Lymphomas were studied in kindreds with autoimmune lymphoproliferative syndrome (ALPS; Canale-Smith syndrome), a disorder of lymphocyte homeostasis usually associated with germline Fas mutations. Fas (CD95/APO-1) is a cell surface receptor that initiates programmed cell death, or apoptosis, of activated lymphocytes. Lymphoma phenotype was determined by immunohistochemistry, frequency of CD3+CD4CD8 T-cell–receptor α/β cells by flow cytometry, nucleotide sequences of the gene encoding Fas (APT1, TNFRSF6), and the percentage of lymphocytes undergoing apoptosis in vitro. Of 223 members of 39 families, 130 individuals possessed heterozygous germline Fas mutations. Eleven B-cell and T-cell lymphomas of diverse types developed in 10 individuals with mutations in 8 families, up to 48 years after lymphoproliferation was first documented. Their risk of non-Hodgkin and Hodgkin lymphomas, respectively, was 14 and 51 times greater than expected (each P < .001). Investigation of these 10 patients and their relatives with Fas mutations revealed that all had defective lymphocyte apoptosis and most had other features of ALPS. The tumor cells retained the heterozygous Fas mutations found in the peripheral blood and manifested defective Fas-mediated killing. These data implicate a role for Fas-mediated apoptosis in preventing B-cell and T-cell lymphomas. Inherited defects in receptor-mediated lymphocyte apoptosis represent a newly appreciated risk factor for lymphomas.

Chromosomal translocations leading to the dysregulation of cell division and differentiation contribute to lymphomagenesis.1-3 Another genetic mechanism that contributes to lymphoma development is the acquired failure of lymphocytes to be susceptible to programmed cell death, or apoptosis. Some lymphoma cells overexpress Bcl-2, a protein that inhibits apoptosis.4-6 Each of these predisposing molecular and cytogenetic defects arise within individual somatic cells, but it is unclear whether they relate to the hereditary risk for lymphoma.

The recent recognition of the autoimmune lymphoproliferative syndrome (ALPS; also called the Canale-Smith syndrome) afforded us the opportunity to explore its cause, inherited defects in genes that promote lymphocyte apoptosis, as an additional contributor to lymphoma development.7-18 ALPS is characterized by chronic lymphadenopathy and splenomegaly of early onset, autoimmune phenomena, and expanded populations of T-cell receptor α/β (TCRα/β) CD3+CD4CD8, or double-negative, T cells (DNTCs). Among over 70 reported kindreds with ALPS,7-16 most are cases of ALPS type Ia that are associated with mutations in the APT1 (TNFRSF6) gene encoding Fas (CD95/Apo-1). Patients with mutations in theFas ligand gene (TNFSF6)16 possess ALPS type Ib. Cases with mutations in caspase 10 are termed ALPS type II,17 and those without defined mutations are termed ALPS type III.11,18 

Fas is a transmembrane receptor in the tumor necrosis factor receptor (TNFR) gene family. B- and T- lymphocyte apoptosis is initiated by binding of the Fas ligand to Fas.19 Fas transduces the death signal through its cytoplasmic “death domain,” the binding site for proteins that activate cysteine proteases (caspases) that mediate the apoptosis cascade.20-22 The prominent lymphoid abnormalities in ALPS in the absence of significant pathology in other organ systems suggest that the Fas-Fas ligand apoptosis pathway has 2 primary roles: the elimination of unneeded and potentially deleterious lymphocytes, and the killing of virus-infected cells.19 We now ask whether Fas-mediated apoptosis plays a role in the prevention of lymphoid malignancy.

Study subjects

One hundred members of 26 families studied at the National Institutes of Health (NIH) were found to have deleterious Fas mutations: 6 members of 5 of these families developed B-cell lymphomas (Figure 1 and Table1). In the course of this work, we learned of 4 more lymphomas among 30 individuals with Fas mutations in 13 additional families including 2 cases in Germany23 and 2 individuals studied in New York.

Fig. 1.

Structure of the TNFRSF6 (APT1) gene encoding the Fas protein, showing locations of the mutations associated with ALPS and lymphoma.

Exon 9 is expanded to show α-helical regions of the intracellular death domain.67 Mutations are newly described or previously reported from NIH (indicated by black symbols),8,11,15,44 Cornell University, New York (indicated by gray symbols),10,26 or Dresden, Germany (indicated by white symbols).30 Mutations seen in individuals with lymphoma are indicated by boxed symbols.

Fig. 1.

Structure of the TNFRSF6 (APT1) gene encoding the Fas protein, showing locations of the mutations associated with ALPS and lymphoma.

Exon 9 is expanded to show α-helical regions of the intracellular death domain.67 Mutations are newly described or previously reported from NIH (indicated by black symbols),8,11,15,44 Cornell University, New York (indicated by gray symbols),10,26 or Dresden, Germany (indicated by white symbols).30 Mutations seen in individuals with lymphoma are indicated by boxed symbols.

Close modal
Table 1.

Clinical and laboratory profiles of lymphoma patients with germline Fas mutation

Study centerPatientSexAge*AdenopathySplenomegalyAutoimmune diseaseDNTCs (%)Cells killed (%)ALPS1-153Yr from onset1-155LymphomaFas mutation
AnemiaThrombocytopeniaNeutropeniaAutoantibodies
Bethesda 3-II-1 Male 38 − − 6.1 32 22 NLP Hodgkin disease 915A→C 
Bethesda 3-II-3 Male 27 − − ND ND ND − 21 T-cell–rich B lymphoma 915A→C 
Bethesda 26-II-4 Male 54 − 0.7 − 48 Burkitt lymphoma 973A→T 
Bethesda 30-II-7 Female 30 − − 5.9 15 Burkitt lymphoma 1074delT 
Bethesda 45-III-2 Male 12 − 11.7 12 Hodgkin disease 779del11 
Bethesda 55-II-1 Male 19 − − 9.7 15 Follicular B lymphoma 942C→T  
New York P9-II-4 Male 30 − − ND ND 29 − 21 Hodgkin disease IVS7(+2)T→A  
New York P10-I-1 Female 38 − − 18.0 Hodgkin disease 1003C→T 
Germany G3-II-8 Male 31 − − − − ND 4.0 < 2  − Hodgkin disease; marginal zone B-cell lymphoma 1009A→G 
Germany G3-III-4 Female − − 75 12 T-cell lymphoma 1009A→G 
Study centerPatientSexAge*AdenopathySplenomegalyAutoimmune diseaseDNTCs (%)Cells killed (%)ALPS1-153Yr from onset1-155LymphomaFas mutation
AnemiaThrombocytopeniaNeutropeniaAutoantibodies
Bethesda 3-II-1 Male 38 − − 6.1 32 22 NLP Hodgkin disease 915A→C 
Bethesda 3-II-3 Male 27 − − ND ND ND − 21 T-cell–rich B lymphoma 915A→C 
Bethesda 26-II-4 Male 54 − 0.7 − 48 Burkitt lymphoma 973A→T 
Bethesda 30-II-7 Female 30 − − 5.9 15 Burkitt lymphoma 1074delT 
Bethesda 45-III-2 Male 12 − 11.7 12 Hodgkin disease 779del11 
Bethesda 55-II-1 Male 19 − − 9.7 15 Follicular B lymphoma 942C→T  
New York P9-II-4 Male 30 − − ND ND 29 − 21 Hodgkin disease IVS7(+2)T→A  
New York P10-I-1 Female 38 − − 18.0 Hodgkin disease 1003C→T 
Germany G3-II-8 Male 31 − − − − ND 4.0 < 2  − Hodgkin disease; marginal zone B-cell lymphoma 1009A→G 
Germany G3-III-4 Female − − 75 12 T-cell lymphoma 1009A→G 

Presence (+), or absence or lack of confirmation (−) of clinical finding.

ND indicates not done; NLP Hodgkin disease, nodular lymphocyte-predominant Hodgkin disease.

*

When last studied, or died.

Percent of TCR α/βCD4CD8 T cells. The laboratory normal value is < 1.0%.

Percent of cells killed after treatment with an anti-Fas MoAb. The NIH laboratory normal value is 52% ± 12% (mean ± SD).

F1-153

The case does (+) or does not (−) meet the ALPS case definition. Immunophenotyping could not be done to confirm an elevated percentage of DNTCs in subjects who had died.

F1-155

Time in years from manifesting first features of ALPS until lymphoma diagnosis.

All individuals with ALPS-like features within these families with Fas mutations and their available relatives in each of the study centers were enrolled after written informed consent in institutional review board–approved research protocols. All amenable subjects were examined and bled for extensive studies. In all, 223 persons in 39 families were evaluated.

Case definition

Autoimmune lymphoproliferative syndrome was defined as chronic, nonmalignant lymphadenopathy and/or splenomegaly, defective lymphocyte apoptosis in vitro, and 1% or more DNTCs.

Flow cytometry analysis

The lymphocyte phenotypes of peripheral blood mononuclear cells (PBMCs) were determined at each study site by 2- or 3-color flow cytometry, as reported.8,10,11,15,23 

Immunohistochemical analysis

Hematoxylin and eosin-stained sections from formalin- or B5-fixed paraffin-embedded tissue blocks were prepared, and immunophenotypic studies were performed on unstained paraffin sections with a panel of antibodies, as described previously.24,25 

Detection of Fas gene mutation

Genomic DNA was prepared from PBMCs or paraffin-embedded tissue blocks from deceased individuals and screened for Fas mutations using described methods.8,10,23,26 

Cell culture and stimulation

The PBMCs were separated from heparinized venous blood or buffy coat fractions by Ficoll-Hypaque (Pharmacia Fine Chemicals, Piscataway, NJ) density gradients. PBMCs at 106/mL were activated with 10 μg/mL phytohemagglutinin (PHA) in RPMI 1640 with 10% fetal calf serum, 2 mM glutamine, and penicillin-streptomycin. Cultures were maintained in recombinant human interleukin 2 (IL-2) (100-200 IU/mL) (Midwest Medical, Bridgeton, MO) with refeeding every 48 to 72 hours.

Transformation of immortalized Epstein-Barr virus cell lines

The Epstein-Barr virus (EBV)-transformed B-cell lines were prepared and cultured by standard methods.27 

Induction of apoptosis

Fas-mediated apoptosis was induced and assayed in peripheral blood T cells, EBV cell lines, or cell suspensions made from patient tissue with 0.75 μg CH11 or 0.1 μg/mL APO-1, both anti-Fas monoclonal antibodies (MoAbs; Kamiya Biomedical, Thousand Oaks, CA), or as described previously.10,11,23 

Statistical methods

For the analysis of TCR α/β DNTC results, median values for the percentage of total T-cell number were compared by the Wilcoxon 2-sample test. The same method was applied to cell death induced by anti-Fas MoAb in both T and B cells. All statistical tests were 2-tailed.

For estimation of the risk of developing lymphoma, patient years were calculated using the age at the most recent evaluation or when lymphoma was diagnosed. The Poisson distribution was used to determine the probabilities of observing 5 or more non-Hodgkin or Hodgkin lymphoma cases, given the expected numbers (9.4 and 2.6 cases/100 000, respectively) reported in the SEER Cancer Statistics Review for U.S. men and women aged less than 65 years.28 With a median age of 27.8 years (range 1-86 years) and only 5 study individuals with Fas mutations over age 65, the SEER data for the population under age 65 years most closely described the distribution of ages in this study. The 95% confidence interval for the observed to expected ratio was determined, as described.29 

Case histories of lymphoma patients

In NIH family 3 with a Fas alteration (915A→C; T225P; Figure 1) subject 3-II-3 had anemia, cervical adenopathy, and a palpable spleen at age 4 years and underwent 2 nondiagnostic lymph node biopsies before a third at age 25 suggested Hodgkin disease. Chemotherapy and radiation therapy were instituted for stage IIIA disease; however, he died of extensive disease. The pathologic specimens were reviewed (by E.S.J.) and reclassified as histiocyte-rich, T-cell–rich large B-cell lymphoma that was negative for EBV by in situ hybridization (Figure2A).30,31 The lymphoma cells were CD20+ but negative for CD30 and CD15 in a background of T cells and histiocytes.

Fig. 2.

Histopathology and immunopathology of lymphomas in ALPS patients.

(A) T-cell and histiocyte-rich B-cell lymphoma diagnosed in patient 3-II-3. Large polylobated tumor cells, which stained for CD20, are seen in a background of T lymphocytes and histiocytes. (B-D) Nodular lymphocyte-predominant Hodgkin disease diagnosed in patient 3-II-1. (B) A rim of epithelioid granulomas surrounds a large nodule containing neoplastic cells. (C) A single polylobated L&H cell is identified in a background of small lymphocytes. (D) A CD20+ L&H cell is rosetted by a rim of CD20 T cells. Numerous CD20+ small B cells are present in the background. (E) Burkitt lymphoma presenting as an omental mass in patient 26-II-4. Tumor cells are moderate in size with clumped chromatin and basophilic nucleoli. A prominent starry sky is present. (F) Tumor cells uniformly express CD20. The growth fraction was 100% with MIB-1 (Ki-67) staining. Identical histologic and immunophenotypic features were seen in lymphoma involving small bowel in patient 30-III-2. (G) An unusual form of follicular lymphoma was diagnosed in patient 55-II-1. Neoplastic cells surround hyalinized and possibly regressed follicular remnants. (H) CD20 immunohistochemistry emphasized follicular pattern. (I) The neoplastic cells are moderate in size with clumped chromatin. (J) A follicle-center origin for tumor cells is supported by Bcl-6 immunohistochemistry, which selectively stains the neoplastic cells within the follicles. Original magnification (A), ×400; (B), ×60; (C), ×400; (D), ×400; (E), ×400; (F), ×400; (G), ×200; (H), ×200; (I), ×400; and (J), ×250.

Fig. 2.

Histopathology and immunopathology of lymphomas in ALPS patients.

(A) T-cell and histiocyte-rich B-cell lymphoma diagnosed in patient 3-II-3. Large polylobated tumor cells, which stained for CD20, are seen in a background of T lymphocytes and histiocytes. (B-D) Nodular lymphocyte-predominant Hodgkin disease diagnosed in patient 3-II-1. (B) A rim of epithelioid granulomas surrounds a large nodule containing neoplastic cells. (C) A single polylobated L&H cell is identified in a background of small lymphocytes. (D) A CD20+ L&H cell is rosetted by a rim of CD20 T cells. Numerous CD20+ small B cells are present in the background. (E) Burkitt lymphoma presenting as an omental mass in patient 26-II-4. Tumor cells are moderate in size with clumped chromatin and basophilic nucleoli. A prominent starry sky is present. (F) Tumor cells uniformly express CD20. The growth fraction was 100% with MIB-1 (Ki-67) staining. Identical histologic and immunophenotypic features were seen in lymphoma involving small bowel in patient 30-III-2. (G) An unusual form of follicular lymphoma was diagnosed in patient 55-II-1. Neoplastic cells surround hyalinized and possibly regressed follicular remnants. (H) CD20 immunohistochemistry emphasized follicular pattern. (I) The neoplastic cells are moderate in size with clumped chromatin. (J) A follicle-center origin for tumor cells is supported by Bcl-6 immunohistochemistry, which selectively stains the neoplastic cells within the follicles. Original magnification (A), ×400; (B), ×60; (C), ×400; (D), ×400; (E), ×400; (F), ×400; (G), ×200; (H), ×200; (I), ×400; and (J), ×250.

Close modal

This individual's brother, subject 3-II-1, had documented cervical and axillary lymphadenopathy by age 3 years. Six biopsies revealed reactive hyperplasia before age 15, when he developed Coombs-positive hemolytic anemia, hepatosplenomegaly, and thrombocytopenia. Splenectomy was performed at age 18. At age 25, further biopsy showed lymphoma (Figure 2B-D), confirmed as an EBV-negative nodular lymphocyte-predominant Hodgkin disease on review. He received combination chemotherapy for stage IIIA disease; however, he relapsed in 1986 and again in 1992. On re-evaluation in 1995, he was free of tumor and asymptomatic, although diffuse peripheral lymphadenopathy persisted.

Study of NIH family 26 with a heterozygous Fas alteration (973A→T; D244V; Figure 1) identified subject 26-II-4, who presented at age 2 with adenopathy and splenomegaly and was diagnosed with acute leukemia. Splenectomy was performed at age 3, and idiopathic thrombocytopenic purpura occurred at age 23. At age 50, Burkitt lymphoma was recognized in an omental mass and confirmed as an EBV-positive B-cell lymphoma (Figure 2E); the tumor remitted with chemotherapy. At age 54, he developed Coombs-positive hemolysis.

Studies of NIH family 30 with a Fas alteration (1074delT; T227fs [frame-shift]; Figure 1) identified female subject 30-II-7with anemia, adenopathy, and splenomegaly at age 5 weeks, splenectomy and lymph node biopsy at age 7, followed by autoimmune hemolytic anemia and idiopathic thrombocytopenic purpura (ITP). At age 17, a new abdominal mass represented EBV-negative Burkitt lymphoma (Figure 2E,F). She was treated with cyclophosphamide, vincristine, and prednisone. At age 31, she remains free of lymphoma.

NIH patient 45-III-2 is a 12-year-old boy who presented with adenopathy, splenomegaly, anemia, and ITP at ages 5 and 7, which led to splenectomy and lymph node biopsy that showed changes typical of ALPS (Table 1).32 At age 10, his Fas mutation (779def11; K181fs [frameshift]; Figure 1) was found. At 11, he lost weight and experienced marked enlargement of lymph nodes in the neck and chest; a biopsy demonstrated Hodgkin disease, EBV-positive, mixed cellularity subtype. He is currently completing multidrug chemotherapy.

NIH subject 55-II-1 is a 19-year-old man (Table 1). He presented with splenomegaly at age 2, underwent splenectomy at age 4, and experienced chronic adenopathy and multiple episodes of thrombocytopenia treated with glucocorticosteroids or intravenous immunoglobulin. A lymph node biopsy at age 4 was nondiagnostic, but 3 biopsies for enlarging axillary, submandibular, and inguinal nodes at age 17 demonstrated a nonclassifiable B-cell lymphoma with atypical follicular proliferation and chromosomal rearrangements (Figure 2G-J). The axillary lymph node was effaced by an atypical follicular proliferation. The neoplastic cells in it were positive for CD20, CD19, CD79a, and Bcl-6 but negative for CD10 and bcl-2, suggestive of a follicle center origin (Bcl-6+) but unusual for follicular lymphoma (Bcl-2, CD10).33 The cells were negative for surface immunoglobulin and for EBV. Stains for CD5, cyclin D1, CD23, and CD11c were negative. Rearrangements of theIg and TCR-γ genes were examined by polymerase chain reaction (PCR) and showed no clonal bands.34 The cells were negative by PCR for translocations involving the Ig heavy chain gene and the Bcl-2 major breakpoint region and the Bcl-1 major translocation cluster. The submandibular and inguinal lymph nodes showed abnormal follicular B-cell proliferation consistent with lymphoma. The cells were surface Ig, but dim monoclonal cytoplasmic κ light chain expression was detected by flow cytometry. Two of the node biopsies showed identical clonal cytogenetic abnormalities with unbalanced translocations involving multiple chromosomes. His peripheral blood showed a heterozygous Fas mutation (942C→T; R234X; Figure 1). Four cycles of rituximab yielded partial response. With further nodal enlargement, additional biopsies revealed a more aggressive, Bcl-2+ follicular lymphoma that remitted with multidrug chemotherapy.

In New York family 9 with Fas mutations (1003C→T; T254I; Figure 1), subject P9-II-4 manifested adenopathy, splenomegaly, and autoimmune hemolysis at age 5. Over the next decade, he underwent a splenectomy and several nondiagnostic lymph node biopsies. At age 26, he developed night sweats, and further biopsy revealed Hodgkin disease, mixed cellularity type. His maternal aunt (P9-I-5) had adenopathy since infancy and died at age 36 from a T-cell–rich large B-cell lymphoma. Fas sequencing could not be conducted on her available tissue sections, and her case is not included in the analyses below.

In New York family P10 with a Fas mutation (IVS7(+2)T6A; P201fs; Figure1), subject P10-I-1 is a 38-year-old woman with Hodgkin disease, nodular sclerosis type, stage IA, at age 5, treated successfully with local irradiation. Three years later, she developed abdominal and inguinal adenopathy and splenomegaly. Repeat biopsies and splenectomy showed only reactive changes compatible with ALPS,32 but no lymphoma. ITP developed at age 17.

An EBV-positive T-cell lymphoma (G3-III-4) in German family 3 with a Fas mutation (1009A→G; E256G; Figure 1) was reported recently.23 Also reported was individual G3-II-8, who had Hodgkin disease, mixed cellularity subtype, at age 13.23 Seventeen years later, however, he presented with fever, wasting, and night sweats. A malignant, marginal zone non-Hodgkin B-cell lymphoma was identified. He died despite 4 cycles of chemotherapy.

No lymphomas were uncovered in the remaining 31 study families. The specific mutations associated with ALPS in all 39 families are shown in Figure 1.

Flow cytometry

Flow cytometry was performed on peripheral blood lymphocytes from 57 members of the 8 families with lymphomas. Of 36 members with Fas mutations tested, 31 possessed elevated (≥ 1%) TCR α/β DNTCs (median of 4.0%; range 0.5%-75%). All 21 healthy relatives tested without mutations had significantly lower (< 1%) DNTC percentages (P < .001).

Induction of Fas-mediated apoptosis

Fifty-four members of the lymphoma kindreds underwent apoptosis tests. Lymphocytes from 36 individuals with Fas mutations showed depressed lymphocyte apoptosis (median cell loss 5%; range 0%-32%), whereas for 18 subjects lacking Fas mutations, the median percentile of T cells undergoing apoptosis was 44% (range 14%-70%;P < .001). Fas-mediated killing of EBV-transformed B cells was studied in 11 subjects from NIH family 3. In the 4 individuals with mutations, 11% to 25% of B cells died, whereas 33% to 59% of B cells died from 7 relatives with normal Fas alleles (P = .006). Thus, Fas defects result in impaired apoptosis of both B cells and T cells, as noted previously.11 

Fas-mediated killing and genetics in tumor cells

In vitro studies showed that defective apoptosis is a feature of both nonmalignant and lymphomatous cells from ALPS lymph node tissues and that the heterozygous mutation transmitted in the germline is sufficient for the development of lymphoma. Cells from a lymphomatous node of patient 55-II-1 and from a typical nonlymphomatous node from an ALPS patient32 were tested and compared with Jurkat T-cell lymphoma and KK124B Burkitt lymphoma (a gift of Dr Kishor Bhatia) cell lines as well as activated fresh normal human tonsillar lymphocytes. The cells were treated with MoAbs that trigger Fas-mediated cell death or with staurosporine, which kills cells via Fas-independent mitochondrial pathways. Using flow cytometry, it was possible to distinguish the lymphomatous cells from patient 55-II-1 as being relatively large B cells, consistent with their immunohistologic appearance (Figure 2G-I). High percentages of tonsillar lymphocytes, Jurkat cells, and KK124B cells were killed by both apoptosis inducers (Figure 3). Resting lymph node (RLN) cells from ALPS patient 17-II-1were relatively refractory to Fas-mediated death but sensitive to staurosporine, as expected.19 Both the malignant and nonmalignant lymph node B cells from patient 55-II-1 were refractory to Fas killing but sensitive to staurosporine. Thus, malignant transformation did not introduce or mitigate the inherited apoptotic defect.

Fig. 3.

Resistance of lymphoma cells to Fas-induced apoptosis.

Cell suspensions prepared from a lymphomatous lymph node from NIH ALPS proband 55 were subjected to apoptosis induction either via antibody cross-linking of Fas or by 5 μg/mL staurosporine that induces apoptosis by the mitochondrial pathway. The percentages of cells dying are compared with the results with resting lymph node (RLN) cells from patient 55, with cells from a family 17 ALPS patient, with Jurkat T-lymphoma cells, with KK124B Burkitt lymphoma cells, and with normal human tonsil cells. Flow cytometry analysis showed that lymphoma cells from patient 55 were of large size and exhibited B-cell markers and that both the lymphoma cells and the smaller resting lymphocytes expressed normal levels of the Fas receptor on the cell surface.

Fig. 3.

Resistance of lymphoma cells to Fas-induced apoptosis.

Cell suspensions prepared from a lymphomatous lymph node from NIH ALPS proband 55 were subjected to apoptosis induction either via antibody cross-linking of Fas or by 5 μg/mL staurosporine that induces apoptosis by the mitochondrial pathway. The percentages of cells dying are compared with the results with resting lymph node (RLN) cells from patient 55, with cells from a family 17 ALPS patient, with Jurkat T-lymphoma cells, with KK124B Burkitt lymphoma cells, and with normal human tonsil cells. Flow cytometry analysis showed that lymphoma cells from patient 55 were of large size and exhibited B-cell markers and that both the lymphoma cells and the smaller resting lymphocytes expressed normal levels of the Fas receptor on the cell surface.

Close modal

Finally, complementary DNAs (cDNAs) prepared from malignant CD19+ B cells sorted from a node from this patient and nonmalignant cells of a lymph node from patient 31-III-1were sequenced. Multiple cDNA clones prepared from proband 55 retained the 942C→T mutation seen in his peripheral blood and no other Fas mutations, whereas the majority of clones revealed wild-type Fas sequences. From proband 31, similar numbers of clones were mutant and wild type. Thus, the tumor in patient 55, like the reactive node of patient 31, showed only heterozygous Fas mutations.

Risk of lymphoma development

The ratio of observed cases of non-Hodgkin lymphoma (5 cases/3774 patient years, not counting the second lymphoma in subject G3-II-8) among the 130 individuals with mutations to the expected rate among the general population less than 65 years of age (9.4 cases/100 000 patient years)28 was 14 (95% confidence interval [CI] 5- to 33-fold), a significant increase (P < .001). The ratio of observed to expected cases of Hodgkin lymphoma among the general population (5 cases/3774 patient years versus 2.6/100 000 patient years)28 was 51 (95% CI 17- to 119-fold; P < .001).

We examined the possibility that germline Fas mutations and the concomitant defect in lymphocyte apoptosis are hereditary predisposing factors to lymphoma. One hundred thirty members of 39 kindreds that possess inherited Fas mutations were studied; lymphomas cosegregated with Fas mutations in 10 individuals in 8 of the families. These 8 families included 75 members of whom 43, including all 10 individuals with lymphoma, exhibited some or all of the manifestations of ALPS, such as lymphoproliferation, especially of DNTCs, and autoimmunity. The risk of Hodgkin and non-Hodgkin lymphoma in individuals with inherited Fas mutations in our study is 51 and 14 times greater than expected, respectively. We found that both the nonmalignant and malignant lymphocytes from the lymphoma subjects exhibited defective Fas-mediated killing, but they did not have a general apoptosis defect, because the cells responded normally to other apoptosis inducers (Figure3).

A significant feature of lymphoma in ALPS is its diversity.1 The 5 cases of Hodgkin disease were of various types including nodular sclerosing, nodular-lymphocyte predominant, and mixed cellularity. The others represented several histologic types of lymphoma, including a T-cell type, a histiocyte-rich T-cell–rich large B-cell type, a follicular B-cell type, a marginal zone B-cell type, and 2 cases of Burkitt lymphoma. The preponderance of B-cell malignancies is notable given the fact that apoptosis in both T and B cells is abnormal in ALPS and that the major nonmalignant cell types that expand in ALPS are DNTCs, which are believed to be mature T cells that have lost expression of their CD4 and CD8 coreceptors.8,11,19Therefore, Fas defects do not appear to shift the distribution between T and B lymphoid malignancies. Given that our study group is still small, we cautiously infer that Fas apoptosis deficiency affects a general protective mechanism against B-cell malignancy rather than one that controls a particular B-cell subtype. Such a conclusion would be consistent with evidence that Fas governs the fate of developing B cells in the bone marrow as well as mature B cells in the peripheral immune organs.39 

The involvement of Fas mutations in lymphoma is distinct from other apoptosis defects that have been postulated to play a role in lymphoma. Previously, genetic alterations in Bcl-2–related molecules were associated with lymphoid malignancy40-42; however, Bcl-2 protects against an apoptosis pathway emanating from the mitochondrion that was not defective in the lymphoma sample from patient 55 (Figure3).19 By contrast, Fas is a cell surface receptor that promotes apoptosis in response to a specific inducer, Fas ligand, to govern lymphocyte homeostasis during immune responses.19The lymphomas we observed are all associated with alterations in the intracytoplasmic region of the Fas receptor, termed the “death domain.” Individuals with mutations that affect the death domain have the greatest defect in Fas-mediated apoptosis and the greatest genetic penetrance and severity of clinical symptoms.43,44 Therefore, the propensity to develop B lymphomas relates to the severity of apoptosis defect engendered by specific lesions in the Fas gene, implying that the specific apoptotic function of Fas is of primary importance in protecting against lymphoma.

Our findings are in accord with prior data associating defects in Fas function with lymphoid malignancy. Several studies revealed underexpression of Fas in lymphoma45-50 and multiple myeloma,51-54 and somatic chromosomal abnormalities in non-Hodgkin lymphoma in the region of chromosome 10 encoding Fas.55-61 Moreover, somatic Fas mutations have been reported in multiple myeloma, adult T-cell leukemia, childhood T-lineage acute lymphoblastic leukemia,35-37 and in 9 of 150 non-Hodgkin lymphomas.38 Among these 9 cases, 3 were heterozygous and involved the Fas death domain, including 1 with the identical mutation seen in our patient 26-II-4. Three others involved mutations outside the death domain in one allele but deletion of the second Fas allele.38 We demonstrated heterozygosity of the Fas mutation in tumor cells from one of our cases (55-II-1); however, the cumulative evidence indicating that the mutations in all of our patients are dominant-negative implies that all tumors that arose in them involved heterozygous Fas mutations.

In our cases, the average age of ALPS onset was 5 years, whereas the average age of lymphoma diagnosis was 28. The cases where somatic alterations in Fas were described in lymphomas more typically arose later in life.35-38 The lag between the manifestations of ALPS and lymphoma could represent time for other genetic or environmental lymphomagenic factors to operate on the substrate of defective apoptosis. This conclusion is supported by studies of mice defective in Fas apoptosis that show lymphoid malignancies arising in conjunction with expression of the L-myc oncogene or as a consequence of aging.7,62-65 

The mechanism by which Fas defects in ALPS predispose to lymphomas might involve several components. The most obvious possibility is that a general expansion of the lymphoid pool provides a larger target cell population for other transforming events. This does not seem to fully explain our observations, because the greatest expansions are in T cells, yet we observe B-cell malignancies. Nonetheless, the absolute increases in B cells, which are stimulated by the expanded T helper 1 cells, and marked elevations in IL-10 in ALPS could be important for the increased lymphoma incidence.66 Other, more specific mechanisms could also participate. Defects in the cytolytic pathway for killing virally infected cells mediated by Fas could allow EBV or other oncogenic viruses to persist. A similar apoptotic clearance mechanism may operate on transformed B cells. Because Fas ligand is expressed by T cells and not B cells, T cells could eliminate Fas-sensitive malignant cells. The study of lymphoma in ALPS provides a genetic model of germline Fas deficiency that will allow further examination of these issues.

We thank Drs John Bohnsack, Howard Britton, Sandra Buys, Brian Corden, Anthony Infante, Donald Mahoney, Annelise Sitarz, and David Virshup for referring patients for study; the participating families themselves for their cooperation; Ms Jean Whitehouse, Ms Romy Lehmann, and Ms Margaret R. Brown for flow cytometry analysis of peripheral blood; Lindsay A. Middleton, for ascertainment of family data; Dr Maryalice Stetler-Stevenson for flow cytometry analysis of lymph nodes; Drs Lixin Zheng, Gretchen Gibney, Michelle Johnson, and Richard Siegel for assistance with apoptosis assays; Drs Mark Raffeld and Lynn Sorbara for molecular studies; Drs Diane Arthur and Linda Cooley for cytogenetic studies; Dr Douglas Kingma for EBV in situ hybridizations; Uri Lopatin for additional statistical help; Drs Margaret Tucker and Joachim Roesler for helpful discussions; Dr G. Koehler for reviewing selected histologic sections; Drs Louis Staudt and Ian Magrath for critical reviews of the manuscript; and Ms Brenda Rae Marshall for editorial assistance.

The contribution of K.B.E. to this work was supported, in part, by grants from the National Institutes of Health (AR4548I), and the fellowship of C.B. was supported by the Cure for Lymphoma Foundation. A.M.J.P. was supported by the Deutsche Forschungsgememeinschaft (DFG 609/1-1). J.W. was supported by an Arthritis Foundation fellowship.

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 U.S.C. section 1734.

1
Kramer
MH
Hermans
J
Wijburg
E
et al
Clinical relevance of BCL2, BCL6, and MYC rearrangements in diffuse large B-cell lymphoma.
Blood.
92
1998
3152
3162
2
Ong
ST
leBeau
MM
Chromosomal abnormalities and molecular genetics of non-Hodgkin's lymphoma.
Semin Oncol.
25
1998
447
460
3
Aguilera
NS
Bijwaard
KE
Duncan
B
et al
Differential expression of cyclin D1 in mantle cell lymphoma and other non-Hodgkin's lymphomas.
Am J Pathol.
153
1998
1969
1976
4
Ohshima
A
Miura
I
Hashimoto
K
et al
Rearrangements of the BCL6 gene and chromosome aberrations affecting 3q27 in 54 patients with non-Hodgkin's lymphoma.
Leuk Lymphoma.
27
1997
329
334
5
Korsmeyer
SJ
Bcl-2 initiates a new category of oncogenes: regulators of cell death.
Blood.
80
1992
879
886
6
Monni
O
Joensuu
H
Franssila
K
Klefstrom
J
Alitalo
K
Knuutila
S
BCL2 overexpression associated with chromosomal amplification in diffuse large B-cell lymphoma.
Blood.
90
1997
1168
1174
7
Sneller
MC
Straus
SE
Jaffe
ES
et al
A novel lymphoproliferative/autoimmune syndrome resembling murine lpr/gld disease.
J Clin Invest.
90
1992
334
341
8
Fisher
GH
Rosenberg
FJ
Straus
SE
et al
Dominant interfering Fas gene mutations impair apoptosis in a human autoimmune lymphoproliferative syndrome.
Cell.
81
1995
935
946
9
Rieux-Laucat
F
LeDeist
F
Hivroz
C
et al
Mutations in Fas associated with human lymphoproliferative syndrome and autoimmunity.
Science.
268
1995
1347
1349
10
Drappa
J
Vaishnaw
AK
Sullivan
KE
Chu
J-L
Elkon
KB
Fas gene mutations in the Canale-Smith syndrome, an inherited lymphoproliferative disorder associated with autoimmunity.
N Engl J Med.
335
1996
1643
1649
11
Sneller
MC
Wang
J
Dale
JK
et al
Clinical, immunologic, and genetic features of an autoimmune lymphoproliferative syndrome associated with abnormal lymphocyte apoptosis.
Blood.
89
1997
1341
1348
12
Bettinardi
A
Brugnoni
D
Quiròs-Roldan
E
et al
Missense mutations in the Fas gene resulting in autoimmune lymphoproliferative syndrome: a molecular and immunological analysis.
Blood.
89
1997
902
909
13
Canale
VC
Smith
CH
Chronic lymphadenopathy simulating malignant lymphoma.
J Pediatr.
70
1967
891
899
14
LeDeist
F
Emile
J-F
Rieux-Laucat
F
et al
Clinical, immunological and pathological consequences of fas-deficient conditions.
Lancet.
348
1996
719
723
15
Infante
AJ
Britton
HA
deNapoli
T
et al
The clinical spectrum in a large kindred with autoimmune lymphoproliferative syndrome (ALPS) due to a Fas mutation that impairs lymphocyte apoptosis.
J Pediatr.
133
1998
629
633
16
Wu
J
Wilson
J
He
J
Xiang
L
Schur
PH
Mountz
JD
Fas ligand mutation in a patient with systemic lupus erythematosus and lymphoproliferative disease.
J Clin Invest.
98
1996
1107
1113
17
Wang
J
Zheng
L
Lobito
A
et al
Inherited human caspase-10 mutations underly defective lymphocyte and dendritic cell apoptosis in autoimmune lymphoproliferative syndrome, type II.
Cell.
98
1999
47
58
18
Dianzani
U
Bragardo
M
diFranco
D
et al
Deficiency of the Fas apoptosis pathway without Fas gene mutations in pediatric patients with autoimmunity/ lymphoproliferation.
Blood.
89
1997
2871
2879
19
Lenardo
M
Chan
KM
Hornung
F
et al
Mature T lymphocyte apoptosis—immune regulation in a dynamic and unpredictable antigenic environment.
Annu Rev Immunol.
17
1999
221
253
20
Ju
ST
Panka
DJ
Cui
H
et al
Fas (CD95)/FasL interactions required for programmed cell death after T-cell activation.
Nature.
373
1995
444
448
21
Zheng
L
Fisher
G
Miller
RE
Peschon
J
Lynch
DH
Lenardo
MJ
Induction of apoptosis in mature T cells by tumour necrosis factor.
Nature.
377
1995
348
351
22
Tartaglia
LA
Ayres
TM
Wong
GH
Goeddel
DV
A novel domain within the 55 kD TNF receptor signals cell death.
Cell.
74
1993
845
853
23
Peters
AMJ
Kohfink
B
Martin
H
et al
Defective apoptosis due to a point mutation in the death domain of CD95 associated with autoimmune lymphoproliferative syndrome, T-cell lymphoma, and Hodgkin's disease.
Exp Hematol.
27
1999
868
874
24
Jaffe
ES
Raffeld
M
Lymphocyte markers in solid tissue.
Manual of Clinical Laboratory Immunology
4th ed.
Rose
NR
deMacario
EC
Fahey
JL
Friendman
H
Penn
GM
1992
288
297
American Society for Microbiology
Washington, DC
25
Quintanilla-Martinez
L
Thieblemont
C
Fend
F
et al
Mantle cell lymphomas lack expression of p27Kip1, a cyclin-dependent kinase inhibitor.
Am J Pathol.
153
1998
175
182
26
Vaishnaw
AK
Orlinick
JR
Chu
J-L
Krammer
PH
Chao
MV
Elkon
KB
The molecular basis for apoptotic defects in patients with CD95 (Fas/Apo-1) mutations.
J Clin Invest.
103
1999
355
363
27
Tosato
G
Generation of Epstein-Barr virus immortalized B-cell lines.
Current Protocols in Immunology.
Coligan
JE
Kruisbeek
AM
Margulies
DH
Shevach
EM
Strober
W
1995
7.22.1
John Wiley & Sons
New York, NY
28
Ries
LAG
Kosary
CL
Hankey
BF
Miller
BA
SEER Cancer Statistics Review, 1973–1995.
1998
National Cancer Institute
Bethesda, MD
29
Lilienfeld
DE
Stolley
PD
Foundations of Epidemiology
3rd ed.
1994
302
303
Oxford University Press
New York, NY
30
Chittal
SM
Brousset
P
Voigt
J-J
Delsol
G
Large B-cell lymphoma rich in T cells and simulating Hodgkin disease.
Histopathology.
19
1991
211
220
31
Delabie
J
Vandenberghe
E
Kennes
C
et al
Histiocyte-rich B-cell lymphoma: a distinct clinicopathologic entity possibly related to lymphocyte-predominant Hodgkin disease, paragranuloma subtype.
Am J Surg Pathol.
16
1992
37
48
32
Lim
MS
Straus
SE
Dale
JK
et al
Pathological findings in human autoimmune lymphoproliferative syndrome.
Am J Pathol.
153
1998
1541
1550
33
Flenghi
L
Ye
BH
Fizzotti
M
et al
A specific monoclonal antibody (PG-B6) detects expression of the BCL-6 protein in germinal center B cells.
Am J Pathol.
147
1996
405
411
34
Elenitoba-Johnson
KS
Kumar
S
Lim
MS
Kingma
DW
Raffeld
M
Jaffe
ES
Marginal zone B-cell lymphoma with monocytoid B-cell lymphocytes in pediatric patients without immunodeficiency: a report of two cases.
Am J Clin Pathol.
107
1997
92
98
35
Landowski
TH
Qu
N
Buyaksal
I
Painter
JS
Dalton
WS
Mutations in the Fas antigen in patients with multiple myeloma.
Blood.
90
1997
4266
4270
36
Tamiya
S
Etoh
K
Suzushima
H
Takatsuki
K
Matsuoka
M
Mutation of CD95 (Fas/Apo-1) gene in adult T-cell leukemia cells.
Blood.
91
1998
3935
3942
37
Beltinger
C
Kurz
E
Böhler
T
Schrappe
M
Ludwig
W-D
Debatin
K-M
CD95 (APO-1/Fas) mutations in childhood T-lineage acute lymphoblastic leukemia.
Blood.
91
1998
3943
3951
38
Grønbæk
K
Straten
P
Ralfkiaer
E
et al
Somatic Fas mutations in non-Hodgkin's lymphoma: association with extranodal disease and autoimmunity.
Blood.
92
1998
3018
3024
39
Goodnow
CC
Cyster
JG
Hartley
SB
et al
Self-tolerance checkpoints in B lymphocyte development.
Adv Immunol.
59
1995
279
368
40
Lauritzen
AF
Moller
PH
Nedergaard
T
Guldberg
P
Hou-Jensen
K
Ralfkiaer
E
Apoptosis-related genes and proteins in Hodgkin's disease.
APMIS.
107
1999
636
644
41
Harada
N
Hata
H
Yoshida
M
et al
Expression of Bcl-2 family of proteins in fresh myeloma cells.
Leukemia.
12
1998
1817
1820
42
Xerri
L
Hassoun
J
Devilard
E
Birnbaum
D
Birg
F
BCL-X and the apoptotic machinery of lymphoma cells.
Leuk Lymphoma.
28
1998
451
458
43
Martin
DA
Zheng
L
Siegel
RM
et al
Defective CD95/APO-1/Fas signal complex formation in the human autoimmune lymphoproliferative syndrome (ALPS), type I.
Proc Natl Acad Sci U S A.
96
1999
5442
5447
44
Jackson
CE
Fischer
RE
Hsu
AP
et al
Autoimmune lymphoproliferative syndrome with defective Fas: genotype influences penetrance.
Am J Human Genet.
64
1999
1002
1014
45
Aftabuddin
M
Yamadori
I
Yoshino
T
Kondo
E
Akagi
T
Correlation between the number of apoptotic cells and expression of the apoptosis-related antigens Fas, Le(y) and bcl-2 protein in non-Hodgkin lymphomas.
Pathol Int.
45
1995
422
429
46
Gruss
HJ
Dower
SK
Tumor necrosis factor ligand superfamily: involvement in the pathology of malignant lymphomas.
Blood.
85
1995
3378
3404
47
Kondo
E
Yoshino
T
Yamadori
I
et al
Expression of Bcl-2 protein and Fas antigen in non-Hodgkin lymphomas.
Am J Pathol.
145
1994
330
337
48
Moller
P
Henne
C
Leithauser
F
et al
Coregulation of the APO-1 antigen with intercellular adhesion molecule-1 (CD43) in tonsillar B cells and coordinate expression in follicular center B cells and in follicle center and mediastinal B-cell lymphomas.
Blood.
81
1993
2067
2075
49
Nguyen
PL
Harris
NL
Ritz
J
Robertson
MJ
Expression of CD95 antigen and Bcl-2 protein in non-Hodgkin's lymphomas and Hodgkin's disease.
Am J Pathol.
148
1996
847
853
50
Plumas
J
Jacob
MC
Chaperot
L
Molens
JP
Sotto
JJ
Bensa
JC
Tumor B cells from non-Hodgkin's lymphoma are resistant to CD95 (Fas/Apo-1)-mediated apoptosis.
Blood.
91
1998
2875
2885
51
Hata
H
Matsuzaki
H
Takeya
M
et al
Expression of Fas/Apo-1 (CD95) and apoptosis in tumor cells from patients with plasma cell disorders.
Blood.
86
1995
1939
1945
52
Massaia
M
Borrione
P
Attisano
C
et al
Dysregulated Fas and Bcl-2 expression leading to enhanced apoptosis in T cells of multiple myeloma patients.
Blood.
85
1995
3679
3687
53
Shima
Y
Nishimoto
N
Ogata
A
Fujii
Y
Yoshizaki
K
Kishimoto
T
Myeloma cells express Fas antigen/APO-1 (CD95) but only some are sensitive to anti-Fas antibody resulting in apoptosis.
Blood.
85
1995
757
764
54
Westendorf
JJ
Lammert
JM
Jelinek
DF
Expression and function of Fas (APO-1/CD95) in patient myeloma cells and myeloma cell lines.
Blood.
85
1995
3566
3576
55
Manolov
G
Manolova
Y
Marker band in one chromosome 14 from Burkitt's lymphomas.
Nature.
237
1972
33
34
56
Juneja
S
Lukeis
R
Tan
L
et al
Cytogenetic analysis of 147 cases of non-Hodgkin lymphoma: non-random chromosomal abnormalities and histological correlations.
Br J Haematol.
76
1990
231
237
57
Offit
K
Wong
G
Filippa
DA
Tao
Y
Chaganti
RS
Cytogenetic analysis of 434 consecutively ascertained specimens of non-Hodgkin lymphoma: clinical correlations.
Blood.
77
1991
1508
1515
58
Offit
K
Jhanwar
SC
Ladanyi
M
Filippa
DA
Chaganti
RS
Cytogenetic analysis of 434 consecutively ascertained specimens of non-Hodgkin lymphoma: correlations between recurrent aberrations, histology, and exposure to cytotoxic treatment.
Genes Chromosomes Cancer.
3
1991
189
201
59
Speaks
SL
Sanger
WG
Masih
AS
Harrington
DS
Hess
M
Armitage
JO
Recurrent abnormalities of chromosome bands 10q23–25 in non-Hodgkin lymphoma.
Genes Chromosomes Cancer.
5
1992
239
243
60
Lichter
P
Walczak
H
Weitz
S
Behrmann
I
Krammer
PH
The human APO-1 (APT) antigen maps to 10q23, a region that is syntenic with mouse chromosome 19.
Genomics.
14
1992
179
180
61
Xerri
L
Carbuccia
N
Parc
P
Birg
F
Search for rearrangements and/or allelic loss of the fas/APO-1 gene in 101 human lymphomas.
Am J Clin Pathol.
104
1995
424
430
62
Zornig
M
Grzeschiczek
A
Kowalski
MB
Hartmann
KY
Moroy
T
Loss of Fas/Apo-1 receptor accelerates lymphomagenesis in E mu L-MYC transgenic mice but not in animals infected with MoMuLV.
Oncogene.
10
1995
2397
2401
63
Cohen
PL
Eisenberg
RA
Lpr and gld: single gene models of systemic autoimmunity and lymphoproliferative disease.
Annu Rev Immunol.
9
1991
243
269
64
Davidson
WF
Giese
T
Fredrickson
TN
Spontaneous development of plasmacytoid tumors in mice with defective Fas-Fas ligand interactions.
J Exp Med.
187
1998
1825
1838
65
Peng
SL
Robert
ME
Hayday
AC
Craft
J
A tumor-suppressor function for Fas (CD95) revealed in T cell-deficient mice.
J Exp Med.
184
1996
1149
1154
66
Fuss
IJ
Strober
W
Dale
JK
et al
Characteristic T helper 2 T cell cytokine abnormalities in autoimmune lymphoproliferative syndrome, a syndrome marked by defective apoptosis and humoral autoimmunity.
J Immunol.
158
1997
1912
1918
67
Huang
B
Eberstadt
M
Olejniczak
ET
Meadows
RP
Fesik
SW
NMR structure and mutagenesis of the Fas (APO-1/CD95) death domain.
Nature.
384
1996
638
641

Drs Howard Britton and Thomas S. DeNapoli informed us of the recent development of Hodgkin disease, mixed cellularity type, in subject 26-IV-5, a 6-year-old boy previously described with ALPS since age 1 (reference 15).

Author notes

Stephen E. Straus, LCI, NIAID, 31 Center Dr, Rm 5B-37, National Institutes of Health, Bethesda, MD 20892; e-mail:sstraus@nih.gov.

Sign in via your Institution