    305

    TD

    D

    2(VUS)

    913

    1252

    77TT

    T11

    510

    10.16

    55.5

    MPD

    Zp.K15

    82fs

    ——

    ——

    2(VUS)

    1739

    5374

    60G

    T15

    712

    30.16

    66.7

    KRT3

    4p.Q18

    8K—

    DB

    N

    2(VUS)

    336

    8734

    72A

    T12

    113

    40.17

    47.2

    TRAN

    K1p.C24

    90X

    —T

    —A

    2(VUS)

    995

    2678

    18AAATA

    GA

    134

    137

    0.17

    97.8

    ECM2

    p.S4

    86fs

    ——

    ——

    2(VUS)

    X15

    5915

    54GG

    G53

    430.18

    92.7

    ACE2

    p.P49

    2fs

    ——

    ——

    2(VUS)

    119

    9520

    61C

    T63

    740.19

    04.3

    MINOS1

    p.S1

    33F

    ——

    ——

    2(VUS)

    1689

    6574

    21C

    T18

    416

    10.19

    68.3

    CPNE7

    p.H45

    0Y—

    DD

    D

    2(VUS)

    1870

    5260

    84G

    A85

    760.21

    24.3

    NET

    O1

    p.S1

    49L

    —T

    DD

    2(VUS)

    353

    5356

    80A

    T61

    550.21

    33.8

    CACN

    A1D

    p.N13

    9I—

    DD

    D

    2(VUS)

    178

    4308

    24C

    A12

    114

    90.21

    57.4

    FUBP1

    p.E1

    89X

    —T

    —A

    2(VUS)

    123

    7802

    393

    GT

    7312

    70.21

    97.5

    RYR

    2p.R23

    36I

    —D

    DD

    2(VUS)

    832

    6568

    5C

    T18

    213

    80.22

    09.1

    CSMD1

    p.E6

    03K

    —T

    DD

    2(VUS)

    1193

    5539

    92CACAGCAGCG

    C93

    960.22

    67.0

    VSTM

    5p.15

    6_15

    9del

    ——

    ——

    2(VUS)

    111

    5840

    44C

    T48

    610.22

    94.2

    PTCH

    D2

    p.T8

    03M

    —D

    DD

    2(VUS)

    146

    6509

    54G

    T55

    480.25

    54.2

    TSPA

    N1

    p.V2

    18L

    —T

    DD

    2(VUS)

    1286

    7284

    3AGATA

    A12

    013

    20.25

    811

    .0CL

    EC4D

    p.D13

    6fs

    ——

    ——

    (Con

    tinue

    don

    followingpa

    ge)

    Hijikata et al

    552 © 2020 by American Society of Clinical Oncology

    Downloaded from ascopubs.org by Tokyo Daigaku Ikagaku Kenkyujo on June 1, 2020 from 157.082.096.062Copyright © 2020 American Society of Clinical Oncology. All rights reserved.

  • TABLEA1

    .Cod

    ingVa

    riantsIden

    tified

    From

    Who

    le-ExomeSe

    quen

    cing

    (Con

    tinue

    d)Tier

    Grad

    ing

    (driv

    er)

    Chrom

    Positio

    nRe

    fALT

    Depth

    (tumor)

    Depth

    (normal)

    VAF

    −Log

    10

    Pa

    Gene

    AminoAc

    idCh

    ange

    COSM

    IC70

    IDSIFT

    PolyPh

    en-2

    MutationTaster

    2(VUS)

    2064

    4532

    CG

    2534

    0.28

    02.9

    SCRT2

    p.G23

    6A—

    DD

    D

    2(VUS)

    1915

    8068

    29G

    A15

    113

    60.28

    512

    .0CY

    P4F1

    2p.R40

    0Q—

    DD

    N

    2(VUS)

    1739

    2217

    48G

    T12

    212

    80.28

    712

    .2KR

    TAP2

    -4p.T1

    17N

    —T

    —N

    2(VUS)

    146

    6509

    55T

    A52

    420.28

    84.1

    TSPA

    N1

    p.V2

    18E

    —D

    DD

    2(VUS)

    1720

    1568

    52G

    T51

    740.29

    45.4

    SPEC

    C1p.S8

    78I

    COSM

    1609

    946

    TD

    D

    2(VUS)

    810

    3287

    953

    CT

    155

    113

    0.29

    712

    .4UBR5

    p.G22

    05R

    —D

    DD

    2(VUS)

    1480

    2714

    80C

    A12

    190

    0.29

    89.8

    NRXN

    3p.S9

    44Y

    ——

    DD

    2(VUS)

    2043

    3535

    25A

    G41

    320.31

    73.5

    WISP2

    p.S1

    42G

    —T

    PN

    2(VUS)

    X13

    8633

    26A

    T11

    286

    0.32

    110

    .0F9

    p.K18

    8X—

    T—

    A

    2(VUS)

    729

    6395

    4G

    T10

    988

    0.33

    010

    .5CA

    RD11

    p.S6

    18Y

    —D

    DD

    2(VUS)

    313

    5871

    427

    AG

    9710

    70.34

    012

    .1MSL

    2p.L9

    9P—

    DB

    D

    2(VUS)

    1953

    5181

    31A

    G21

    121

    30.34

    123

    .4ER

    VV-1

    p.N26

    3S—

    ——

    2(VUS)

    218

    6609

    095

    CG

    7010

    10.34

    310

    .5FS

    IP2

    p.T2

    34S

    —D

    —N

    2(VUS)

    2147

    6274

    32C

    A42

    720.35

    76.2

    LSS

    p.E3

    79D

    —D

    DD

    2(VUS)

    542

    7970

    23G

    A53

    630.37

    77.8

    CCDC1

    52p.E1

    75K

    —T

    PD

    2(VUS)

    773

    0119

    83G

    T46

    480.39

    16.5

    MLX

    IPL

    p.L3

    78I

    —T

    DN

    2(VUS)

    1944

    0966

    99G

    A51

    290.39

    24.6

    IRGQ

    p.P45

    1S—

    TD

    N

    2(VUS)

    729

    6395

    5A

    G11

    488

    0.40

    413

    .3CA

    RD11

    p.S6

    18P

    —D

    DD

    2(VUS)

    1548

    0552

    95A

    T71

    116

    0.40

    812

    .8SE

    MA6

    Dp.L2

    47F

    —D

    DD

    2(VUS)

    1946

    9156

    85C

    T15

    615

    60.41

    019

    .6CC

    DC8

    p.R12

    8H—

    DD

    D

    2(VUS)

    1168

    3694

    16C

    CC

    9867

    0.42

    98.0

    PPP6

    R3

    p.A76

    0fs

    ——

    ——

    2(VUS)

    1256

    4818

    67C

    A77

    780.45

    512

    .8ER

    BB3

    p.Y2

    65X

    —T

    —D

    2(VUS)

    1876

    7552

    35G

    A83

    830.45

    813

    .5SA

    LL3

    p.A10

    82T

    COSM

    3388

    512

    TP

    N

    2(VUS)

    739

    0464

    68G

    A91

    800.46

    214

    .0PO

    U6F

    2splicing

    ——

    —N

    2(VUS)

    1232

    1375

    70GTG

    TG

    5147

    0.47

    18.3

    KIAA

    1551

    p.12

    28_

    1228

    del

    ——

    ——

    2(VUS)

    642

    9930

    08G

    A72