To evaluate the prognostic significance of clinicobiologic and pathological features in angioimmunoblastic T-cell lymphoma (AITL), 157 AITL patients were retrieved from the GELA LNH87-LNH93 randomized clinical trials. One hundred forty-seven patients received a cyclophosphamide, doxorubicin, vincristine, and prednisone (CHOP)–like regimen with intensified courses in half of them. Histologically, 41 cases were classified as “rich in large cells” and 116 as “classic” (including 19 rich in epithelioid cells, 14 rich in clear cells, and 4 with hyperplastic germinal centers). Sixty-two cases were scored for CD10 and CXCL13 expression according to the abundance of positive lymphoid cells. Median age was 62 years, with 81% advanced stage, 72% B symptoms, 65% anemia, 50% hypergammaglobulinemia, and 66% elevated LDH. Overall 7-year survival was 30%. In multivariate analysis, only male sex (P = .004), mediastinal lymphadenopathy (P = .041), and anemia (P = .042) adversely affected overall survival. Increase in large cells and high level of CD10 and CXCL13 did not affect survival. Intensive regimen did not improve survival. In conclusion, AITL is a morphologically heterogeneous T-cell lymphoma commonly expressing CXCL13 and CD10 and carrying few prognostic factors. It portends a poor prognosis even when treated intensively. However, AITL is not always lethal with 30% of patients alive at 7 years.

Angioimmunoblastic T-cell lymphoma (AITL) represents a distinct clinicopathological entity, among nodal peripheral T-cell lymphomas.1  It generally occurs in elderly patients presenting with generalized lymphadenopathy, hepatosplenomegaly, anemia, and hypergammaglobulinemia. Histologic features of AITL include partial or complete effacement of the lymph node architecture by a polymorphous infiltrate that is typically associated with a proliferation of follicular dendritic cells (FDCs) and a prominent arborization of high endothelial venules. The neoplastic cells display minimal cytologic atypia. They are small- to medium-sized cells typically with a clear cytoplasm and they usually aggregate in small clusters. They account for only a fraction of the infiltrate and are admixed with a reactive population of small lymphocytes, eosinophils, plasma cells, histiocytes, and large lymphoid, sometimes Reed-Sternberg–like B cells that are often infected by Epstein-Barr virus (EBV).2  AITL shows a morphologic spectrum and increase in T-cell immunoblasts may be observed.1,3  Although it has not been thoroughly investigated, it was suggested that the increase in T-cell immunoblasts would indicate transformation into a peripheral T-cell lymphoma, unspecified (PTCL/U).3  An increase in EBV-infected B cells may also occur, and, in rare cases, an overt diffuse large B-cell lymphoma develops.4-7  Recent data concerning the identity of the normal cellular counterpart of AITL are emerging. It is now believed that AITL derives from a follicular helper T-cell (TFH) subset.8-14  This subset of T cells would be located at the boundary between the mantle zone and the germinal center light zone and is supposed to provide help to germinal center B cells during their terminal differentiation.8,10  The tumor cells usually express CD4, CD10, Bcl6, and CXCL13, a phenotype that is unique among T-cell lymphomas.11-14 

AITL is rare accounting for approximately 2% of all non-Hodgkin lymphomas.15  Due to the rarity of the disease, there have been relatively little data concerning the impact of clinical, biologic, and morphologic features of angioimmunoblastic lymphadenopathy with dysproteinemia (AILD) and/or AITL on survival and outcome.16-23  Randomized clinical trials do not exist and few reports if any included an extended follow up. In this report, we attempted to identify the prognostic significance of different pathological, biologic, and clinical features after a long follow up of consecutive AITL patients treated with chemotherapy according to LNH87 and LNH93 protocols conducted by the Groupe d'Etude des Lymphomes de l'Adulte (GELA). In particular, since rare cases of B-cell lymphomas have been reported to arise after AILD or AITL4-7  and since it was suggested that the increase in T cells could evolve into a PTCL/U,3  we intended to study the prognostic relevance of the increase (> 10%) in large atypical cells.

Patient selection

Eligibility criteria for this study included patients with confirmed diagnosis of AITL after histopathological and immunohistochemical review. The 157 AITL patients are a subset of 6700 patients enrolled in the LNH87 and LNH93 GELA trials for intermediate- and high-grade lymphomas. Some of the clinical features have been previously reported in 68 patients.24 

Histologic analysis

At the time of enrollment in the LNH87 and LNH93 protocols, lymph node biopsies were reviewed by pathologists of the GELA group and initially classified according to the Working Formulation25  and updated Kiel classification,26  based on morphologic examination of slides stained with hematoxylin-eosin and Giemsa and on immunohistochemistry comprising at least CD20 and CD3ϵ. For the purpose of the present study, 4 expert hematopathologists reviewed all T-cell lymphoma cases and completed the phenotype in order to reclassify them according to the WHO classification.1  In AITL cases, the diagnosis was based on the presence of the following 5 criteria: partial or diffuse effacement of the nodal architecture, vascular proliferation with prominent arborization of high endothelial venules, extrafollicular meshwork of FDCs, atypical population of CD3+ T cells, and large CD20+ B cells.

To uncover the prognostic significance of the increase in large cells, cases were assigned to 2 major categories: “rich in large cells” (>10% large B and/or T cells) and “classic,” the latter including cases rich in clear cells, rich in epithelioid cells, and with hyperplastic germinal centers (Figure 1A-E).

Figure 1

Histopathologic spectrum of AITL and patterns of CXCL13 expression. Panels A-E illustrate the large spectrum of angioimmunoblastic lymphoma. Panels F,G illustrate CXCL13 immunostaining patterns: (A) classic-type, (B) rich in large cells, (C) with hyperplastic germinal centers, (D) rich in clear cells, (E) rich in epithelioid cells (hematoxylin eosin stain); (F) score 1, (G) score 2, and (H) score 3. Images were captured by a Zeiss microscope (Carl Zeiss, Heidelberg, Germany); original magnifications ×25 (C) and ×250 (A,B,D-H). The different morphologic subtypes and the level of CXCL13 expression did not influence survival.

Figure 1

Histopathologic spectrum of AITL and patterns of CXCL13 expression. Panels A-E illustrate the large spectrum of angioimmunoblastic lymphoma. Panels F,G illustrate CXCL13 immunostaining patterns: (A) classic-type, (B) rich in large cells, (C) with hyperplastic germinal centers, (D) rich in clear cells, (E) rich in epithelioid cells (hematoxylin eosin stain); (F) score 1, (G) score 2, and (H) score 3. Images were captured by a Zeiss microscope (Carl Zeiss, Heidelberg, Germany); original magnifications ×25 (C) and ×250 (A,B,D-H). The different morphologic subtypes and the level of CXCL13 expression did not influence survival.

Close modal

Immunohistochemical analysis

Immunohistochemistry was performed on deparaffinized tissue sections using an indirect immunoperoxidase method. After appropriate antigen retrieval, slides were stained for CD20, CD3ϵ, and as far as possible CNA42 and/or CD21 antigens (DakoCytomation, Glostrup, Denmark). CD4, CD2, CD5, and CD7 (DakoCytomation) were performed in 55, 45, 54, and 42 cases, respectively. One-hundred twenty-one cases were tested for the presence of EBV using in situ hybridization with probes specific for the EBV-encoded small RNA (EBER) sequences and/or antibodies to latent membrane protein-1 (LMP-1) (DakoCytomation). Finally, 62 cases, for which additional slides were available, were evaluated for CD10 (56C6; Novocastra, Newcastle, United Kingdom) and CXCL13 (R&D systems, Minneapolis, MN) expression. The abundance of positive lymphoid tumor cells was evaluated semiquantitatively according to a score varying from 0 to 3 as follows (Figure 1F-H): 0 indicates negative/very few positive lymphoid cells; 1, scattered positive lymphoid cells with no or one occasional aggregate; 2, many scattered positive lymphoid cells with more than one aggregate; and 3, sheets of positive lymphoid cells. For detection of CXCL13 expression, an amplification system was used as recently reported.13  Positive internal controls included polymorphonuclear leukocytes and residual germinal center B cells for CD10 and FDCs and TFHs for CXCL13. Cases without positive internal control were excluded from the analysis.

Images were captured with a Zeiss Axioskop2 microscope (Carl Zeiss, Heidelberg, Germany) and Neofluar 100×/0.1 NA optical lenses (Zeiss). Photographs were taken with a DP70 Olympus camera (Olympus, Tokyo, Japan). Image acquisition was performed with Olympus DP Controller 2002, and images were processed with Adobe Photoshop version 7.0 (Adobe Systems, San Jose, CA).

Staging

Patients were clinically staged according to the Ann Arbor classification. Initial investigations included a complete medical history and physical examination; computed tomography of the chest, abdomen, and pelvis; bone marrow biopsy; and biologic evaluation including hemoglobin level, Coombs test, lymphocyte and platelet counts, and gammaglobulin, LDH, albumin, and microglobulin levels (Table 1). Patients were randomized to treatment into one of the 4 groups of LNH87 and 7 groups of LNH93 protocols according to a score taking into account age and a number of prognostic factors of the age-adjusted international prognostic index (aaIPI) score.27  Patients were required to sign an informed consent approved by Hôpital Saint Louis (Paris, France) institutional review board in accordance with the Declaration of Helsinki. They were excluded from the trial if they had positive serologic tests for HIV or human T-lymphotropic virus 1 (HTLV1).

Table 1

Univariate analysis: survival estimates according to patients' characteristics

CharacteristicNo.%7-y OS, %P7-y EFS, %P
Sex    .004  .094 
    Male 95/157 60 23 — 21 — 
    Female 62/157 40 39 — 24 — 
Age*    .071  .114 
    Not older than 60 y 72/157 — 21 — 17 — 
    Older than 60 y 85/157 — 36 — 27 — 
Performance status    .721  .613 
    0 to 1 79/157 50 27 — 17 — 
    More than 1 78/157 50 31 — 27  
B symptoms 112/155 72 27 .125 22 .276 
Ann Arbor stage    .425  .859 
    I to II 30/156 19 32 — 14 — 
    III to IV 126/156 81 28 — 24 — 
Bulky 10 cm or more 27/102 26 22 .130 14 .282 
No. of extranodal sites    .094  .106 
    0 to 1 75/139 54 34 — 28 — 
    More than 1 64/139 46 28 — 20 — 
Extranodal involvement       
    Liver 38/154 25 22 .130 19 .378 
    Spleen 86/155 55 26 .143 21 .490 
    Skin 46/154 30 25 .266 19 .389 
    Lung 15/153 10 18 .546 20 .858 
Bone marrow 71/151 47 29 .775 23 .813 
Effusion/edema/ascites 16/62 26 25 .895 19 .525 
Polyarthritis/arthralgia 12/76 16 25 .810 25 .263 
Skin rash 36/82 44 26 .598 19 .747 
IPI score    .799  .737 
    0 to 1 15/132 11 24 — 14 — 
    2 to 3 67/132 51 26 — 20 — 
    4 to 5 50/132 38 38 — 32 — 
PIT score    .106  .016 
    0 to 1 38/144 26 26 — 18 — 
    2 50/144 35 15 — — 
    3 to 4 56/144 39 38 — 34 — 
Anemia, Hb level no higher than 120 g/L 101/155 65 26 .046* 22 .158 
Positive Coombs test 30/92 33 39 .299 35 .188 
Lymphopenia no higher than 0.7 × 109/L 77/156 49 29 .799 19 .930 
Thrombocytopenia no higher than 150 × 109/L 33/121 20 26 .219 18 .363 
Hypereosinophilia more than 0.5 × 109/L 12/38 32 33 .324 25 .375 
Hypergammaglobulinemia more than 12 g/L 73/146 50 36 .260 26 .294 
Increased serum LDH level 98/149 66 29 .794 23 .329 
Hypoalbuminemia less than 35 g/L 71/143 50 28 .160 24 .227 
Serum β2 microglobulin level more than 250 nM 71/107 66 31 .911 22 .938 
CharacteristicNo.%7-y OS, %P7-y EFS, %P
Sex    .004  .094 
    Male 95/157 60 23 — 21 — 
    Female 62/157 40 39 — 24 — 
Age*    .071  .114 
    Not older than 60 y 72/157 — 21 — 17 — 
    Older than 60 y 85/157 — 36 — 27 — 
Performance status    .721  .613 
    0 to 1 79/157 50 27 — 17 — 
    More than 1 78/157 50 31 — 27  
B symptoms 112/155 72 27 .125 22 .276 
Ann Arbor stage    .425  .859 
    I to II 30/156 19 32 — 14 — 
    III to IV 126/156 81 28 — 24 — 
Bulky 10 cm or more 27/102 26 22 .130 14 .282 
No. of extranodal sites    .094  .106 
    0 to 1 75/139 54 34 — 28 — 
    More than 1 64/139 46 28 — 20 — 
Extranodal involvement       
    Liver 38/154 25 22 .130 19 .378 
    Spleen 86/155 55 26 .143 21 .490 
    Skin 46/154 30 25 .266 19 .389 
    Lung 15/153 10 18 .546 20 .858 
Bone marrow 71/151 47 29 .775 23 .813 
Effusion/edema/ascites 16/62 26 25 .895 19 .525 
Polyarthritis/arthralgia 12/76 16 25 .810 25 .263 
Skin rash 36/82 44 26 .598 19 .747 
IPI score    .799  .737 
    0 to 1 15/132 11 24 — 14 — 
    2 to 3 67/132 51 26 — 20 — 
    4 to 5 50/132 38 38 — 32 — 
PIT score    .106  .016 
    0 to 1 38/144 26 26 — 18 — 
    2 50/144 35 15 — — 
    3 to 4 56/144 39 38 — 34 — 
Anemia, Hb level no higher than 120 g/L 101/155 65 26 .046* 22 .158 
Positive Coombs test 30/92 33 39 .299 35 .188 
Lymphopenia no higher than 0.7 × 109/L 77/156 49 29 .799 19 .930 
Thrombocytopenia no higher than 150 × 109/L 33/121 20 26 .219 18 .363 
Hypereosinophilia more than 0.5 × 109/L 12/38 32 33 .324 25 .375 
Hypergammaglobulinemia more than 12 g/L 73/146 50 36 .260 26 .294 
Increased serum LDH level 98/149 66 29 .794 23 .329 
Hypoalbuminemia less than 35 g/L 71/143 50 28 .160 24 .227 
Serum β2 microglobulin level more than 250 nM 71/107 66 31 .911 22 .938 

OS indicates overall survival; EFS, event-free survival; IPI, International Prognostic Index; and PIT, prognostic index for peripheral T-cell lymphoma, unspecified.

*

Median patient age was 62 years (range, 20-89 years).

Median hemoglobin level was 100 g/L (range, 30-120 g/L).

Treatment

The results of both LNH87 and LNH93 protocols have in large part been published.28-36  Briefly, for younger patients, the reference arm was 3 to 4 courses of dose-intensive doxorubicin, cyclophosphamide, vindesine, bleomycin, and prednisone (ACVBP) followed by sequential consolidation36  (50 patients), and the experimental arm consisted of the following: 8 cycles of methotrexate, bleomycin, doxorubicin, cyclophosphamide, vincristine, and dexamethasone (mBACOD) in patients with no adverse factor28  (LNH 87-1, 11 patients); 4 cycles of ACVBP-like with a second randomization between sequential consolidation and high-dose chemotherapy (CBV) with stem cell rescue for patients younger than 56 years with at least one adverse factor29  (LNH 87-2, 7 patients); 4 alternating induction cycles of teniposide, ifosfamide, mitoxantrone, courses of etoposide, ifosfamide, and mitoxantrone (VIM) and doxorubicin, cyclophosphamide, vindesine, and methotrexate (ACVM) in patients between 56 and 69 years of age with at least one adverse factor30  (LNH 87-3, 14 patients); 3 cycles of cyclophosphamide, doxorubicin, vincristine, and prednisone (CHOP) with involved field radiotherapy for patients with 0 aaIPI factors31  (LNH 93-1, 2 patients); 4 cycles of ACVBP-like followed by a modified consolidation for patients with 1 aaIPI factor (LNH 93-2, 2 patients); and 4 cycles of ACVBP-like followed by high-dose chemotherapy with peripheral blood stem cell rescue for patients younger than 60 years with 2 or more factors32  (LNH 93-3, 8 patients).

For elderly patients, the arms were as follows: 4 cycles of CHOP with or without additional involved field radiotherapy for patients with 0 aaIPI factors33  (LNH 93-4, 4 patients); 8 cycles of CHOP or ACVBP for patients younger than 70 years with 1 to 3 aaIPI factors34  (LNH 93-5, 23 patients); 6 cycles of CHOP-like or reduced CHOP for patients 70 years or older and PS less than 235  (LNH 87-4 and LNH 93-6, 29 patients); oral chemotherapy with etoposide, chlorambucil, and prednisone for patients 70 years or older with a PS 2 or more (LNH 93-7, 7 patients).

Primary treatment failure included change to another therapy including chemotherapy and/or autologous or allogeneic treatment.

Response to treatment was considered as complete response (CR), unconfirmed complete response (CRu), partial response, or failure according to the International Workshop criteria.37 

Statistical analysis

Patients' characteristics and remission rates were compared using chi-square test. Event-free survival (EFS) was defined as the time interval between randomization to primary treatment failure, relapse, and death from any cause or last follow up. Overall survival (OS) was defined as the time interval between randomization to last follow up or death from any cause. Estimates of survival were calculated according to the Kaplan-Meier method38  and were compared using the log-rank test.39  Cox proportional hazards regression analysis with OS and EFS as the dependent variables was used to detect potential independent prognostic factors. Differences were considered statistically significant when the 2-sided P value was less than .05. All statistical analyses were performed using the Statistical Application System software (SAS, version 913; SAS Institute, Cary, NC).

Clinical and biologic characteristics of AITL patients at presentation

One-hundred fifty-seven patients were included in the study; they were treated between April 1987 and October 1999. Table 1 summarizes their main clinical and biologic characteristics. The median age was 62 years (range: 20-89 years). Most patients (126/156; 81%) presented with an advanced-stage III-IV, and 21% (29/139) had more than 2 extranodal involved sites. B symptoms were observed in 72% (112/155). Laboratory investigations showed the presence of anemia in 65% (101/155), a positive Coombs test in 33% (30/92), hypergammaglobulinemia more than 12 g/L in 50% (73/146), and elevated serum LDH level in 66% (98/149) of patients. Sixty percent (58/97) and 38% (24/63) of anemic patients had bone marrow involvement and a positive Coombs test, respectively. The international prognostic index (IPI) score was more than 1 in 89% (117/132) of patients, and the prognostic index for PTCL/U (PIT) score40  (scoring system including age > 60 years, performance status [PS] ≥ 2, LDH ≥ normal, and bone marrow involvement) was more than 2 in 39% (56/144) of patients.

Pathological findings

By definition, all cases displayed FDC hyperplasia demonstrated by morphology and/or CNA42/CD21 immunostaining, as well as a variable proportion of CD20 large B cells within an atypical T-cell population. Although clonality could be assessed in only a minority of cases (data not shown), the review process retained cases that disclosed typical histopathological and immunophenotypic features for AITL, and that would not be mistaken for a reactive process, with all but 4 cases showing diffuse effacement of the normal architecture. According to their cytologic aspect, 41 cases were classified as rich in large cells and 116 as classic (Table 2) including 19 rich in epithelioid cells, 14 rich in clear cells, and 4 with hyperplastic germinal centers. As indicated in Table 2, CD3 was positive in 100%, CD4 in 95%, CD2 in 91%, CD5 in 85%, and CD7 in 50% of tested cases. A total of 111 cases were studied for EBV using EBER in situ hybridization. Among the 103 cases with interpretable results, 25 (24%) were negative for EBV, whereas 78 (76%) contained a variable number of EBER-positive cells. In the majority of cases, EBER-positive cells were scattered, with the exception of 19 cases in which there were sheets of EBV-positive cells. Ten additional cases that were not investigated by EBER in situ hybridization disclosed scattered blasts positive for LMP-1. Among the 52 cases interpretable for CD10 expression, 71% disclosed aggregates or sheets of CD10+ cells with atypical features (scored 2 + 3). Scattered CD10+ lymphoid cells (score 1)—the neoplastic nature of which could not be determined—were observed in 12% and CD10 was negative (score 0) in 17% of our cases. Forty-five of 62 sections were adequate for CXCL13 interpretation. CXCL13 staining was found, as a dot reinforcement of the Golgi area, in aggregates or sheets of atypical cells (score 2 + 3) in 73% of cases, and in scattered lymphoid cells (score 1) in the remaining 27%. Overall, at least one marker was scored positive in 86% of the interpretable cases and both markers were negative in 14% of them.

Table 2

Univariate analysis: survival estimates according to histopathologic and phenotypic* characteristics of AITL patients

Pathological findingNo.%7-y OS, %P7-y EFS, %P
Classic 116 74 24 .444 19 .350 
    Rich in epithelioid cells 19 13 — — — — 
    Rich in clear cells 14 — — — — 
    With HGC 04 03 — — — — 
Rich in large cells 41 26 41 — 31 — 
CD10 52   .634  .870 
    Score 0 09 17 27 — 14 — 
    Score 1 06 12 67 — 33 — 
    Score 2 25 48 24 — 29 — 
    Score 3 12 23 17 — — 
CXCL13 45   .865  .724 
    Score 0 — — — — 
    Score 1 12 27 33 — 17 — 
    Score 2 24 53 23 — 20 — 
    Score 3 09 20 33 — — 
EBV (ISH) 103   .557  .386 
    Negative 25 24 15 — — 
    Positive 78 76 30 — 21 — 
Pathological findingNo.%7-y OS, %P7-y EFS, %P
Classic 116 74 24 .444 19 .350 
    Rich in epithelioid cells 19 13 — — — — 
    Rich in clear cells 14 — — — — 
    With HGC 04 03 — — — — 
Rich in large cells 41 26 41 — 31 — 
CD10 52   .634  .870 
    Score 0 09 17 27 — 14 — 
    Score 1 06 12 67 — 33 — 
    Score 2 25 48 24 — 29 — 
    Score 3 12 23 17 — — 
CXCL13 45   .865  .724 
    Score 0 — — — — 
    Score 1 12 27 33 — 17 — 
    Score 2 24 53 23 — 20 — 
    Score 3 09 20 33 — — 
EBV (ISH) 103   .557  .386 
    Negative 25 24 15 — — 
    Positive 78 76 30 — 21 — 

HGC indicates hyperplastic germinal centers; OS, overall survival; EFS, event-free survival; and ISH, in situ hybridization.

*

CD3, CD4, CD2, CD5, and CD7 were positive in tumor cells in 100% (157/157), 95% (52/55), 91% (41/45), 85% (46/54), and 50% (21/42), respectively.

Clinical outcome and prognostic parameters in the overall population

All but 10 patients received an anthracycline-based chemotherapy. CR/CRu was achieved in 46% of patients after induction therapy. No difference could be observed between the different chemotherapeutic arms of the randomized studies including stem cell transplantation. An attempt was made to put together all the intensive regimens versus the conventional ones, and it was not possible to discriminate a better or a worse outcome according to treatment.

Thirty-one (20%) of 156 patients developed severe infection and 82 (53%) of 156 developed severe neutropenia during therapy (≥ grade 3 toxicity). Sixty-six (42%) of 156 patients relapsed or progressed under treatment. Forty-five of them received salvage chemotherapy and 12 of them received autologous or allogeneic treatment.

Follow-up could be assessed in 156 patients. The median follow-up was 68 months (range: 3.77-161.47 months). The 2-, 5-, and 7-year OS rates were 51% (CI: 42.85-58.77), 33% (CI: 25.63-40.95), and 29% (CI:, 21.19-36.73) respectively (Figure 2A), reaching an apparent plateau level around 6 years. The 2-, 5-, and 7-year EFS rates were 38% (CI: 30.66-46.06), 29% (CI: 21.54-36.02), and 23% (CI: 15.57-29.94), respectively (Figure 2B). Overall, there were 107 deaths. Patients died from lymphoma (57%), treatment toxicity (7%), infection (7%), lymphoma and toxicity (7%), lymphoma and infection (5%), toxicity and infection (4%), lymphoma and toxicity and infection (1%), other cancer (4%), or from other or unknown reason (8%). Of note, 2 of the 7 patients older than 70 years remained alive and free of disease after 5 and 10 years of follow-up.

Figure 2

Survival of the 156 patients with angioimmunoblastic T-cell lymphoma. Overall survival (A) and event-free survival (B).

Figure 2

Survival of the 156 patients with angioimmunoblastic T-cell lymphoma. Overall survival (A) and event-free survival (B).

Close modal

On univariate analysis (Table 1), male sex (P = .004), mediastinal lymphadenopathy (P = .013), and anemia (P = .046) turned out to be poor prognostic factors for OS. Anemia correlated with bone marrow involvement (P < .001) and showed a trend toward an association with a positive Coombs test (P = .098). Bone marrow involvement and positive Coombs test did not influence survival, however. Mediastinal lymphadenopathy (P = .026) and PIT score (P = .016) influenced EFS. However, on careful examination of PIT survival curves according to score, patients with scores 3 to 4 were doing better than those with scores 0 to 1 and 2. The same applied to age (P = .071), where patients older than 60 years were doing better than younger ones with regard to OS. Since it is likely that these unexpected findings represent a false positive selection (ie, alpha = 0.05) due to sampling fluctuations among the numerous parameters tested in our study, we decided not to include age and PIT in the multivariate analysis. On multivariate analysis, male sex (P = .004), mediastinal lymphadenopathy (P = .041), and anemia (P = .042) remained as independent prognostic factors for OS (Table 3). Mediastinal lymphadenopathy (P = .053) and male sex (P = .067) were at the limits of statistical significance for EFS. OS and EFS differed significantly in favor of patients who developed CR/CRu (P < .001). IPI was not predictive of survival (Figure 3A). The different histologic subtypes, the presence of EBV-positive cells, as well as CD10 and CXCL13 scoring did not have any influence on survival (Table 2). Of note, cases with sheets of EBV-positive cells paralleled those that were rich in large cells (P = .0219). The impact of the clinicopathological prognostic index (including age, LDH > normal, nonambulatory PS, Ki67 ≥ 80%) described by Went et al for PTCL/U patients could not be assessed in our AITL series.41 

Table 3

Parameters influencing overall survival of AITL patients on multivariate analysis

ParameterPRelative risk95% CI low95% CI high
Male sex .004 1.856 1.221 2.819 
Anemia, Hb no higher than 120 g/dL .042 1.570 1.017 2.425 
Mediastinal lymphadenopathy .041 1.535 1.017 2.316 
ParameterPRelative risk95% CI low95% CI high
Male sex .004 1.856 1.221 2.819 
Anemia, Hb no higher than 120 g/dL .042 1.570 1.017 2.425 
Mediastinal lymphadenopathy .041 1.535 1.017 2.316 
Figure 3

Overall survival of angioimmunoblastic T-cell lymphoma patients. Overall survival figures according to IPI (A) and PIT (B) scores are shown.

Figure 3

Overall survival of angioimmunoblastic T-cell lymphoma patients. Overall survival figures according to IPI (A) and PIT (B) scores are shown.

Close modal

Comparison between the 2 histologic categories: classic and rich in large cells

Overall, the 2 populations classic and rich in large cells were not statistically different in terms of clinical features, laboratory findings, and treatment response (P = .676) except for a higher frequency of elevated serum LDH (P = .027) and β2 microglobulin levels (P = .046) within the group rich in large cells. The distribution of OS and EFS curves was not statistically different between these 2 groups either.

To the best of our knowledge, the present series of AITL patients is the largest reported so far in which all patients were included in prospective randomized clinical trials. In this series, we further extend the peculiar clinical and biologic findings of AITL. We found very few prognostic factors for this disease, and we confirm its poor prognosis despite a first-line intensive CHOP-like chemotherapy with a curative intent in most patients. We also emphasize its morphologic heterogeneity as well as the common expression of CD10 and CXCL13 determined on routinely fixed samples.

From a clinical point of view and in keeping with previous reports,16-23,42-44  we observed that AITL is a disease of the elderly presenting with systemic manifestations and features known to be poor prognostic factors for B-cell lymphomas: 54% of our patients were older than 60 years, 50% had an altered PS, 81% presented with stage III-IV disease, 46% had more than one extranodal site involvement, and 66% had an elevated LDH level. The clinical symptoms and biologic signs are not specific of AITL but their association appears to be very suggestive of the disease. Notably, skin rash (44%), arthritis/arthralgia (16%), pleural effusion/ascites/edema (26%), eosinophilia (32%), hypergammaglobulinemia (50%), and positive Coombs test (33%) appear to represent distinctive manifestations of AITL.

A small number of studies sought to identify prognostic factors in AITL/AILD, yielding controversial results.16-23  Their results might be hampered by the relatively limited number of patients and/or the heterogeneous therapies including prednisolone alone in a proportion of these patients. In the current study, we could not find any pertinent prognostic factor for this disease besides male sex (P = .004), mediastinal lymphadenopathy (P = .041), and anemia (P = .042) that adversely affected OS on multivariate analysis. The impact of IPI score was rarely defined in the literature for AITL, and in agreement with Lee et al,22  it was curiously not predictive of survival at all. And in contrast to what has been originally reported for PTCL/U,40  a high PIT score was not associated with a worse outcome in our AITL series. Even when PIT was included in the multivariate analysis, it did not significantly influence survival (data not shown). Moreover, in the present study where patients received more intensive chemotherapy overall compared with previous reports, it was not possible to demonstrate any survival benefit for one treatment arm over the other even for those submitted to consolidation with autologous stem cell transplantation. This latter subset analysis is in agreement with what had already been reported by Mounier et al on non–anaplastic T-cell lymphomas and front-line autologous stem cell transplantation.45  ACVBP regimen was not superior to standard CHOP and mBACOD regimens with respect to OS. These data should be interpreted with caution however due to the relatively low number of cases assigned to each treatment arm and obviously require further confirmation in prospective randomized trials. In a retrospective study by Schetelig et al, including 29 AITL patients treated with high-dose chemotherapy followed by autologous stem cell transplantation, CR increased from 45% before high-dose chemotherapy to 76% after, with a 44% and 37% 5-year OS and progression-free survival, respectively.46  Interestingly, similar results have been recently reported in a large series of AITL patients treated with high-dose chemotherapy and autologous stem cell transplantation, with a 59% OS and 42% progression-free survival at 4 years after transplantation.47 

From a pathologic point of view, the present series describes the morphologic heterogeneity of AITL, which could be a source of misdiagnosis to poorly experienced pathologists, especially concerning cases with hyperplastic germinal centers and very rich in epithelioid cells. The former can be confused with a hyperplastic reactive disorder and the latter with Lennert lymphoma or Hodgkin lymphoma. This is reflected by the large error rate (50%) in the referred cases described by Attygalle et al11  and by our very low recruitment of cases with hyperplastic germinal centers, a pattern that has recently received attention since it was described in 1998.48 

Few reports have tried to identify histologic prognostic features, but none of them would be of proven clinical value.18,20,22,49  The current study specifically attempted to investigate the potential prognostic relevance for the increase in large cells, but it failed to show any significance on outcome for this increase. In this respect, the presence of sheets of EBV-infected cells did not affect survival in this series of patients treated with polychemotherapy. Moreover, the number of CD10+ and CXCL13+ atypical cells, which is supposed to represent the neoplastic counterpart, did not show any influence on survival. The different morphologic subtypes, and in particular the abundance of clear cells, were not predictive of survival either.

It has been recently shown that CXCL13, a chemokine critically involved in B-cell migration into germinal centers, was highly up-regulated in the TFH subset.50-52  Subsequently, we and others have shown that CXCL13 was expressed by neoplastic cells of most AITLs and that CD10 as well as TFH markers such as CXCL13 belong to the molecular gene signature of AITL.9,13,14  CD10 and CXCL13 were proposed as sensitive immunohistochemical markers in AITL.11,13,14  This finding was confirmed in the present study where at least one marker stained clusters of atypical lymphocytes in the majority (86%) of cases. However, in the few cases (14%) where it was difficult to assign the neoplastic nature for the rare scattered stained cells on purely morphologic grounds, both markers were considered negative. In such circumstances, the diagnosis of AITL should rely on a combination of classical morphologic criteria together with clinical and biologic features.

It was hypothesized that the clinical effects of AITL are due to marked dysregulation of the immune system rather to direct complications of tumor growth.11,14,53  This hypothesis can be supported by our findings that high IPI and PIT scores—which are supposed to reflect increase in tumor burden—did not correlate with a worse outcome, and by the fact that eosinophilia and hypergammaglobulinemia are more often encountered in AITL than in other subtypes of T-cell lymphomas.24  Moreover, a recent gene expression study of highly enriched tumor samples of AITL showed that nearly 90% of AITL signature was contributed by nonneoplastic cells.9  The AITL profiling portrait included genes involved in the humoral immune response and in the modulation of vasculogenesis and extracellular matrix and genes encoding various chemokines mediating the recruitment of inflammatory cells. These findings are in line with the peculiar histologic features of AITL where tumor cells are greatly outnumbered by the surrounding reactive cells and found in intimate contact with the expanding meshwork of FDCs. As a whole, these results support the idea considering AITL as an immunologically functional disease in which the clinical behavior is determined by the resultant cross talk between the malignant cells and the immunologic micro-environment. An improved understanding of the interactions between neoplastic cells and microenvironment in AITL would offer the possibility of identifying new targets for rationale design of future treatments. In this respect, immunomodulatory approaches54-57  deserve some consideration in future prospective randomized trials.

In conclusion, despite various intensive regimens with an anthracycline-based chemotherapy, AITL, compared with other non-Hodgkin lymphomas, pursues an aggressive clinical course. AITL does not present any pertinent prognostic factor besides, naturally, the achievement of a complete response to therapy. However, after a long follow-up, we observed a trend for a plateau level reaching a survival probability of approximately 30% at 6 years, thus providing a threshold for evaluation of new drugs or strategies including allograft with reduced intensity regimen. A longer follow-up is needed to determine whether these patients are eventually cured of their disease.

The online version of this article contains a data supplement.

This study was presented at the 48th annual meeting of the American Society of Hematology, Orlando, FL, December 2006.58 

The publication costs of this article were defrayed in part by page charge payment. Therefore, and solely to indicate this fact, this article is hereby marked “advertisement” in accordance with 18 USC section 1734.

We thank Sylvie Sousa, Chafika Coppeaux, Nicolas Nio, and Marion Fournier for their skillful assistance in data management and Nadine Martin-Garcia for her technical assistance.

Contribution: C.G. and P.G. designed the study; N. Mourad, C.G., and P.G. wrote the paper; N. Mounier performed the statistical analysis; N. Mourad, N. Mounier, J.B., C.G., and P.G. controlled and analyzed the data; A.F., C.J.L.M.M., J.D., and P.G. reviewed the pathological material; J.-F.E. performed part of the EBER studies; N. Mounier, E.R., A.D., R.B., A.B., C.H., B.C., and C.G. participated in data collection; all authors checked the final version of the paper.

A complete list of the members of the Groupe d'Etude des Lymphomes de l'Adulte appears in Document S1, available on the Blood website (see the Supplemental Materials link at the top of the online article).

Conflict-of-interest disclosure: The authors declare no competing financial interests.

Correspondence: Philippe Gaulard, Département de Pathologie et Inserm U841, Hôpital Henri Mondor, Créteil–F-94000, France; e-mail: philippe.gaulard@hmn.aphp.fr.

1
Jaffe
 
ES
Harris
 
NL
Stein
 
H
Vardiman
 
JW
World Health Organization classification of tumours: Pathology and genetics, tumours of haematopoietic and lymphoid tissues.
2001
Lyon, France
IARC Press
2
Weiss
 
LM
Jaffe
 
ES
Liu
 
XF
Chen
 
YY
Shibata
 
D
Medeiros
 
LJ
Detection and localization of Epstein-Barr viral genomes in angioimmunoblastic lymphadenopathy and angioimmunoblastic lymphadenopathy-like lymphoma.
Blood
1992
, vol. 
79
 (pg. 
1789
-
1795
)
3
Feller
 
AC
Diebold
 
J
Histopathology of Nodal and Extranodal Non-Hodgkin's Lymphomas.
2004
Berlin Heidelberg, Germany
Springer-Verlag
4
Abruzzo
 
LV
Schmidt
 
K
Weiss
 
LM
, et al. 
B-cell lymphoma after angioimmunoblastic lymphadenopathy: a case with oligoclonal gene rearrangements associated with Epstein-Barr virus.
Blood
1993
, vol. 
82
 (pg. 
241
-
246
)
5
Matsue
 
K
Itoh
 
M
Tsukuda
 
K
Kokubo
 
T
Hirose
 
Y
Development of Epstein-Barr virus-associated B cell lymphoma after intensive treatment of patients with angioimmunoblastic lymphadenopathy with dysproteinemia.
Int J Hematol
1998
, vol. 
67
 (pg. 
319
-
329
)
6
Lome-Maldonado
 
C
Canioni
 
D
Hermine
 
O
, et al. 
Angio-immunoblastic T cell lymphoma (AILD-TL) rich in large B cells and associated with Epstein-Barr virus infection: a different subtype of AILD-TL?
Leukemia
2002
, vol. 
16
 (pg. 
2134
-
2141
)
7
Attygalle
 
AD
Kyriakou
 
C
Dupuis
 
J
, et al. 
Histologic evolution of angioimmunoblastic T-cell lymphoma in consecutive biopsies: clinical correlation and insights into natural history and disease progression.
Am J Surg Pathol
2007
, vol. 
31
 (pg. 
1077
-
1088
)
8
Krenacs
 
L
Schaerli
 
P
Kis
 
G
Bagdi
 
E
Phenotype of neoplastic cells in angioimmunoblastic T-cell lymphoma is consistent with activated follicular B helper T cells.
Blood
2006
, vol. 
108
 (pg. 
1110
-
1111
)
9
de Leval
 
L
Rickman
 
DS
Thielen
 
C
, et al. 
The gene expression profile of nodal peripheral T-cell lymphoma demonstrates a molecular link between angioimmunoblastic T-cell lymphoma (AITL) and follicular helper T cells (TFH).
Blood
2007
, vol. 
109
 (pg. 
4952
-
4963
)
10
Vinuesa
 
CG
Tangye
 
SG
Moser
 
B
Mackay
 
CR
Follicular B helper T cells in antibody response and autoimmunity.
Nat Rev Immunol
2005
, vol. 
5
 (pg. 
853
-
865
)
11
Attygalle
 
A
Al Jehani
 
R
Diss
 
TC
, et al. 
Neoplastic T cells in angioimmunoblastic T-cell lymphoma express CD10.
Blood
2002
, vol. 
99
 (pg. 
627
-
633
)
12
Dogan
 
A
Attygalle
 
AD
Kyriakou
 
C
Angioimunoblastic T-cell lymphoma.
Br J Haematol
2003
, vol. 
121
 (pg. 
681
-
691
)
13
Dupuis
 
J
Boye
 
K
Martin
 
N
, et al. 
Expression of CXCL13 by neoplastic cells in Angioimmunoblastic T cell-lymphoma (AITL): a new diagnostic marker providing evidence that AITL derives from follicular helper T cells.
Am J Surg Pathol
2006
, vol. 
30
 (pg. 
490
-
494
)
14
Grogg
 
KL
Attygalle
 
AD
Macon
 
WR
Remstein
 
ED
Kurtin
 
PJ
Dogan
 
A
Angioimmunoblastic T-cell lymphoma: a neoplasm of germinal-center T-helper cells?
Blood
2005
, vol. 
106
 (pg. 
1501
-
1502
)
15
A clinical evaluation of the International Lymphoma Study Group classification of non-Hodgkin's lymphoma: The Non-Hodgkin's Lymphoma Classification Project.
Blood
1997
, vol. 
89
 (pg. 
3909
-
3918
)
16
Pangalis
 
GA
Moran
 
EM
Nathwani
 
BN
Zelman
 
RJ
Kim
 
H
Rappaport
 
H
Angioimmunoblastic lymphadenopathy: long term follow up study.
Cancer
1983
, vol. 
52
 (pg. 
318
-
321
)
17
Archimbaud
 
E
Coiffier
 
B
Bryon
 
PA
Vasselon
 
C
Brizard
 
CP
Viala
 
JJ
Prognostic factors in angioimmunoblastic lymphadenopathy.
Cancer
1987
, vol. 
59
 (pg. 
208
-
212
)
18
Aozasa
 
K
Ohsawa
 
M
Fujita
 
MQ
, et al. 
Angioimmunoblastic lymphadenopathy: review of 44 patients with emphasis on prognostic behavior.
Cancer
1989
, vol. 
63
 (pg. 
1625
-
1629
)
19
Siegert
 
W
Nerl
 
C
Agthe
 
A
, et al. 
Angioimmunoblastic lymphadenopathy (AILD)-type T-cell lymphoma: prognostic impact of clinical observations and laboratory findings at presentation: The Kiel Lymphoma Study Group.
Ann Oncol
1995
, vol. 
6
 (pg. 
659
-
664
)
20
Ch'ang
 
HJ
Su
 
IJ
Chen
 
CH
, et al. 
Angioimmunoblastic lymphadenopathy with dysproteinemia: lack of prognostic value of clear cell morphology.
Oncology
1997
, vol. 
54
 (pg. 
193
-
198
)
21
Pautier
 
P
Devidas
 
A
Delmer
 
A
, et al. 
Angioimmunoblastic-like T-cell non Hodgkin's lymphoma: outcome after chemotherapy in 33 patients and review of the literature.
Leuk Lymphoma
1999
, vol. 
32
 (pg. 
545
-
552
)
22
Lee
 
SS
Rudiger
 
T
Odenwald Roth
 
S
Starostik
 
P
Muller-Hermelink
 
HK
Angioimmunoblastic T cell lymphoma is derived from mature T-helper cells with varying expression and loss of detectable CD4.
Int J Cancer
2003
, vol. 
103
 (pg. 
12
-
20
)
23
Lachenal
 
F
Berger
 
F
Ghesquières
 
H
, et al. 
Angioimmunoblastic T-cell lymphoma: clinical and laboratory features at diagnosis in 77 patients.
Medicine
2007
, vol. 
86
 (pg. 
282
-
292
)
24
Gisselbrecht
 
C
Gaulard
 
P
Lepage
 
E
, et al. 
Prognostic significance of T-cell phenotype in aggressive non-Hodgkin's lymphomas: Groupe d'Etude des Lymphomes de l'Adulte (GELA).
Blood
1998
, vol. 
92
 (pg. 
76
-
82
)
25
National Cancer Institute sponsored study of classifications of non-Hodgkin's lymphomas: summary and description of a working formulation for clinical usage: The Non-Hodgkin's Lymphoma Pathologic Classification Project.
Cancer
1982
, vol. 
49
 (pg. 
2112
-
2135
)
26
Stansfeld
 
AG
Diebold
 
J
Noel
 
H
, et al. 
Updated Kiel classification for lymphomas.
Lancet
1998
, vol. 
1
 (pg. 
292
-
293
)
27
A predictive model for aggressive non-Hodgkin's lymphoma: The International Non-Hodgkin's Lymphoma Prognostic Factors Project.
N Engl J Med
1993
, vol. 
329
 (pg. 
987
-
994
)
28
Tilly
 
H
Mounier
 
N
Lederlin
 
P
, et al. 
Randomized comparison of ACVBP and m-BACOD in the treatment of patients with low-risk aggressive lymphoma: the LNH87–1 study: Groupe d'Etude des Lymphomes de l'Adulte.
J Clin Oncol
2000
, vol. 
18
 (pg. 
1309
-
1315
)
29
Haioun
 
C
Lepage
 
E
Gisselbrecht
 
C
, et al. 
Survival benefit of high-dose therapy in poor-risk aggressive non-Hodgkin's lymphoma: final analysis of the prospective LNH87–2 protocol: a Groupe d'Etude des Lymphomes de l'Adulte study.
J Clin Oncol
2000
, vol. 
18
 (pg. 
3025
-
3030
)
30
Bosly
 
A
Lepage
 
E
Coiffier
 
B
, et al. 
Outcome is not improved by the use of alternating chemotherapy in elderly patients with aggressive lymphoma.
Hematol J
2001
, vol. 
2
 (pg. 
279
-
285
)
31
Reyes
 
F
Lepage
 
E
Ganem
 
G
, et al. 
ACVBP versus CHOP plus radiotherapy for localized aggressive lymphoma.
N Engl J Med
2005
, vol. 
352
 (pg. 
1197
-
1205
)
32
Gisselbrecht
 
C
Lepage
 
E
Molina
 
T
, et al. 
Shortened first-line high-dose chemotherapy for patients with poor-prognosis aggressive lymphoma.
J Clin Oncol
2002
, vol. 
20
 (pg. 
2472
-
2479
)
33
Bonnet
 
C
Fillet
 
G
Mounier
 
N
, et al. 
CHOP alone compared with CHOP plus radiotherapy for localized aggressive lymphoma in elderly patients: a study by the Groupe d'Etude des Lymphomes de l'Adulte.
J Clin Oncol
2007
, vol. 
25
 (pg. 
787
-
792
)
34
Tilly
 
H
Lepage
 
E
Coiffier
 
B
, et al. 
Intensive conventional chemotherapy (ACVBP regimen) compared with standard CHOP for poor-prognosis aggressive non-Hodgkin lymphoma.
Blood
2003
, vol. 
102
 (pg. 
4284
-
4289
)
35
Bastion
 
Y
Blay
 
JY
Divine
 
M
, et al. 
Elderly patients with aggressive non-Hodgkin's lymphoma: disease presentation, response to treatment, and survival: a Groupe d'Etude des Lymphomes de l'Adulte study on 453 patients older than 69 years.
J Clin Oncol
1997
, vol. 
15
 (pg. 
2945
-
2953
)
36
Coiffier
 
B
Gisselbrecht
 
C
Herbrecht
 
R
Tilly
 
H
Bosly
 
A
Brousse
 
N
LNH-84 regimen: a multicenter study of intensive chemotherapy in 737 patients with aggressive malignant lymphoma.
J Clin Oncol
1989
, vol. 
7
 (pg. 
1018
-
1026
)
37
Cheson
 
BD
Horning
 
SJ
Coiffier
 
B
Report of an international workshop to standardize response criteria for non-Hodgkin's lymphomas: NCI Sponsored International Working Group.
J Clin Oncol
1999
, vol. 
17
 (pg. 
1244
-
1253
)
38
Kaplan
 
EL
Meier
 
P
Non parametric estimation for incomplete observations.
J Am Stat Assoc
1958
, vol. 
53
 (pg. 
457
-
481
)
39
Mantel
 
N
Evaluation of survival data and two new rank order statistics arising in its consideration.
Cancer Chemother Rep
1966
, vol. 
50
 (pg. 
163
-
170
)
40
Gallamini
 
A
Stelitano
 
C
Calvi
 
R
, et al. 
Peripheral T-cell lymphoma unspecified (PTCL-U): a new prognostic model from a retrospective multicentric clinical study.
Blood
2004
, vol. 
103
 (pg. 
2474
-
2479
)
41
Went
 
P
Agostinelli
 
C
Gallamini
 
A
, et al. 
Marker expression in peripheral T-cell lymphoma: a proposed clinical-pathologic prognostic score.
J Clin Oncol
2006
, vol. 
24
 (pg. 
2472
-
2479
)
42
Frizzera
 
G
Moran
 
E
Rappaport
 
H
Angio-immunoblastic lymphadenopathy with dysproteinemia.
Am J Med
1975
, vol. 
1
 (pg. 
1070
-
1073
)
43
Lukes
 
R
Tindle
 
B
Immunoblastic lymphadenopathy: a hyperimmune entity resembling Hodgkin's disease.
N Eng J Med
1975
, vol. 
292
 (pg. 
1
-
8
)
44
Tobinai
 
K
Minato
 
K
Ohtsu
 
T
, et al. 
Clinicopathologic, immunophenotypic and immunogenotypic analyses of immunoblastic lymphadenopathy-like T-cell lymphoma.
Blood
1998
, vol. 
72
 (pg. 
1000
-
1006
)
45
Mounier
 
N
Gisselbrecht
 
C
Briere
 
J
, et al. 
All aggressive lymphoma subtypes do not share similar outcome after front-line autotransplantation: a matched-control analysis by the Groupe d'Etude des Lymphomes de l'Adulte (GELA).
Ann Oncol
2004
, vol. 
15
 (pg. 
1790
-
1797
)
46
Schetelig
 
J
Fetscher
 
S
Reichle
 
A
, et al. 
Long-term disease-free survival in patients with angioimmunoblastic T-cell lymphoma after high-dose chemotherapy and autologous stem cell transplantation.
Haematologica
2003
, vol. 
88
 (pg. 
1272
-
1278
)
47
Kyriakou
 
C
Canals
 
C
Goldstone
 
A
, et al. 
High-dose therapy and autologous stem-cell transplantation in angioimmunoblastic lymphoma: complete remission at transplantation is the major determinant of outcome: Lymphoma Working Party of the European Group for Blood and Marrow Transplantation.
J Clin Oncol
 
Prepublished on December 10, 2007, as DOI 10.1200/JCO-2008–12-6219
48
Ree
 
HJ
Kadin
 
ME
Kikuchi
 
M
, et al. 
Angioimmunoblastic lymphoma (AILD-type T-cell lymphoma) with hyperplastic germinal centers.
Am J Surg Pathol
1998
, vol. 
22
 (pg. 
643
-
655
)
49
Nathwani
 
BN
Rappaport
 
H
Moran
 
EM
Pangalis
 
GA
Kim
 
H
Malignant lymphoma arising in angioimmunoblastic lymphadenopathy.
Cancer
1978
, vol. 
41
 (pg. 
578
-
606
)
50
Kim
 
CH
Lim
 
HW
Kim
 
JR
Rott
 
L
Hillsamer
 
P
Butcher
 
EC
Unique gene expression program of human germinal center T helper cells.
Blood
2004
, vol. 
104
 (pg. 
1952
-
1960
)
51
Chtanova
 
T
Tangye
 
SG
Newton
 
R
, et al. 
T follicular helper cells express a distinctive transcriptional profile, reflecting their role as non-Th1/Th2 effector cells that provide help for B cells.
J Immunol
2004
, vol. 
173
 (pg. 
68
-
78
)
52
Ansel
 
KM
Ngo
 
VN
Hyman
 
PL
, et al. 
A chemokine-driven positive feedback loop organizes lymphoid follicles.
Nature
2000
, vol. 
406
 (pg. 
309
-
314
)
53
Grogg
 
KL
Attygalle
 
AD
Macon
 
WR
Remstein
 
ED
Kurtin
 
PJ
Dogan
 
A
Expression of CXCL13, a chemokine highly upregulated in germinal center T-helper cells, distinguishes angioimmunoblastic T-cell lymphoma from peripheral T-cell lymphoma, unspecified.
Mod Pathol
2006
, vol. 
19
 (pg. 
1101
-
1107
)
54
Advani
 
R
Warnke
 
R
Sikic
 
BI
Horning
 
S
Treatment of angioimmunoblastic T-cell lymphoma, with cyclosporine.
Ann Oncol
1997
, vol. 
8
 (pg. 
601
-
603
)
55
Quintini
 
G
Iannitto
 
E
Barbera
 
V
, et al. 
Response to low-dose oral methotrexate and prednisone in two patients with angio-immunoblastic lymphadenopathy-type T-cell lymphoma.
Hematol J
2001
, vol. 
2
 (pg. 
393
-
395
)
56
Siegert
 
W
Nerl
 
C
Meuthen
 
I
, et al. 
Recombinant human interferon-alpha in the treatment of angioimmunoblastic lymphadenopathy: results in 12 patients.
Leukemia
1991
, vol. 
5
 (pg. 
892
-
895
)
57
Ong
 
ST
Koeppen
 
H
Larson
 
RA
Olopade
 
OI
Successful treatment of angioimmunoblastic lymphadenopathy with dysproteinemia with fludarabine.
Blood
1996
, vol. 
88
 (pg. 
2354
-
2355
)
58
Mourad
 
N
Mounier
 
N
Briere
 
J
, et al. 
Angioimmunoblastic T-cell lymphoma: a clinicopathologic study of 158 patients treated in GELA trials.
Blood
2006
, vol. 
108
 pg. 
121a
  
Abstract 397
Sign in via your Institution