Hepatic involvement by T-cell neoplasms: a clinicopathologic study of 40 cases RSS PDFダウンロード Philippa Li MD, Dongwei Zhang MD, PhD, Jiehao Zhou MD, PhD, Peng Li MD, PhD, Yulei Shen MD, PhD, Zenggang Pan MD, PhD, Andrew G. Evans MD, PhD and Xiaoyan Liao MD, PhD Human Pathology, 2020-12-01, Volume 106, Pages 1-12, Copyright c 2020 Elsevier Inc. Open reading mode Summary Hepatic involvement by a T-cell neoplasm is rare and often challenging to diagnose in liver biopsies. We collected 40 cases of T-cell neoplasms diagnosed in the liver from five large academic institutions to assess the clinicopathologic features. The patients included 11 women and 29 men, with a median age of 54 (range: 2?75) years and a high mortality rate (31/37, 83.8%). Fourteen (35%) patients were diagnosed with hepatosplenic T-cell lymphoma (HSTCL), 13 (32.5%) peripheral T-cell lymphoma, not otherwise specified (PTCL-NOS), and 13 (32.5%) other types of T-cell neoplasms. Patients with HSTCL were much younger and had worse survival than PTCL-NOS and other T-cell neoplasms ( P < 0.05). On imaging studies, 20 cases (50%) showed abnormalities, including 10 with mass lesions that correlated with normal or cholestatic pattern enzyme elevation. Histomorphological analysis revealed four main patterns; with the exception of mass forming lesions (pattern 4; n = 8), cases with sinusoidal predominant (pattern 1; n = 12), portal predominant with sinusoidal infiltrates (pattern 2; n = 13) or lobular aggregates (pattern 3; n = 5) demonstrated small to medium lymphocytes resembling a reactive/inflammatory process. In addition, we described two cases of T-cell large granular lymphocytic leukemia that mimicked HSTCL, and a case of aggressive post-transplant lymphoproliferative disorder that developed after chronic Epstein-barr virus (EBV) infection, suggesting the importance of EBV testing in some lymphoma cases. As the largest cohort of T-cell neoplasms in liver, our study provides critical data on disease frequency, distribution, and clinicopathologic features that are essential for accurate diagnosis. 要約 T細胞腫瘍の肝への浸潤はまれであり,肝生検での診断が困難な場合が多い。我々は、5つの大規模学術施設から肝臓内T細胞腫瘍と診断された40例を収集し、臨床病理学的特徴を評価した。患者は女性11名、男性29名で、年齢中央値は54歳(範囲:2-75)、死亡率は高い(31/37、83.8%)。 14名(35%)が肝脾T細胞リンパ腫(HSTCL)、13名(32.5%)が特定不能の末梢T細胞リンパ腫(PTCL-NOS)、13名(32.5%)がその他のタイプのT細胞腫瘍と診断された。HSTCLの患者はPTCL-NOSや他のT細胞腫瘍に比べて非常に若く、生存率も悪かった( P < 0.05)。 画像検査では、20例(50%)に異常が認められ、そのうち10例は正常または胆汁性パターンの酵素上昇と相関する腫瘤性病変を有していた。組織形態学的解析では,腫瘤性病変(パターン4,n=8)を除き,洞様優位(パターン1,n=12),洞様浸潤を伴う門脈優位(パターン2,n=13),葉状集積(パターン3,n=5)例では,反応・炎症過程に似た小〜中程度のリンパ球が確認され,4パターンが主であった. さらに,HSTCLを模倣したT細胞性大顆粒リンパ球性白血病2例と,Epstein-barrウイルス(EBV)慢性感染後に発症した侵攻性移植後リンパ増殖性疾患1例を挙げ,一部のリンパ腫症例におけるEBV検査の重要性を示唆した。 本研究は、肝細胞新生物の最大規模のコホートとして、正確な診断に不可欠な疾患頻度、分布、臨床病理学的特徴に関する重要なデータを提供するものである。 1 Introduction The liver is not commonly involved with non-Hodgkin lymphoma, occurring next in frequency to lymph node, spleen, and bone marrow. Most hepatic lymphomas are secondary involvement by a systemic disease, frequently diffuse large B-cell lymphoma, comprising nearly half of the cases reported [ 1 , 2 ]. Approximately 12% of lymphomas in the liver are attributed to a T-cell lineage, mostly peripheral T-cell lymphoma, not otherwise specified (PTCL-NOS, 9%), followed by anaplastic large cell lymphoma (ALCL, 2%) and hepatosplenic T-cell lymphoma (HSTCL, 1%) [ 1 ]. Owing to their rarity, T-cell lymphomas in the liver were often reported in single case reports or small case series [ 3 , 4 ], lacking epidemiologic and systemic clinicopathologic analysis. The initial lymphoma diagnosis, particularly for T-cell neoplasms, can be very challenging in small liver biopsies [ 3 ]. For example, both HSTCL and T-cell large granular lymphocytic leukemia (T-LGL) typically infiltrate the hepatic sinusoids without discrete mass formation, mimicking a reactive condition. Other lymphomas can have portal and lobular infiltrates, indistinguishable from viral hepatitis or drug-induced liver injury by histology alone. Accurate diagnosis relies on clinical history, immunohistochemistry (IHC) to determine T-cell lineage and antigen aberrancy, polymerase chain reaction analysis on T-cell receptors ( TCRs ) to establish clonality, and sometimes cytogenetic or molecular studies to detect disease-specific alterations in certain lymphomas [ 5 , 6 ]. This poses significant challenges when limited biopsy material is available for evaluation, emphasizing the importance of pattern recognition and selective ancillary tests. Herein, we performed a multi-institutional retrospective review of various types of T-cell neoplasms diagnosed in liver specimens to analyze the essential clinical and histopathologic features. はじめに 肝臓は非ホジキンリンパ腫の発生頻度が低く、リンパ節、脾臓、骨髄に次ぐ頻度である。ほとんどの肝リンパ腫は全身性疾患による二次的な病変であり、びまん性大細胞型B細胞リンパ腫が多く、報告例のほぼ半分を占める [ 1 , 2 ]。肝臓のリンパ腫の約12%はT細胞系譜に起因し、そのほとんどは特定不能の末梢T細胞リンパ腫(PTCL-NOS、9%)、次いで未分化大細胞リンパ腫(ALCL、2%)および肝脾T細胞リンパ腫(HSTCL、1%)である[1]。まれな疾患のため, 肝臓のT細胞リンパ腫はしばしば単一症例報告または小規模ケースシリーズで報告されており[3 , 4]、疫学的および全身的な臨床病理学的解析がおこなわれていない。 リンパ腫の初期診断、特にT細胞腫瘍の診断は、小さな肝生検では非常に困難である[3]。例えば、HSTCLとT細胞性大顆粒リンパ球性白血病(T-LGL)の両者は、典型的に肝類洞に浸潤し、反応性の状態を模倣して、個別の腫瘤形成はみられない。  その他のリンパ腫では、門脈および小葉に浸潤を認め、組織学のみではウイルス性肝炎または薬剤誘発性肝障害と区別できないことがある。正確な診断は、臨床歴、T細胞系譜と抗原異常を決定する免疫組織化学(IHC)、クローナリティを確立するためのT細胞受容体(TCR)のポリメラーゼ連鎖反応分析、そして時には特定のリンパ腫における疾患特異的変化を検出する細胞遺伝学や分子学に依存する [ 5 , 6 ]。生検材料が限られている場合、これは大きな課題となり、パターン認識と選択的な補助検査の重要性が強調される。 ここでは、肝検体で診断されたさまざまな種類のT細胞腫瘍について、多施設共同によるレトロスペクティブレビューを行い、必須の臨床的・病理組織学的特徴を分析した。 2 Materials and methods 2.1 Patients and data collection A text-based search in the electronic pathology database between 2000 and 2020 was performed collaboratively across five large academic institutions in the United States. Clinical information regarding patients’ age, sex, imaging findings, relevant medical history, clinical presentation before liver biopsy, treatment, follow-up, and disease outcomes was collected. When multiple liver specimens were available for the same patient, the imaging study and liver function test most pertinent to the final diagnosis were recorded. In particular, the image findings were divided into 4 categories: (1) sizable mass lesion (mass); (2) focal nonspecific abnormality without mass formation (abnormal); (3) hepatomegaly (enlargement); (4) no abnormality (normal). The liver function tests, including alanine aminotransferase (ALT), aspartate aminotransferase, alkaline phosphatase (ALP), and total bilirubin levels were recorded for the highest value within one month before the biopsy or autopsy. A ratio (R) value, defined as ALT/upper limit of normal: ALP/upper limit of normal, was applied to designate the pattern of liver injury, namely, hepatitis pattern if R > 5, cholestatic pattern if R < 2, and mixed pattern if R is between 2 and 5 [ 7 ]. This study was approved by the institutional review boards of all participating medical centers. 2 材料と方法 2.1 患者およびデータ収集 2000 年から 2020 年までの電子病理データベースにおけるテキストベースの検索を、米国内の 5 つの大規模学術機関にまたがる共同作業で行った。患者の年齢、性別、画像所見、関連する病歴、肝生検前の臨床像、治療、経過観察、疾患の転帰に関する臨床情報を収集した。同一患者で複数の肝標本が得られた場合、最終診断に最も関連した画像検査と肝機能検査を記録した。特に、画像所見は、(1)大きめの腫瘤性病変(腫瘤)、(2)腫瘤を形成しない局所的非特異的異常(異常)、(3)肝腫大(腫大)、(4)異常なし(正常)の4カテゴリーに分類された。アラニンアミノトランスフェラーゼ(ALT)、アスパラギン酸アミノトランスフェラーゼ、アルカリホスファターゼ(ALP)、総ビリルビン値などの肝機能検査は、生検または剖検前1カ月以内の最高値を記録した。ALT/正常値上限と定義される比率(R)値。ALP/正常値上限と定義される比率(R)値を適用し,肝障害のパターンを指定した.すなわち,R>5なら肝炎パターン,R<2なら胆石症パターン,Rが2〜5なら混合パターンである[7].この研究は、参加したすべての医療機関の施設審査委員会の承認を得ている。 www.DeepL.com/Translator(無料版)で翻訳しました。 2.2 Lymphoma diagnosis, classification, and histomorphologic analysis The pathology report and diagnosis for each case were reviewed by expert hematopathologists at each contributing institution. The classification of T-cell neoplasms was based on the 2017 WHO Classification of Tumors of Hematopoietic and Lymphoid Tissues [ 8 ]. Specific types of T-cell neoplasms diagnosed in this study included HSTCL, peripheral T-cell lymphoma, not otherwise specified (PTCL-NOS), T-LGL, ALK-negative ALCL, cutaneous T-cell lymphoma (CTCL) specifically mycosis fungoides, angioimmunoblastic T-cell lymphoma (AITL), post-transplant lymphoproliferative disorder (PTLD), extranodal NK/T-cell lymphoma, nasal type (NK/T), T-prolymphocytic leukemia (T-PLL), and T-cell acute lymphoblastic leukemia (T-ALL). When applicable, a “primary” hepatic lymphoma is defined as lymphoma either confined to the liver or having initial and major liver involvement before spreading to other anatomic sites. A “secondary” hepatic lymphoma is defined when there is prior or concurrent systemic disease, or the same lymphoma has been diagnosed at other anatomic sites before liver involvement [ 91011 ]. The histomorphology and infiltration pattern of lymphoma cells in relationship to liver parenchyma (portal, sinusoidal, and lobular), as well as the size of lymphoma cells in comparison to the nuclei of macrophages or endothelial cells (small, medium, and large) were recorded for each case based on slide review. 2.3 Statistical analysis Clinicopathologic characteristics were coded as numerical (e.g. age), categorical (e.g. R value, lymphoma infiltration pattern, and lymphoma classification), or dichotomous (e.g. sex). Student's t-test, chi-square or Fisher's exact test, and Kaplan-Meier survival analysis were performed for statistical significance using Prism 8.3.0 (GraphPad Software, San Diego, CA, USA). P -values less than 0.05 were considered statistically significant. 2.2 リンパ腫の診断、分類、および組織形態学的解析 各症例の病理報告書と診断は、各寄稿施設の専門的な血液病理医によって検討された。T細胞腫瘍の分類は,2017年WHO造血器・リンパ系組織の腫瘍分類[ 8 ]に基づいて行った。 本研究で診断されたT細胞腫瘍の具体的な種類は、HSTCL、他に特定されない末梢性T細胞リンパ腫(PTCL-NOS)、T-LGL、ALK陰性ALCL、皮膚T細胞リンパ腫(CTCL)特に菌状息肉症である。血管免疫芽球性 T 細胞リンパ腫(AITL)、移植後リンパ増殖性疾患(PTLD)、節外 NK/T 細胞リンパ腫(NK/T)、T 前リンパ球性白血病(T-PLL)、T 細胞急性リンパ芽球性白血病(T-ALL)等がある。 該当する場合、「原発性」肝リンパ腫は、肝臓に限局したリンパ腫、または他の解剖学的部位に転移する前に最初に主要な肝臓への浸潤を有するリンパ腫と定義される。二次性」肝リンパ腫は、全身性疾患の既往または併発がある場合、または同じリンパ腫が肝転移前に他の解剖学的部位で診断された場合に定義される[ 91011 ]。 肝実質との関係におけるリンパ腫細胞の組織形態と浸潤パターン(門脈、類洞、小葉)、およびマクロファージや内皮細胞の核と比較したリンパ腫細胞のサイズ(小、中、大)は、スライドレビューに基づいて各症例のために記録された。 2.3 統計解析 臨床病理学的特徴は、数値(例:年齢)、カテゴリー(例:R値、リンパ腫浸潤パターン、リンパ腫分類)、または二項対立(例:性)でコード化された。Prism 8.3.0 (GraphPad Software, San Diego, CA, USA) を用いて、Student's t-test, chi-square or Fisher's exact test, Kaplan-Meier survival analysis を行い、統計的有意性を検討した。0.05未満のP値は統計的に有意であるとみなした。 3 Results 3.1 Demographic and clinical data A total of 40 patients, 11 women and 29 men, were included in this study ( Table 1 and Supplementary Table 1 ). The median age was 54 (range: 2?75) years; 4 (10%) were pediatric patients (<18 years old). Fever (n = 13), jaundice (n = 8), and weight loss (n = 8) were the most common presenting clinical symptoms. More than half of the cases (n = 24) showed pancytopenia in peripheral blood. Liver function test ranged from normal to the highest of 2200 IU/L for ALT, 1300 IU/L for ALP, and 28.7 mg/dL for total bilirubin. The majority of cases (n = 20) presented with cholestatic pattern liver enzyme abnormality, followed by hepatitis pattern (n = 6) and mixed (n = 1). Nine patients had normal liver function test at the time of biopsy. On imaging studies, half of the patients (n = 20) had no overt abnormalities, 10 had discrete mass lesions, 6 diffuse hepatomegaly/enlargement, and 4 focal abnormality other than mass. A total of 19 (47.5%) cases were liver primary, whereas 21 (52.5%) were secondary involvement by a previously diagnosed lymphoma/leukemia. The most common diagnoses were HSTCL (n = 14, 35%) and PTCL-NOS (n = 13, 32.5%), totaling more than two-thirds of this study cohort. The remaining 13 patients were diagnosed as follows: T-LGL (n = 2, 5%), ALCL (n = 2, 5%), CTCL (n = 2, 5%), AITL (n = 2, 5%), PTLD (n = 2, 5%), NK/T (n = 1, 2.5%), T-PLL (n = 1, 2.5%), and T-ALL (n = 1, 2.5%). Chemotherapy was the main treatment choices for all T-cell neoplasms. Six patients, including 3 with HSTCL, 3 with PTCL-NOS and 1 with T-PLL underwent stem cell transplant. 成果 3.1 人口統計学的データおよび臨床データ 女性11名、男性29名、合計40名の患者が本研究に参加した(表1、補足表1)。年齢の中央値は54歳(範囲:2?75)、4人(10%)が小児患者(18歳未満)であった。 発熱(n = 13),黄疸(n = 8),体重減少(n = 8)が最も一般的な臨床症状であった.半数以上の症例(n = 24)では,末梢血で汎血球減少が認められた.肝機能検査は正常値からALT2200IU/L,ALP1300IU/L,総ビリルビン28.7mg/dLと最高値に達した.肝酵素異常は,大多数(n=20)が胆汁酸型を呈し,次いで肝炎型(n=6),混合型(n=1)であった. 生検時に肝機能検査が正常であった患者は9名であった。画像検査では,半数(n=20)に明らかな異常はなく,10人に離散的な腫瘤性病変,6人にびまん性の肝腫大/肥大,4人に腫瘤以外の局所的な異常が認められた。合計19例(47.5%)が肝臓原発であったのに対し、21例(52.5%)は既診断のリンパ腫/白血病による二次的病変であった。最も多い診断はHSTCL(n=14、35%)とPTCL-NOS(n=13、32.5%)で、合計するとこの研究コホートの3分の2を超えていた。  残りの13名は以下のように診断されました。T-LGL(n = 2, 5%), ALCL(n = 2, 5%), CTCL(n = 2, 5%), AITL(n = 2, 5%), PTLD(n = 2, 5%), NK/T(n = 1, 2.5%), T-PLL(n = 1, 2.5%), および T-ALL(n = 1, 2.5%), である。化学療法は、すべてのT細胞腫瘍に対する主要な治療法の選択肢であった。HSTCL 3例、PTCL-NOS 3例、T-PLL 1例を含む6例は、幹細胞移植を受けた。 Table 1 Clinical and pathological characteristics of T-cell neoplasms in liver. Patient Diagnosis Age Sex Pertinent clinical history F/U (days) Outcome Imaging R value Histology pattern Main immunophenotype Clonality Molecular/cytogenetics 1 HSTCL (n = 14, 35%) 3 F Mixed phenotype acute leukemia, s/p CTx 107 DOD Abnormal NL Sinusoidal + portal CD3+, CD4-, CD8-, CD5-, CD56-, TIA-1+, TDT- TCRgd Isochromosome 7q 2 55 M Cutaneous DLBCL, s/p CRTx and SCT 99 DOD Normal 0.1 Portal + sinusoidal CD3+, CD4+, CD7-, CD30-, CD56-, CD57+/?, TIA1+ TCRab 3 60 M None 65 DOD Normal 0.2 Sinusoidal CD2+, CD3+, CD4-, CD8-, CD5-, CD7+, CD30-, TIA1+, granzyme B- TCRgd 4 31 M Recurrent HSTCL, s/p CTx and SCT 215 DOD Normal 1.2 Sinusoidal CD2+, CD3-, CD4-, CD8-, CD7+/?, TIA1+, granzyme B-, EBV- TCRgd Monosomy 7 5 49 M Presented with HLH 6 DOD Normal 1.1 Portal + sinusoidal CD2-, CD3+, CD4-, CD8-, CD5-, CD7- 46,XY,del(13)(q14q22), 47,idem,+X and 92,XXYY 6 47 M None 32 DOD Enlarged 32.2 Sinusoidal CD2+, CD3+, CD4-, CD8-, CD5+, CD7-, CD30-, CD56+/?, EBV- 7 71 M None 14 DOD Normal 0.4 Portal + Sinosoidal CD3+, CD25+, CD30-, EBV- 8 4 M None 0 DOD Normal N/A Portal aggregates + sinusoidal CD2+, CD3+, CD4-, CD8-, CD5-, CD7+, CD25-, CD30-, CD56+, TIA1+ TCRgd 9 30 M Ulcerative colitis 89 DOD Normal 14.2 Sinosoidal CD3+, CD4-, CD8+, CD5-, CD16+, CD56+, CD57+ TCRgd 10 21 M None N/A LTF Enlarged NL Sinusoidal CD2+, CD3+, CD4-, CD8+, CD5-, CD7+, CD25-, CD57- TCRab 11 60 F None 37 DOD Enlarged 0.3 Sinusoidal + portal CD3+, CD4-, CD8-, CD5+/?, CD30-, CD56-, CD57+/?, TIA1+, EBV- TCRgd 12 30 M Ulcerative colitis, PSC, AIH NA DOD Abnormal 0.7 Sinusoidal CD2+/?, CD3+, CD4-, CD8-, CD5-, CD7+, CD30-, CD56+, TIA1+, EBV-, ALK- 13 33 M None 619 DOD Normal 1.2 Portal CD2+, CD3+, CD4-, CD8+, CD5+/?, CD7+/?, CD56-, CD57-, TIA1+, granzyme B+, EBV- TCRgd 14 31 M Presented with HLH 13 DOD Normal 1.4 Sinusoidal + portal CD2-, CD3+, CD4-, CD8-, CD5-, CD7+/?, TIA1+, EBV- TCRgd 15 PTCL-NOS (n = 13, 32.5%) 61 M None 76 DOD Mass NL Mass CD2+, CD3+, CD4-, CD8-, CD5-, CD7+/?, CD30+/?, CD56-, TIA1- 16 70 F None 14 DOD Normal 5.5 Portal + lobular aggregates CD2+, CD3+, CD4+, CD8+, CD5-, CD7-, TIA1+, granzyme B- TCRab 17 55 F PTCL in LN x 4 yrs, s/p CTx N/A LTF Mass N/A portal large aggregates + lobules CD3+, CD4+, CD8-, CD5+, CD7+/?, CD30-, EBV-, ALK- 18 27 M EVAN'S syndrome 2600 Alive Mass 1.7 Mass CD3+, CD4+, CD8-, CD5+, CD7+/?, CD30-, CD56-, graznyme B+ TCRab 19 69 M PTCL in LN x 2 mos 213 DOD Normal NL Portal large aggregates + sinosoidal CD2+, CD3+, CD4-, CD8+, CD7+, CD30-, CD56+/?, TIA1+, granzyme B+, EBV-, ALK- TCRab STAT5 mutation 20 59 F PTCL in LN x 8 yrs, s/p SCT 25 DOD Normal 1.5 Portal + sinusoidal CD3+, CD8-, CD5+, CD7-, Trisome 7 21 57 M Involving liver first then LN 30 DOD Normal 0.9 Portal massive CD2+, CD3+, CD4+, CD5+, EBV- 22 54 M PTCL in LN x 1 yr 72 DOD Normal 0.4 Portal + sinusoidal CD2+, CD3+, CD4+, CD8-, CD5+, CD7+/?, CD25+, CD30+/?, CD56+/?, TIA1+, granzyme B-, EBV- 23 70 M PTCL in LN x 1yr, plasma cell dyscrasia 206 DOD Abnormal N/A Portal CD2+, CD3+, CD4+, CD5+, CD7+, EBV- TCRab 24 51 M PTCL in LN x 6 mos 35 Alive Enlarged 0.2 Portal CD3+, CD4+, CD8+/?, CD5+, CD7+/?, CD30+/?, CD57+/?, PD1+, BCL6-, CD10-, CXCL13-, EBV- TCRab Normal cytogenetics 25 54 M Synchronous LN and liver involvment N/A LTF Normal 9.7 Portal + lobular + sinusoidal CD3+, CD4+, CD8-, CD5+, CD30-, CD56-, TIA1+, EBV- TCRab Normal cytogenetics 26 25 M PTCL in LN x 3 mos 1800 DOD Enlarged 0.4 Sinusoidal CD2+, CD3+, CD8+, CD5+, CD7+, CD30-, CD56+/?, CD57+, TIA1+, granzyme B+, EBV- TCRab Normal cytogenetics 27 73 M PTCL in BM x 1 yr 1629 Alive Enlarged NL Portal + sinusoidal CD2+, CD3+, CD4+, CD8-, CD5+/?, CD7+/?, CD30- TCRab Normal cytogenetics 28 T-LGL (n = 2, 5%) 74 M None 132 Alive Normal NL Sinusoidal CD2+, CD3+, CD4-, CD8+, CD5-, CD7-, CD30-, CD56-, CD57+, TIA1+, granzyme B+, EBV-, ALK- TCRab 29 2 F S/p liver transplant for biliary atresia 108 Alive Normal 3.5 Sinusoidal + portal CD2+ CD3+ CD4-CD8+ CD5+ CD7 ± CD56- CD57+ TIA1+ Granzume B + EBV- TCRab 30 ALCL (n = 2, 5%) 71 M Involving liver first, then CSF 52 DOD Mass 0.7 Mass CD3+, CD30+, ALK- 31 62 M ALCL x 13 yrs 360 DOD Mass 1.9 Mass CD2+, CD4+/?, CD30+, EBV-, ALK- 32 CTCL (n = 2, 5%) 73 F Skin x 3 yrs 850 DOD Normal 0.2 Unknown CD2+, CD3+, CD4-, CD8-, CD5+, CD7-, CD30-, ALK- 33 53 M Skin, s/p CRTx; Hepatitis C 152 DOD Mass 0.4 Portal + sinusoidal CD2+, CD3+, CD4+, CD8-, CD5+, CD7-, CD30-, CD56-, CD57-, EBV- 34 AITL (n = 2, 5%) 75 F AITL in LN x 12 yrs, s/p CTx 658 Alive Mass NL Mass CD2+, CD3+, CD8+, CD30+, PD1+, Bcl6+, CD10+/?, EBV- 35 43 M AITL in LN x 2 yrs; Hepatitis C 747 DOD Normal 5.3 Unknown CD2+, CD3+, CD4+, CD8-, CD5+, CD7-, CD30-, EBV+ TCRab 36 PTLD 46 F S/p Liver transplant for congenital hepatic fibrosis 4 DOD Normal 0.8 Portal aggregates + sinusoidal CD3+, CD4+, CD8-, CD30+, CD56-, CD57-, TIA1+, granzyme B+, EBV+, ALK- TCRab 37 PTLD 9 F Presented with HLH, s/p SCT 321 DOD Mass NL mass CD2+, CD3+, CD4+, CD8-, CD5-, CD7-, CD30+, CD56+, CD57-, TIA1+, granzyme B+, EBV+, ALK- TCRab 38 NK/T 62 M Skin x 1 yr 1142 DOD Normal 16.7 Portal + lobular CD3+, CD4-, CD8-, CD7+, CD56+, TIA1+, granzyme B+, EBV+ 39 T-PLL 55 F T-PLL in BM x 1 yr, s/p CTx and SCT 2505 DOD Mass NL Portal + sinusoidal CD3+, CD4+, CD8-, CD5+, CD7+ Complex abnormality a 40 T-ALL 38 M T-ALL in BM x 3 mos 70 DOD Mass NL Mass CD2+, CD3+, CD5-, CD7- フルサイズの画像を見る HSTCL, hepatosplenic T-cell lymphoma; PTCL-NOS, peripheral T-cell lymphoma, not otherwise specified; T-LGL, T-cell large granular lymphocytic leukemia; ALCL, anaplastic large cell lymphoma; CTCL, cutaneous T-cell lymphoma; AITL, angioimmunoblastic T-cell lymphoma; PTLD, post-transplant lymphoproliferative disorder; NK/T, NK/T-cell lymphoma; T-PLL, T-prolymphocytic leukemia; T-ALL, T-cell acute lymphoblastic leukemia; B, biopsy; A, autopsy; BM, bone marrow; LN, lymph node; CSF, cerebrospinal fluid; s/p, status post; CTx, chemotherapy; CRTx, chemoradiation; SCT, stem cell transplant; DLBCL, diffuse large B-cell lymphoma; HLH, hemophagocytic lymphohistiocytosis; PSC, primary sclerosing cholangitis; AIH, autoimmune hepatitis; yr, year; mo, month; ICE, ifosfamide, carboplatin and etoposide; EPOCH, etoposide, prednisone, vincristine, cyclophosphamide and doxorubicin hydrochloride; CVAD, cyclophosphamide, vincristine, doxorubicin, and dexamethasone; ESHAP, etoposide, methylprednisolone, cytarabine, cisplatin; CHOP, cyclophosphamide, doxorubicin, vincristine, and prednisone, DHAP, dexamethasone, cytarabine, cisplatin; BEAM, carmustine, etoposide, cytarabine, melphalan; CHOEP, cyclophosphamide, doxorubicin, vincristine, etoposide and prednisone; FMR, fludarabine, mitoxantrone and rituximab; FC, fludarabine, cyclophosphamide; S-HAM, sequential high-dose cytarabine and mitoxantrone; N/A, not applicable; F/U, follow-up; DOD, dead of disease; LTF, lost to follow-up; R, ratio; NL, normal; EBV, Epstein-barr virus; TCRs, T-cell receptors. ? CBC, complete blood count; WBC, white blood cell (x1000/uL); Hb, hemoglobin (g/dL); Hct, hematocrit (%); Plt, platelets (x1000/uL). a 52?53,XY,add(2)(q35),del(6)(p21),?add(6)(?p23),+add(7)(q36)x2,+8,+20,+20,+21,+21,+i(21)(q10),+mar[cp14]/46,XY[6]. HSTCL、肝脾性T細胞リンパ腫;PTCL-NOS、他に特定されない末梢性T細胞リンパ腫;T-LGL、T細胞大顆粒リンパ球性白血病;ALCL、未分化大細胞リンパ腫。CTCL、皮膚T細胞リンパ腫、AITL、血管免疫芽球性T細胞リンパ腫、PTLD、移植後リンパ増殖性疾患、NK/T、NK/T細胞リンパ腫、T-PLL、T-前リンパ球性白血病。T-ALL、T細胞性急性リンパ芽球性白血病;B、生検;A、剖検;BM、骨髄;LN、リンパ節;CSF、脳脊髄液;s/p、状態後;CTx、化学療法;CRTx、化学放射線療法。SCT、幹細胞移植;DLBCL、びまん性大細胞型B細胞リンパ腫;HLH、血球貪食性リンパ組織球症;PSC、原発性硬化性胆管炎;AIH、自己免疫性肝炎;yr、year;mo、month.ICE、イホスファミド、カルボプラチン、エトポシド;EPOCH、エトポシド、プレドニゾン、ビンクリスチン、シクロホスファミド、塩酸ドキソルビシン;CVAD、シクロホスファミド、ビンクリスチン、ドキソルビシン、デキサメタゾンの4種類。ESHAP、エトポシド、メチルプレドニゾロン、シタラビン、シスプラチン;CHOP、シクロホスファミド、ドキソルビシン、ビンクリスチン、プレドニゾン、DHAP、デキサメタソン、シタラビン、シスプラチン。BEAM、カルムスチン、エトポシド、シタラビン、メルファラン;CHOEP、シクロホスファミド、ドキソルビシン、ビンクリスチン、エトポシド、プレドニゾン;FMR、フルダラビン、ミトキサントロン、リツキシマブ;FC、フルダラビン、シクロフォスファミド。S-HAM、シタラビンとミトキサントロンの大量投与;N/A、適用外;F/U、フォローアップ;DOD、病死;LTF、フォローアップ不能;R、比率;NL、正常;EBV、エプスタインバーウイルス;TCR、T細胞受容体。 ? CBC、全血球数、WBC、白血球(x1000/uL)、Hb、ヘモグロビン(g/dL)、Hct、ヘマトクリット(%)、Plt、血小板(x1000/uL)。 a 52?53,XY,add(2)(q35),del(6)(p21),?add(6)(?p23),+add(7)(q36)x2,+8,+20,+20,+21,+21,+i(21)(q10),+mar[cp14]/46,XY[6]. Upon follow-up, 31 of 37 (83.8%) patients died of lymphoma after a median of 82 (range: 0?2505) days, whereas 6 (16.2%) were alive after a median of 395 (range: 35?2600) days. Three patients were lost to follow-up. Both HSTCL and PTCL-NOS were highly lethal with a short overall survival, with even worse prognosis for HSTCL by Kaplan-Meier survival analysis when compared to non-HSTCL cases ( Fig. 1 ). There were no significant clinical and survival differences between patients with “primary” vs. “secondary” T-cell neoplasms in liver (data not shown). Fig. 1 Kaplan-Meier survival analysis in patients with T-cell neoplasms in liver. (A) Patients of HSTCL had worse clinical course than PTCL-NOS, but the difference did not reach statistical significance ( P = 0.078). The clinical course was significantly worse in patients with HSTCL when compared with other types of T-cell lymphomas except HSTCL and PTCL-NOS (A), or when compared with all non-HSTCL T-cell neoplasms (B). HSTCL, hepatosplenic T-cell lymphoma; PTCL-NOS, peripheral T-cell lymphoma, not otherwise specified. 3.2 Histologic analysis and correlation with imaging and laboratory results Histomorphologic analysis and correlation with imaging studies and liver enzymes were performed on all but two cases. Four major infiltration patterns were summarized ( Fig. 2 ): sinusoidal predominant (pattern 1; n = 12), portal predominant with sinusoidal infiltrates (pattern 2; n = 13), portal and lobular infiltrates with lymphoid aggregates (pattern 3; n = 5), and mass forming (pattern 4; n = 8). In both patterns 1 and 2, the hepatic architecture was relatively preserved with no discrete lymphoid nodules or large aggregates. Pattern 3 was characterized by lymphoid aggregates or small nodules predominantly in both portal tracts and lobules ( Fig. 3 ). In pattern 4, the lymphoma formed mass lesions and effaced the underlying hepatic architecture by high-grade tumor cells with frequent mitoses and apoptosis ( Fig. 4 ). Pattern 4 almost always correlated with a sizable “mass” lesion on imaging studies ( Table 1 and Supplementary Table 1 ). In all patterns, monotonous lymphoid infiltrates and/or larger cell size were usually the features that prompted lymphoma work-up, especially in cases without a prior lymphoma diagnosis. 追跡調査の結果、37人中31人(83.8%)が中央値82日(範囲:0?2505)後にリンパ腫で死亡し、一方、6人(16.2%)が中央値395日(範囲:35?2600)後に生存していた。3名の患者は追跡調査から外れた。HSTCLとPTCL-NOSはともに致死率が高く、全生存期間は短く、HSTCLの予後はKaplan-Meier生存解析で非HSTCL例と比較してさらに悪い(図1)。肝臓のT細胞腫瘍の「原発性」と「続発性」の患者間には、臨床的にも生存期間的にも有意な差は認められなかった(データは示されていない)。 図1 肝臓のT細胞腫瘍の患者におけるKaplan-Meier生存率分析。(A)HSTCLの患者はPTCL-NOSよりも臨床経過が悪かったが、その差は統計的に有意ではなかった(P = 0.078)。HSTCLとPTCL-NOSを除く他のT細胞リンパ腫と比較した場合(A)、あるいはHSTCL以外のすべてのT細胞腫瘍と比較した場合(B)、HSTCL患者の臨床経過は有意に不良であった。HSTCLは肝脾T細胞リンパ腫、PTCL-NOSは他に特定されない末梢T細胞リンパ腫。 3.2 組織学的解析と画像診断・検査結果との相関性 2例を除くすべての症例で、組織形態学的解析と画像検査および肝酵素との相関が行われた。4つの主要な浸潤パターンが要約された(図2):洞様優勢(パターン1;n=12)、洞様浸潤を伴う門脈優勢(パターン2;n=13)、リンパ凝集塊を伴う門脈および小葉浸潤(パターン3;n=5)、腫瘤形成(パターン4;n=8)であった。パターン1、2ともに、肝構造は比較的保たれており、個別のリンパ節や大きな凝集塊は認められなかった。パターン3では、門脈と小葉の両方にリンパ球の集合体や小結節が多く見られた(図3)。パターン4では、リンパ腫は腫瘤病変を形成し、頻繁な有糸分裂とアポトーシスを伴う高悪性度腫瘍細胞によって、その下の肝構造を消してしまった(Fig. 4)。パターン4は、画像検査においてほとんど常に大きな「腫瘤」病変と相関していた( 表1および補足表1 )。すべてのパターンにおいて、単調なリンパ球浸潤と、より大きな細胞サイズが、特にリンパ腫の診断歴のない症例において、通常リンパ腫のワークアップを促す特徴であった。 フルサイズの画像を開く Fig. 2 Histomorphologic analysis of T-cell neoplasms in liver, and correlation with imaging studies and R value of liver enzyme tests. Number in “()” indicates total number of cases. NL, normal, N/A, not applicable or available. フルサイズの画像を開く Fig. 3 T-cell neoplasms exhibiting histology patterns 1 (A?C), 2 (D?F), and 3 (G?I). Pattern 1 was represented by HSTCL with small-sized (A) and medium-sized (B) lymphoid cells expanding the hepatic sinusoid, and T-LGL with classic sinusoidal linear distribution (C). Pattern 2 was represented by HSTCL (D and E) and PTCL-NOS (F) showing portal and sinusoidal infiltrates. Pattern 3 was represented by PTCL-NOS (G and H) and NK/T (I) demonstrating prominent lymphoid aggregates in portal tracts and heavy lobular infiltrates. Magnification: 200×. HSTCL, hepatosplenic T-cell lymphoma; PTCL-NOS, peripheral T-cell lymphoma, not otherwise specified. フルサイズの画像を開く Fig. 4 T-cell neoplasms exhibiting histology pattern 4 with massive infiltration effacing underlying hepatic structures, represented by ALCL (A and B), AITL (C), PTCL-NOS (D), T-cell PTLD (E), and T-ALL (F). Magnification: 200×. PTCL-NOS, peripheral T-cell lymphoma, not otherwise specified; ALCL, anaplastic large cell lymphoma; AITL, angioimmunoblastic T-cell lymphoma; PTLD, post-transplant lymphoproliferative disorder. The lymphoma infiltration pattern did not always correlate with the pattern of liver function tests; however, the liver enzyme elevation (R > 5) occurred only in patterns 1 and 3, where lymphoma cells showed predominant sinusoidal infiltration (2 HSTCL), or lobular aggregates with hepatocyte injury (2 PTCL-NOS and 1 NK/T). Patterns 2 and 4 more often correlated with a cholestatic enzyme elevation or normal liver function tests ( Fig. 1 ). 図2 肝臓のT細胞腫瘍の組織形態学的解析、および画像検査や肝酵素検査のR値との相関。()」内の数字は総症例数を示す。NL, normal, N/A, not applicable or available. 図3 組織型パターン1(A?C)、2(D?F)、3(G?I)を示すT細胞腫瘍。パターン1は、肝類洞を拡大する小型(A)および中型(B)のリンパ球を伴うHSTCLと、古典的な類洞線状分布を有するT-LGLで表される(C)。パターン2はHSTCL(D、E)、PTCL-NOS(F)で、門脈および類洞への浸潤を示す。パターン3はPTCL-NOS(G、H)とNK/T(I)で、門脈のリンパ球の凝集と小葉浸潤が顕著であった。倍率は200倍。HSTCLはhepatosplenic T-cell lymphoma、PTCL-NOSはperipheral T-cell lymphoma、not otherwise specified。 フルサイズの画像を開く 図4. ALCL(AおよびB)、AITL(C)、PTCL-NOS(D)、T細胞PTLD(E)およびT-ALL(F)で表される、大量の浸潤で肝細胞の下層構造を侵す組織像パターン4のT細胞性新生物。倍率:200倍。PTCL-NOSはperipheral T-cell lymphoma, not otherwise specified;ALCLはanaplastic large cell lymphoma;AITLはangioimmunoblastic T-cell lymphoma;PTLDはpost-transplant lymphoproliferation disorder。 リンパ腫の浸潤パターンと肝機能検査のパターンは必ずしも相関しないが、肝酵素の上昇(R>5)はパターン1と3にのみ見られ、リンパ腫細胞が優勢な類洞浸潤(HSTCL 2例)、または肝細胞障害を伴う小葉凝集塊(PTCL-NOS 2例、NK/T 1例)が見られた。パターン2と4は、胆汁酸酵素の上昇や肝機能検査が正常であることと相関することが多かった(図1)。 3.3 Comparisons between HSTCL and PTCL-NOS Since HSTCL and PTCL-NOS were the most frequent diagnoses in our cohort, each comprising approximately one third of the total cases, we compared these two lymphoma types regarding clinicopathologic features ( Table 2 ). Patients with HSTCL were much younger than those of PTCL-NOS (median age 31 vs. 55, P = 0.018). Both were male predominant. For HSTCL, 2 patients had prior history of hematopoietic malignancies other than HSTCL, 2 had history of ulcerative colitis, and 2 presented with hemophagocytic lymphohistiocytosis (HLH) before lymphoma diagnosis. Most patients with HSTCL had either spleen and/or bone marrow involvement with typical sinusoidal infiltration. Involvement of the appendix and lymph node in one case (patient 8) was also noted. On imaging studies, three cases showed hepatomegaly and two revealed focal abnormality, whereas all others had normal imaging findings. Liver function tests were predominantly cholestatic (n = 9/14, 64%). Immunophenotypes were mostly CD3+/CD4-/CD8-/TIA1+/TCRγδ consistent with non-activated cytotoxic gamma/delta T-cells. In comparison, patients with PTCL-NOS usually had no history of other lymphomas or immunosuppression. The majority had established PTCL-NOS diagnoses in lymph nodes (10/13, 77%), spleen (2/13, 15%), and/or bone marrow (2/13, 15%) before liver involvement. Only two patients (patients 15 and 18) had exclusive liver involvement, both showing mass lesions on imaging studies and a normal or cholestatic liver function test, suggesting “primary” hepatic PTCL-NOS. Histologically, all HSTCL cases presented as small to medium-sized lymphoid cells with sinusoidal infiltrates corresponding to pattern 1 (9/14, 64%) or pattern 2 (5/14, 36%), whereas PTCL-NOS showed all four histology patterns though the majority demonstrated portal infiltrates corresponding to patterns 2 or 3 (9/13, 69%) ( Figs. 2 and 3 ). In contrast to HSTCL, immunophenotypes in PTCL-NOS were mostly CD3+/CD4+/CD8-/TCRαβ, with rare CD4/CD8 double negative or double positive cases. Although all patients with HSTCL died of disease after a median follow-up of 51 (range: 0?619) days, at least one patient with primary hepatic PTCL-NOS survived after stem cell transplant on a follow-up of 2600 days. 3.3 HSTCLとPTCL-NOSの比較 HSTCLとPTCL-NOSは我々のコホートで最も頻繁に診断され、それぞれ全症例の約1/3を占めていたので、臨床病理学的特徴についてこの2つのタイプを比較した(表2)。  HSTCLの患者はPTCL-NOSの患者よりはるかに若かった(年齢中央値31 vs. 55、P = 0.018)。両者とも男性優位であった。HSTCLでは,2名がHSTCL以外の造血器悪性腫瘍の既往,2名が潰瘍性大腸炎の既往,2名がリンパ腫診断前に血球貪食性リンパ組織球症(HLH)を呈していた。 HSTCLの患者の多くは,脾臓および骨髄のいずれかに病変を有し,典型的な類洞浸潤を伴っていた.1例(患者8)では、虫垂とリンパ節への浸潤も認められた。画像検査では、3例に肝腫大、2例に局所的な異常が認められたが、他はすべて正常であった。肝機能検査では胆汁酸が主体であった(n = 9/14、64%)。免疫表現型はCD3+/CD4-/CD8-/TIA1+/TCRγδが多く、非活性化細胞傷害性γ/δT細胞と一致した。  一方、PTCL-NOSの患者は通常、他のリンパ腫や免疫抑制の既往がなかった。大半の患者は、肝臓病変の前にリンパ節(10/13、77%)、脾臓(2/13、15%)、骨髄(2/13、15%)のPTCL-NOSの診断が確立していた。   2例(15例と18例)だけが肝臓に病変があり、画像診断で腫瘤性病変を示し、肝機能検査は正常または胆汁性で、「原発性」肝性PTCL-NOSが示唆された。 組織学的には,HSTCLではパターン1(9/14,64%)あるいはパターン2(5/14,36%)に相当する類洞浸潤を伴う小ー中型リンパ球が全例に認められたが,PTCL-NOSではパターン2あるいは3(9/13,69%)に相当する門脈浸潤が大多数に認められた(図2,3).HSTCLとは対照的に、PTCL-NOSの免疫型はCD3+/CD4+/CD8-/TCRαβが多く、まれにCD4/CD8の二重陰性や二重陽性の症例もみられた。HSTCLは中央値51日(範囲:0?619日)の経過観察で全例が死亡したが、肝原発PTCL-NOSの少なくとも1例は2600日の経過観察で幹細胞移植後生存していた。 Table 2 Comparisons between HSTCL and PTCL-NOS. 5--> HSTCL (n = 14) PTCL-NOS (n = 13) P value Age, median, range 32 (3?71) 57 (26?73) 0.015 Sex 2F, 12M 3F, 10M NS Immunosuppression due to prior malignancy or IBD Prior malignancy 2 0 NS IBD 2 0 None 10 13 R value Cholestatic 9 6 Hepatitic 2 2 NS Normal 2 3 N/A 1 2 Imaging findings Mass 0 3 NS Enlarged 3 3 Abnormal 2 1 Normal/None 9 6 Histology 1. Sinusoidal 6 1 0.008 2. Portal + sinusoidal 8 5 3. Portal + lobular 0 4 4. Massive 0 3 Outcomes DOD 13 8 NS Alive 0 3 LTF 1 2 Follow-up days 51 (0?619) 76 (14?2600) NS フルサイズの画像を見る IBD, inflammatory bowel disease; DOD, died of disease; LTF, lost to follow-up; NS, not significant; HSTCL, hepatosplenic T-cell lymphoma; PTCL-NOS, peripheral T-cell lymphoma, not otherwise specified. 3.4 Other lymphomas Two T-LGL were identified in our cohort ( Table 1 and Supplementary Table 1 ). One was a 74-year-old men (patient 28) with no significant past medical history, the other one was a 2-year-old women (patient 29) with a complicated history including embryonic biliary atresia status post liver transplant, EBV viremia, and upper gastrointestinal B-cell PTLD, polymorphic type. Both had liver biopsies demonstrating sinusoidal T-cell infiltration mimicking HSTCL ( Fig. 3 C), but an immunophenotype of CD3+/CD8+/TIA1+/Granzyme B+/CD57+/EBV-/TCRαβ+. Both patients responded to treatment well and continued to do well on clinical follow-up. The two patients with T-cell PTLD had very aggressive clinical course and died of disease within one year of diagnosis. The first was a 46-year-old women (patient 36) status after liver transplant for congenital hepatic fibrosis. Liver biopsy showed portal/sinusoidal infiltrates with an immunophenotype of CD4+/TIA+/Granzyme B+/EBER+ ( Fig. 5 A). The second was a 9-year-old women (patient 37) with a history of persistent active hepatitis (ALT up to 1430 IU/L, R = 25.3) clinically suspicious for HLH. Liver biopsy showed portal/lobular small mature lymphocytic infiltrates, retrospectively stained positive for EBV ( Fig. 5 B). She underwent 9 of 10 HLA-matched unrelated donor bone marrow transplant but soon developed multiple liver mass lesions. The liver biopsy showed a high-grade lymphoma with an immunophenotype of CD4+/CD56+/TIA-1+/Granzyme B+/EBV LMP-1+ ( Figs. 4D and 5 C). 3.4 その他のリンパ腫 我々のコホートでは2例のT-LGLが同定された(表1および補足表1)。一人は74歳の男性(患者28)で過去に目立った病歴はなく、もう一人は2歳の女性(患者29)で、肝移植後の胚性胆道閉鎖状態、EBVウイルス血症、上部消化管B細胞PTLD、多型などの複雑な病歴を有していた。両者とも肝生検でHSTCLに類似した類洞T細胞浸潤を認めたが(図3 C)、免疫表現型はCD3+/CD8+/TIA1+/Granzyme B+/CD57+/EBV-/TCRαβ+であった。両者とも治療への反応は良好で、臨床的なフォローアップも良好に継続した。 T細胞性PTLDの2例は、非常に侵襲的な臨床経過を示し、診断後1年以内に病死した。一人目は46歳の女性(患者36)で、先天性肝線維症に対する肝移植後の状態であった。肝生検では、CD4+/TIA+/Granzyme B+/EBER+の免疫表現型を持つ門脈/類洞浸潤を示した(図5 A)。もう一人は、臨床的にHLHが疑われる活動性肝炎(ALTが1430IU/Lまで、R=25.3)が持続する9歳の女性(患者37)であった。肝生検では門脈/小葉の小成熟リンパ球浸潤が認められ、レトロスペクティブにEBV陽性と染色された(図5 B)。HLA適合10例中9例の非血縁ドナー骨髄移植を受けたが、まもなく多発性肝腫瘤病変を発症した。肝生検では、免疫表現型が CD4+/CD56+/TIA-1+/グランザイムB+/EBV LMP-1+ の高悪性度リンパ腫が認められた(図 4D と 5 C)。 フルサイズの画像を開く Fig. 5 Epstein-barr virus (EBV) testing in liver biopsies. EBV was detected in a PTLD with portal and sinusoidal infiltrates (A), the 9-year-old with chronic EBV infection in pre-transplant liver biopsy (B) and massive high-grade T-cell PTLD after bone marrow transplant (C), and an extranodal NK/T-cell lymphoma, nasal type (D). Magnification: 200×. PTLD, post-transplant lymphoproliferative disorder. The two ALK-negative ALCL cases, one likely being liver “primary”, occurred in elderly men and presented as mass lesions on imaging, with mild cholestatic pattern liver function abnormality. Histology revealed high-grade cytology with characteristic hallmark cells ( Fig. 4 A and B). T-ALL and T-PLL, both secondary liver involvement by a systemic disease, presented as mass lesions and near normal liver function tests. Two CTCL (mycosis fungoides), two AITL, and one NK/T had prior diagnosis of the same lymphoma in skin or lymph node. The one case of extranodal NK/T-cell lymphoma, nasal type demonstrated portal and lobular infiltrates (pattern 3), marked hepatotoxicity (R = 16.7), and strong EBER positivity ( Fig. 5 D). With the exception of one AITL, all patients died of disease upon follow-up. 図5 肝生検におけるEpstein-barrウイルス(EBV)検査。門脈・類洞浸潤を伴うPTLD(A)、移植前肝生検で慢性EBV感染を認め(B)、骨髄移植後に大量の高グレードT細胞性PTLD(C)、節外NK/T細胞リンパ腫、鼻型(D)でEBVを検出した9歳の症例。倍率:200倍。PTLD、移植後リンパ増殖性疾患。 ALK陰性のALCL2例(1例は肝「原発」である可能性が高い)は、高齢男性に発症し、画像上では腫瘤性病変として認められ、軽度の胆汁性パターン肝機能異常が見られた。組織学的には、特徴的なホールマーク細胞を伴う高悪性度の細胞診が行われた(図4 A、B)。T-ALLとT-PLLは全身性疾患による二次性肝障害で,腫瘤性病変を呈し,肝機能検査はほぼ正常であった。CTCL(菌状息肉症)2例、AITL2例、NK/T1例は、皮膚またはリンパ節に同じリンパ腫の診断歴があった。節外性NK/T細胞リンパ腫の1例は、門脈および小葉への浸潤(パターン3)、著しい肝障害(R = 16.7)、強いEBER陽性を示した(図5 D)。AITL1名を除き、全例が経過観察中に死亡した。 4 Discussion In this multi-institutional study, we reviewed a total of 40 cases of T-cell neoplasms diagnosed in the liver to characterize the clinicopathologic features. The types of neoplasms in this study covered nearly 70% of the WHO classification of mature T- and NK-cell neoplasms, the largest and most comprehensive to date. The majority of diagnoses were made in liver biopsies with pertinent clinical history and necessary ancillary tests for confirmation. Our study showed that hepatic T-cell neoplasms occurred in a wide age range, were male predominant, and had a generally dismal prognosis. The integrated correlation analysis on imaging studies, laboratory tests, and histomorphology patterns may serve as a diagnostic guideline for surgical pathologists who frequently encounter the liver biopsies before hematopathology consultation and lymphoma work-up. Hepatic involvement by lymphoma can be difficult to distinguish from reactive conditions or other malignancies [ 12 ]. Unlike other primary or metastatic neoplasm in the liver, lymphoma may not form a mass lesion [ 2 ]; instead, the lymphoma cells infiltrate into the hepatic sinusoids or portal tracts, compressing the hepatic plate and biliary outflow, causing liver enzyme elevation [ 131415 ]. The damage to liver is more likely a pushing effect rather than hepatotoxic, which explains why ALP elevation is more frequent than transaminitis. The abnormal liver function tests together with an infiltrative growth pattern without mass formation frequently mimics drug-induced liver injury or viral infection. Although radiographic and laboratory tests are not always reliable in detecting lymphoma [ 15 ], correlation with histomorphology increases awareness and diagnostic accuracy. On the other hand, a mass-forming lymphoma may be easier to recognize as a malignancy but can be difficult to distinguish from other primary or metastatic non-hematopoietic neoplasms [ 16 , 17 ]. Thus, pattern recognition, morphologic suspicion, and prudent work-up are key to reaching proper diagnosis. HSTCL accounted for approximately one-third of all T-cell neoplasms in our cohort. In line with the literature showing that approximately 20% HSTCL occurred in patients with immunosuppression [ 18 ], a similar percentage of patients with inflammatory bowel disease or prior diagnosis of other lymphoma/leukemia was found in our case series. HSTCL is a well-known underlying etiology for HLH [ 19 ], an observation again recapitulated in our study. The sinusoidal infiltration pattern is characteristic for HSTCL, but not exclusive, as T-LGL and PTCL-NOS can have similar histomorphology, therefore relying on IHC or other ancillary tests for definitive diagnosis. Indeed, a European study published in 2007 showed that HSTCL only accounted for 1 of 19 T-cell lymphomas involving the liver [ 1 ], strikingly lower than the proportion of HSTCL in our series. The difference may be partially attributed to geographic factors but also likely due to more extensive diagnostic tests to accurately distinguish HSTCL from other lymphomas. T-LGL serves as a top differential diagnosis for HSTCL by histology. Immunophenotypically, T-LGL is typically CD8+/TIA1+/Granzyme B+/CD57+/TCRαβ+, despite some variants and overlaps [ 20 , 21 ]. T-LGL is usually indolent with lower mortality, in contrast to the rapid progression of HSTCL. A takeaway from the pediatric T-LGL case in our cohort is to keep in mind that although T-LGL usually occurs in elderly patients with no associated risk factors, it happens in all age groups, particularly those with immunosuppression due to solid organ transplant or treatment for inflammatory bowel disease [ 22 , 23 ]. PTCL-NOS represented the second largest group of T-cell neoplasm in our study. As a diagnosis of exclusion, PTCL-NOS refers to a heterogeneous group of mature T cell lymphomas that cannot be classified into a specific category according to the current WHO classification [ 24 , 25 ]. Consistent with a general lack of a specific etiology, no prior malignancy diagnosis other than PTCL-NOS nor history of inflammatory bowel disease were identified in our case cohort. The majority were that of CD4+/CD8-or CD4-/CD8+ T cells, frequently TCR-αβ+, and with loss of one or more pan-T-cell antigens. While most PTCL-NOS were diagnosed in lymph nodes, at least two cases showed initial or predominant liver presentation with mass-forming lesions, for which we suspected a primary hepatic PTCL-NOS. Of note, primary hepatic PTCL-NOS is extremely rare, some associated with EBV infection [ 4 ]. Although we did not find significant survival differences between “primary” and “secondary” lymphomas in our study, it may warrant further studies to compare larger cohorts in each group for potential significant clinical implications. 考察 この多施設共同研究において、我々は肝臓で診断されたT細胞腫瘍40例を検討し、その臨床病理学的特徴を明らかにした。本研究で対象とした新生物はWHOの成熟T細胞およびNK細胞新生物の分類の70%近くをカバーし、これまでで最大かつ最も包括的なものであった。診断の大部分は肝生検で行われ、適切な臨床歴と確認のために必要な補助的検査が行われた。我々の研究は、肝T細胞腫瘍が幅広い年齢層に発生し、男性優位であり、一般に悲惨な予後であることを明らかにした。画像検査、臨床検査、組織形態学的パターンに関する統合的な相関分析は、血液病理学的診察やリンパ腫ワークアップの前に肝生検に頻繁に遭遇する外科病理医にとって診断ガイドラインとなる可能性がある。 リンパ腫による肝障害は、反応性疾患や他の悪性腫瘍との鑑別が困難なことがある[ 12 ]。肝臓の他の原発性または転移性新生物とは異なり、リンパ腫は腫瘤病変を形成しないことがある[2];その代わりに、リンパ腫細胞が肝類洞や門脈に浸潤して肝板や胆道流出を圧迫し、肝酵素の上昇を引き起こす[131415]。肝臓へのダメージは、肝毒性よりもむしろプッシュ効果である可能性が高く、ALP上昇がトランスアミン炎よりも頻度が高い理由を説明している。肝機能検査の異常は、腫瘤を形成しない浸潤性増殖パターンとともに、しばしば薬剤性肝障害やウイルス感染症を模倣する。X線検査や臨床検査は、リンパ腫の検出において必ずしも信頼できるものではないが [ 15 ] 、組織形態学との相関は、認知度と診断精度を高める。一方、腫瘤形成性リンパ腫は悪性腫瘍として認識しやすいが、他の原発性または転移性の非造血性新生物との鑑別が困難な場合がある [ 16 , 17 ]。したがって、パターン認識、形態学的疑い、および慎重なワークアップが、適切な診断に至る鍵である。 HSTCLは、我々のコホートにおけるすべてのT細胞腫瘍の約3分の1を占めた。約20%のHSTCLが免疫抑制のある患者に発生したという文献 [ 18 ]と同様に、炎症性腸疾患または他のリンパ腫/白血病の診断歴のある患者も我々のケースシリーズで認められた。HSTCLはHLHの基礎疾患としてよく知られており[19]、この観察結果は我々の研究でも再現されている。洞様浸潤パターンはHSTCLに特徴的であるが、T-LGLとPTCL-NOSが類似の組織形態を示すことがあるため、排他的ではなく、確定診断にはIHCや他の補助的検査に頼ることになる。実際、2007年に発表されたヨーロッパの研究では、肝臓を侵すT細胞リンパ腫19例中HSTCLは1例のみであり[ 1 ]、我々のシリーズにおけるHSTCLの割合よりも驚くほど低いことが示されている。この差は部分的には地理的な要因に起因しているかもしれないが、HSTCLを他のリンパ腫と正確に区別するために、より広範な診断検査が行われたためと思われる。 T-LGLは組織学的にHSTCLの鑑別診断の上位に位置づけられる。免疫表現型的には、T-LGLは、いくつかの変異や重複はあるものの、典型的にはCD8+/TIA1+/Granzyme B+/CD57+/TCRαβ+である[ 20 , 21 ]。T-LGLは、HSTCLの急速な進行とは対照的に、通常、死亡率の低い低年齢層です。我々のコホートにおける小児T-LGL症例から得られる教訓は、T-LGLは通常、関連する危険因子のない高齢者に発生するが、すべての年齢層、特に固形臓器移植や炎症性腸疾患の治療による免疫抑制のある患者に起こることを念頭に置いておくことだ[ 22 , 23 ]。 PTCL-NOSは、我々の研究においてT細胞腫瘍の2番目に大きなグループであった。除外診断として、PTCL-NOSは、現在のWHO分類に従って特定のカテゴリーに分類できない成熟T細胞リンパ腫の異質なグループを指す[ 24 , 25 ]。特定の病因の一般的な欠如と一致して、PTCL-NOS以外の悪性腫瘍の診断歴や炎症性腸疾患の既往は、我々のケースコホートで確認されなかった。大多数は、CD4+/CD8またはCD4-/CD8+ T細胞のもので、しばしばTCR-αβ+で、1つ以上の汎T細胞抗原が失われていた。ほとんどのPTCL-NOSはリンパ節で診断されたが、少なくとも2例は腫瘤形成性病変を伴う初発または優勢な肝臓症状を示し、我々は肝原発性PTCL-NOSを疑った。注目すべきは、原発性肝性PTCL-NOSは極めて稀であり、一部はEBV感染に関連していることである[ 4 ]。我々の研究では、「原発性」と「二次性」のリンパ腫の間に生存率の有意差は認められなかったが、臨床的に重要な意味を持つ可能性があるため、各グループの大規模コホートを比較するさらなる研究が必要であろう。 EBV is etiologically linked to a wide range of lymphoproliferative disorders and malignant lymphomas [ 26 , 27 ]. In our study, EBV infection was associated with at least 3 types of lymphoproliferative disorders: NK/T, AITL, and PTLD. Interestingly, one PTLD case developed after bone marrow transplant was retrospectively found to have chronic active EBV infection in pre-transplant liver biopsy, stressing the importance of EBV test for some lymphoma types. EBV + T-cell PTLD is uncommon, and rare cases of synchronous EBV + B- and T-cell PTLD have been reported [ 28 ]. Interestingly, the current WHO classification of T-cell PTLD covers a wide range of T-cell neoplasms including HSTCL and T-LGL in the clinical setting of solid organ or bone marrow transplant [ 8 ], which makes the two HSTCL and one T-LGL in our cohort classifiable as EBV-negative T-cell PTLD under that umbrella, revealing the complexity of lymphoma etiology and overlaps in classification. Although only a limited number of cases were tested, cytogenetics and molecular studies have been playing an increasingly important role in lymphoma diagnosis and management [ 29 ]. In our cohort, isochromosome 7q was applied to confirm HSTCL in a very complicated case with a prior diagnosis of mixed phenotype acute leukemia, stressing the diagnostic value of cytogenetics in certain cases. The molecular consequences of isochromosome 7q in HSTCL is unknown, although many candidate genes have been proposed [ 30 ]. Other genetic or molecular abnormalities are not disease-specific but are helpful in differential diagnosis. For example, activating STAT5 mutations have been reported in multiple PTCL-NOS, HSTCL, and some aggressive variant of T-LGL [ 313233 ], suggesting STAT signaling pathway as a common driver mutation in certain groups of T-cell lymphomas. Overlaps at the molecular levels and IHC between AITL and PTCL-NOS were also reported [ 34 ]. One study identified shared molecular features between NK-cell lymphoma and a group of non-HSTCL T-cell lymphoma with sensitivity to a novel aurora kinase A inhibitor [ 35 ], suggesting that molecular tests may not only help understanding lymphomagenesis, but also revealing therapeutic targets. EBVは、広範なリンパ増殖性疾患や悪性リンパ腫と病因的に関連している[ 26 , 27 ]。我々の研究では、EBV感染は少なくとも3つのタイプのリンパ増殖性障害と関連していた。NK/T、AITL、およびPTLDである。興味深いことに、骨髄移植後に発症したPTLDの1例は、移植前の肝生検で慢性的に活動性のEBV感染が確認され、いくつかのリンパ腫タイプに対するEBV検査の重要性が強調された。EBV+T細胞PTLDは珍しく、EBV+B細胞およびT細胞PTLDの同期例がまれに報告されている[ 28 ]。興味深いことに、現在のWHOのT細胞PTLDの分類は、固形臓器または骨髄移植の臨床環境におけるHSTCLとT-LGLを含む広範囲のT細胞腫瘍を対象としています[ 8 ]。このため、我々のコホートのHSTCL 2つとT-LGL 1つは、その傘下のEBV陰性T細胞PTLDとして分類可能で、リンパ腫病因と分類における重複がいかに複雑かが明らかにされています。 限られた症例しか検査されていないが、細胞遺伝学と分子研究は、リンパ腫の診断と管理においてますます重要な役割を果たすようになってきている[ 29 ]。我々のコホートでは、混合表現型急性白血病の先行診断を受けた非常に複雑な症例において、HSTCLを確定するために7q染色体を適用し、特定の症例における細胞遺伝学の診断的価値を強調している。HSTCLにおける7q染色体の分子的帰結は不明であるが、多くの候補遺伝子が提案されている[ 30 ]。その他の遺伝子異常や分子異常は、疾患特異的ではないが、鑑別診断に有用である。例えば、活性化STAT5変異は、複数のPTCL-NOS、HSTCL、およびT-LGLのいくつかの侵攻型変異で報告されており[313233]、STATシグナル伝達経路が特定のグループのT細胞リンパ腫に共通のドライバー変異であることが示唆されている。AITLとPTCL-NOSの間の分子レベルおよびIHCにおける重複も報告されている[34 ]。ある研究では、NK細胞リンパ腫と、新規オーロラキナーゼA阻害剤に感受性のある非HSTCL T細胞リンパ腫のグループに共通の分子的特徴を同定し、分子検査はリンパ腫の発生を理解するだけでなく、治療標的も明らかにできる可能性を示唆している[ 35 ]。 5 Conclusion In summary, we performed the largest comprehensive study on T-cell neoplasms in the liver to characterize the clinicopathologic features, covering a wide range of low-grade vs. high-grade, precursor vs. mature, and primary vs. secondary T-cell lymphomas/proliferative disorders. An integrated analysis on imaging, laboratory liver function tests, histomorphologic features, and immunophenotypes was performed to provide clues for proper workup and accurate diagnosis. We focused specifically on common entities such as HSTCL, PTCL-NOS, T-LGL, and PTLD, the diagnostic pitfalls, and lessons learned from challenging cases. As a heterogeneous group of lymphomas that is difficult to define and classify, future studies may focus on large comprehensive testing such as next-generation sequencing for more accurate diagnosis, better classification, and targeted therapy strategies. 結論 要約すると、我々は肝臓のT細胞腫瘍に関する最大の包括的研究を行い、低悪性度対高悪性度、前駆体腫瘍対成熟細胞腫瘍、原発性対二次性T細胞リンパ腫/増殖性障害の広い範囲に及ぶ臨床病理学的特徴を明らかにした。画像診断、肝機能検査、組織形態学的特徴、免疫表現型を統合的に解析し、適切なワークアップと正確な診断のための手がかりを提供した。特に、HSTCL、PTCL-NOS、T-LGL、PTLDなどの一般的な疾患、診断の落とし穴、困難な症例から得られた教訓に焦点を当てた。定義や分類が困難な異種リンパ腫群として、今後の研究では、より正確な診断、より良い分類、標的治療戦略のために、次世代シーケンサーのような大規模な包括的検査に焦点を当てることが考えられます。 Acknowledgements The authors would like to thank the Department of Pathology and Laboratory Medicine at University of Rochester Medical Center for technical support. Appendix A Supplementary data The following is the supplementary data to this article: Multimedia component 1 Competing interests: None. References 1. Loddenkemper C., Longerich T., Hummel M., Ernestus K., Anagnostopoulos I., Dienes H.P., et. al.: Frequency and diagnostic patterns of lymphomas in liver biopsies with respect to the WHO classification. Virchows Arch 2007; 450: pp. 493-502. <% viewInSource %><% crossRef %> 2. Bach A.G., Behrmann C., Holzhausen H.J., Spielmann R.P., Surov A.: Prevalence and imaging of hepatic involvement in malignant lymphoproliferative disease. Clin Imag 2012; 36: pp. 539-546. <% viewInSource %><% crossRef %> 3. Bai Y.F., Liu J.M., Zhang X.M., Jiang C.Z., Xu X., Zheng S.S.: Percutaneous liver biopsy: retrospective study of primary and secondary hepatic lymphoma in twenty-one patients. Hepatobiliary Pancreat Dis Int 2017; 16: pp. 58-64. <% viewInSource %> 4. Ramai D., Ofori E., Nigar S., Reddy M.: Primary hepatic peripheral T-cell lymphoma associated with Epstein-Barr viral infection. World J Hepatol 2018; 10: pp. 347-351. <% viewInSource %><% crossRef %> 5. Alonsozana E.L., Stamberg J., Kumar D., Affe E.S., Medeiros L.J., Frantz C., et. al.: Isochromosome 7q: the primary cytogenetic abnormality in hepatosplenic gammadelta T cell lymphoma. Leukemia 1997; 11: pp. 1367-1372. <% viewInSource %><% crossRef %> 6. Duyster J., Bai R.Y., Morris S.W.: Translocations involving anaplastic lymphoma kinase (ALK). Oncogene 2001; 20: pp. 5623-5637. <% viewInSource %><% crossRef %> 7. Robles-Diaz M., Lucena M.I., Kaplowitz N., Stephens C., Medina-Caliz I., Gonzalez-Jimenez A., et. al.: Use of Hy's law and a new composite algorithm to predict acute liver failure in patients with drug-induced liver injury. Gastroenterology 2014; 147: pp. 109-118 e105. <% viewInSource %><% crossRef %> 8. Swerdlow S.H.C.E., Harris N.L., et. al.: World Health Organization. WHO classification of tumours of haematopoietic and lymphoid tissues.2017. WHO <% viewInSource %> 9. Noronha V., Shafi N.Q., Obando J.A., Kummar S.: Primary non-Hodgkin's lymphoma of the liver. Crit Rev Oncol Hematol 2005; 53: pp. 199-207. <% viewInSource %><% crossRef %> 10. Swadley M.J., Deliu M., Mosunjac M.B., Gunthel C.J., Nguyen M.L., Hanley K.Z.: Primary and secondary hepatic lymphomas diagnosed by image-guided fine-needle aspiration: a retrospective study of clinical and cytomorphologic findings. Am J Clin Pathol 2014; 141: pp. 119-127. <% viewInSource %> 11. Rajesh S., Bansal K., Sureka B., Patidar Y., Bihari C., Arora A.: The imaging conundrum of hepatic lymphoma revisited. Insights Imaging 2015; 6: pp. 679-692. <% viewInSource %><% crossRef %> 12. Khan S.M., Cottrell B.J., Millward-Sadler G.H., Wright D.H.: T-cell-rich B-cell lymphoma presenting as liver disease. Histopathology 1993; 23: pp. 217-224. <% viewInSource %><% crossRef %> 13. Belliveau R.E., Abt A.B., Wiernik P.H.: Hepatic enzymes in Hodgkin's and non Hodgkin's lymphoma. Tumori 1979; 65: pp. 215-219. <% viewInSource %><% crossRef %> 14. Lee J.A., Jeong W.K., Min J.H., Kim J.: Primary hepatic lymphoma mimicking acute hepatitis. Clin Mol Hepatol 2013; 19: pp. 320-323. <% viewInSource %> 15. Biemer J.J.: Hepatic manifestations of lymphomas. Ann Clin Lab Sci 1984; 14: pp. 252-260. <% viewInSource %> 16. Rajwanshi A., Srinivas R., Upasana G.: Malignant small round cell tumors. J Cytol 2009; 26: pp. 1-10. <% viewInSource %><% crossRef %> 17. Yu L., Yan L.L., Yang S.J.: Sarcomatoid variant of ALK- anaplastic large cell lymphoma involving multiple lymph nodes and both lungs with production of proinflammatory cytokines: report of a case and review of literature. Int J Clin Exp Pathol 2014; 7: pp. 4806-4816. <% viewInSource %><% crossRef %> 18. Belhadj K., Reyes F., Farcet J.P., Tilly H., Bastard C., Angonin R., et. al.: Hepatosplenic gammadelta T-cell lymphoma is a rare clinicopathologic entity with poor outcome: report on a series of 21 patients. Blood 2003; 102: pp. 4261-4269. <% viewInSource %><% crossRef %> 19. Pizzi M., Covey S., Mathew S., Liu Y.C., Ruan J., Leonard J.P., et. al.: Hepatosplenic T-cell lymphoma mimicking acute myeloid leukemia. Clin Lymphoma, Myeloma & Leukemia 2016; 16: pp. e47-50. <% viewInSource %> 20. Yabe M., Medeiros L.J., Wang S.A., Tang G., Bueso-Ramos C.E., Jorgensen J.L.: Distinguishing between hepatosplenic T-cell lymphoma and gammadelta T-cell large granular lymphocytic leukemia: a clinicopathologic, immunophenotypic, and molecular analysis. Am J Surg Pathol 2017; 41: pp. 82-93. <% viewInSource %> 21. Shi Y., Wang E.: Hepatosplenic T-cell lymphoma: a clinicopathologic review with an emphasis on diagnostic differentiation from other T-cell/natural killer-cell neoplasms. Arch Pathol Lab Med 2015; 139: pp. 1173-1180. <% viewInSource %> 22. Qiu Z.Y., Tian G.Y., Zhang Z., Zhang Y.Q., Xu W., Li J.Y.: Large granular lymphocytosis after transplantation. Oncotarget 2017; 8: pp. 81697-81708. <% viewInSource %><% crossRef %> 23. Kondoh K., Morimoto M., Keino D., Oyama R., Nagae C., Ashikaga T., et. al.: T-cell large granular lymphocyte leukemia in a child with anemia and Crohn's disease. Pediatr Int 2013; 55: pp. 111-114. <% viewInSource %> 24. Jaffe E.S., Nicolae A., Pittaluga S.: Peripheral T-cell and NK-cell lymphomas in the WHO classification: pearls and pitfalls. Mod Pathol 2013; 26: pp. S71-S87. <% viewInSource %><% crossRef %> 25. Weisenburger D.D., Savage K.J., Harris N.L., Gascoyne R.D., Jaffe E.S., MacLennan K.A., et. al.: Peripheral T-cell lymphoma, not otherwise specified: a report of 340 cases from the International Peripheral T-cell Lymphoma Project. Blood 2011; 117: pp. 3402-3408. <% viewInSource %><% crossRef %> 26. Ekstrom-Smedby K.: Epidemiology and etiology of non-Hodgkin lymphoma--a review. Acta Oncol 2006; 45: pp. 258-271. <% viewInSource %><% crossRef %> 27. Gru A.A., Haverkos B.H., Freud A.G., Hastings J., Nowacki N.B., Barrionuevo C., et. al.: The epstein-barr virus (EBV) in T cell and NK cell lymphomas: time for a reassessment. Curr Hematol Malig Rep 2015; 10: pp. 456-467. <% viewInSource %><% crossRef %> 28. Yin C.C., Medeiros L.J., Abruzzo L.V., Jones D., Farhood A.I., Thomazy V.A.: EBV-associated B- and T-cell posttransplant lymphoproliferative disorders following primary EBV infection in a kidney transplant recipient. Am J Clin Pathol 2005; 123: pp. 222-228. <% viewInSource %> 29. Heavican T.B., Bouska A., Yu J., Lone W., Amador C., Gong Q., et. al.: Genetic drivers of oncogenic pathways in molecular subgroups of peripheral T-cell lymphoma. Blood 2019; 133: pp. 1664-1676. <% viewInSource %><% crossRef %> 30. Finalet Ferreiro J., Rouhigharabaei L., Urbankova H., van der Krogt J.A., Michaux L., Shetty S., et. al.: Integrative genomic and transcriptomic analysis identified candidate genes implicated in the pathogenesis of hepatosplenic T-cell lymphoma. PLoS One 2014; 9: <% viewInSource %> 31. Rajala H.L., Eldfors S., Kuusanmaki H., van Adrichem A.J., Olson T., Lagstrom S., et. al.: Discovery of somatic STAT5b mutations in large granular lymphocytic leukemia. Blood 2013; 121: pp. 4541-4550. <% viewInSource %><% crossRef %> 32. Andersson E.I., Rajala H.L., Eldfors S., Ellonen P., Olson T., Jerez A., et. al.: Novel somatic mutations in large granular lymphocytic leukemia affecting the STAT-pathway and T-cell activation. Blood Canc J 2013; 3: pp. e168. <% viewInSource %><% crossRef %> 33. Nicolae A., Xi L., Pittaluga S., Abdullaev Z., Pack S.D., Chen J., et. al.: Frequent STAT5B mutations in gammadelta hepatosplenic T-cell lymphomas. Leukemia 2014; 28: pp. 2244-2248. <% viewInSource %><% crossRef %> 34. Manso R., Gonzalez-Rincon J., Rodriguez-Justo M., Roncador G., Gomez S., Sanchez-Beato M., et. al.: Overlap at the molecular and immunohistochemical levels between angioimmunoblastic T-cell lymphoma and a subgroup of peripheral T-cell lymphomas without specific morphological features. Oncotarget 2018; 9: pp. 16124-16133. <% viewInSource %><% crossRef %> 35. Iqbal J., Weisenburger D.D., Chowdhury A., Tsai M.Y., Srivastava G., Greiner T.C., et. al.: Natural killer cell lymphoma shares strikingly similar molecular features with a group of non-hepatosplenic gammadelta T-cell lymphoma and is highly sensitive to a novel aurora kinase A inhibitor in vitro. Leukemia 2011; 25: pp. 348-358. <% viewInSource %><% crossRef %>