Hairy cell leukemia variant (HCLv) presents with high disease burden, lack of typical antigens like CD25, and poor response to standard treatments like cladribine. Occasionally, patients with classic HCL respond poorly. Clinical and molecular features of HCL and HCLv has not been compared. Rearrangements expressing immunoglobulin VH chain were sequenced, including 22 from 20 patients with HCLv and 63 from 62 patients with classic HCL. Most patients were seeking relapsed/refractory trials, representing a poor-prognosis population. VH4-34, a gene commonly used in autoimmune disorders, was observed in 8 (40%) HCLv and 6 (10%) classic (P = .004) HCL patients. Compared with 71 VH4-34 rearrangements, 14 VH4-34+ rearrangements were more frequently (P < .001) unmutated, defined as greater than 98% homologous to germline sequence. VH4-34+ patients had greater white blood cell counts at diagnosis (P = .002), lower response rate (P < .001) and progression-free survival (P = .007) after initial cladribine, and shorter overall survival from diagnosis (P < .001). Response and survival were more closely related to VH4-34 status than to whether or not patients had HCLv. VH4-34+ HCL is an important disorder that only partly overlaps with the previously described HCLv. Response to initial single-agent cladribine therapy is suboptimal; these patients should be considered for alternative approaches, including antibody-related therapy.

Hairy cell leukemia variant (HCLv) is a B-cell disorder, recognized for nearly 30 years, which accounts for 10% of hairy-cell leukemia (HCL) cases. Morphology of variant cells was reported to be intermediate between that of classic HCL and prolymphocytic leukemias.1-3  Patients typically present with leukocytosis rather than leukopenia and often lack the neutropenia, anemia, and/or thrombocytopenia with which classic HCL patients present.1,2,4  By flow cytometry, B-cell antigens FMC7, CD11c, CD20, CD22, and surface immunoglobulin are strongly positive in both classic HCL and HCLv, whereas HCLv differs from classic HCL by lack of CD25, HC-2, and CD123 and by expression of CD27.3-7  CD103 is usually positive in both but can be negative in HCLv.2  HCLv lacking both CD25 and CD103 may be difficult to differentiate from splenic marginal zone lymphoma (SMZL)/splenic lymphoma with villous lymphocytes without also relying on morphologic differences between HCL and SMZL.2,6,7  In contrast to the high complete remission and overall response rates of classic HCL to the administration of purine analogs pentostatin and cladribine,8-10  response in patients with HCLv is limited to partial responses in approximately 50% of patients.2-4,11,12  Several complete responses of HCLv to monoclonal antibody-based therapy with and without chemotherapy have been reported.13-17 

Like other mature B lymphocytes, the malignant cells in HCL patients have 1 or sometimes 2 different rearrangements for immunoglobulin heavy chain. Significant variability can be observed in the third complementarity determining region (CDR3), comprising the variable heavy (VH), Dbl homology, and junctional heavy domains. We previously reported a molecular characterization of 24 such rearrangements in 23 HCL patients18  and discussed previous studies in which 70 rearrangements in 69 patients were described.19-23  We reported that of 4 patients in our series with unmutated rearrangements, defined as greater than 98% homology to germline sequence, 3 presented with high tumor burden consistent clinically with variant disease. However, the CD25/VH status in these 3 patients was CD25/VH4-34+, CD25/VH4-34, and CD25+/VH4-34+,18  suggesting that VH4-34 expression and adverse clinical behavior may not be confined to HCLv diagnosed by immunophenotype. A more recent VH study in HCL reported that 5 of 38 cases were HCLv, 2 of which were unmutated VH4-34.24  Another study reported that 5 of 83 cases were HCLv, and 5 classic cases were VH4-34+, 3 of which were unmutated.25  To better understand the association of VH4-34 and other VH genes with the variant immunophenotype and to determine whether molecular features could be prognostically important, independent of the diagnosis of classic HCL or HCLv, we studied immunoglobulin rearrangements and clinical factors in 82 HCL patients, 20 of whom had HCLv.

Patients and controls

Blood for DNA study was obtained as part of sample acquisition protocols with informed consent approved by the NCI Investigator's Review Board and in accordance with the Declaration of Helsinki. All samples were retrieved between 2001 and 2008. Of the 85 rearrangements in 82 patients examined, 24 rearrangements in 23 patients were published previously.18  Diagnoses of classic HCL and HCLv were rendered by a hematopathologist (M.S.-S.), based upon morphology and immunophenotype and according to World Health Organization classification.26 

HCL was characterized by a proliferation of medium-sized lymphoid cells with mature chromatin; circumferential hairy-like cytoplasmic projections; reticulin fibrosis and interstitial infiltration in bone marrow; tartrate-resistant acid phosphatase (TRAP) positivity; bright coexpression of CD20, CD22, and CD11c; and expression of CD25, CD103, and CD123. HCLv was characterized by a proliferation of abnormal lymphoid cells varying from medium to large in size, often with prominent nucleoli and irregular nuclear contours, with hairy-like cytoplasmic projections. Like classic HCL, HCLv usually demonstrated bright coexpression of CD20, CD22, and CD11c and expression of CD103 but was negative for CD25 and frequently CD123.2,5,26  TRAP, when available, usually was positive. SMZL, which was excluded from this study, was diagnosed based upon morphology and immunophenotype, namely relatively small lymphoid cells with occasional cytoplasmic projections in polar orientation; moderate coexpression of CD20, CD22, and dim-to-moderate CD11c; and negativity for TRAP, CD103, and CD123.2,5,26  For each of the 82 patients assigned a diagnosis of HCL, the leukemic cells were examined in the same National Institutes of Health (NIH) laboratory for morphology and flow cytometry. In addition, bone marrow biopsy samples were evaluated at NIH for 63 patients, and bone marrow biopsy reports were obtained for 17 others. Of 32 asplenic patients, in 5 the spleen tissue was examined at NIH, and the pathology reports were obtained for 13 others.

cDNA synthesis and characterization of rearrangements

Total RNA was extracted from PAXgene Blood RNA tubes (PreAnalytiX GmbH) containing peripheral-blood mononuclear cells by use of the PAXgene Blood RNA kit (PreAnalytiX GmbH). Total RNA from CD11c-sorted cells was purified by use of the MagMax kit (Ambion). First-strand cDNA synthesis was performed by SuperScriptTM III RnaseH-Reverse Transcriptase (Invitrogen) from 1 to 3 μg of total RNA in reaction mix in the presence of Oligo(dT)20 primer (Invitrogen) and 10 mmol/L dNTP mix according to the manufacturer's protocol as described previously.18  cDNA was amplified in a single multiplexed polymerase chain reaction (PCR) consisting of 6 VH framework 1 primers combined with 1 JH consensus primer as designed for the BIOMED-2 study.27 

All reactions were carried out in 100 μL containing 10 pmol of each primer (InVivoScribe Technologies), 200 μmol/L dNTPs, 0.5 U AmpliTaq Gold, and 10X PCR buffer II (Applied Biosystems). The DNA was amplified as follows: denaturation at 95°C for 7 minutes, 37 cycles of 94°C for 1 minute, 55°C for 30 seconds, 72°C for 1 minute, with a final extension of 10 minutes at 72°C. PCR products were analyzed on a 2% agarose gel and visualized with ethidium bromide staining, purified by the use of QIAquick PCR purification kit (QIAGEN), and cloned by the use of the Zero Blunt TOPO PCR Cloning kit (Invitrogen). Cloned PCR products were sequenced by the use of T3 consensus primers and Big-Dye terminators (Applied Biosystems). A monoclonal sequence was defined when at least 5 of 10 clones had a completely identical sequence. Intraclonal and ongoing somatic hypermutation were not observed. No 2 rearrangements from different patients were identical, which was evidence against any being a contaminant. The structure of each rearrangement was determined by sequence analysis by use of the IMGT database28  and the VBASE2 database.29 

Cell purification with CD11c sorting

Peripheral-blood mononuclear cells were purified from 25 mL of heparin-containing peripheral blood of HCL patients by Ficoll-Paque Plus (GE Healthcare Bio-Science AB) by use of the manufacturer's protocol and then counted. The peripheral B-cell population was isolated by the Dynal B-cell–negative isolation kit (Invitrogen Dynal AS) and incubated with 20 μL of Fc receptor blocking reagent (Miltenyi Biotec) for 10 minutes at 4°C. One microgram of CD11c antibody (Santa Cruz Biotechnology) per 106 cells was added to the sample and incubated on ice for 1 hour. Cells were washed by adding 2 mL of 0.1% bovine serum albumin containing phosphate-buffered saline and centrifuged to 1200 rpm for 8 minutes at 4°C. The cells were then incubated with goat anti–mouse IgG Dynabeads (Invitrogen Dynal AS) for 15 minutes at 4°C with gentle tilting and rotation followed by magnetic capture of the CD11c-positive population, followed by addition of TRI Reagent (Ambion Inc) for future RNA purification.

Statistics

Comparisons between groups of patients were performed by the Fisher exact test or by Wilcoxon rank sum test as appropriate. Kaplan-Meier curves were compared by log-rank, and median survival of a population was considered 50% probability of survival. Calculations were performed using SAS, and P values presented were 2-sided.

Assessment of response to initial treatment with cladribine

Patients with sufficient medical records, including dates of treatment, complete blood counts, and bone marrow biopsy reports, were considered evaluable for retrospective response assessment, providing their first purine analog treatment was cladribine. Best response was considered after just the first course of cladribine and was determined on the basis of published criteria for HCL,9  except that hemoglobin had to reach 11 rather than 12 for a complete remission. Also, in patients who were red blood cell transfusion dependent, the 50% improvement in hemoglobin needed for partial response was assumed if the hemoglobin reached 9 g/dL without transfusions. Because bone marrow biopsy was not always used after cladribine, we also determined whether patients achieved hematologic remission, defined as meeting all criteria for complete remission except for the bone marrow biopsy. Date of progression was defined as the first documentation of relapse from partial response in responding patients or progressive disease in nonresponding patients.

Patient characteristics

Of 82 patients with HCL, 62 (76%) were considered classic HCL, whereas 20 (24%) were considered HCLv. Immunophenotypic characteristics are shown in Table 1 for the 26 patients who had either HCLv or VH4-34+ rearrangements or both. The remaining 56 classic HCL patients are summarized in Table 2. HCL cells from all patients expressed the B-cell markers CD19, CD20 (bright in 100%), and CD22 (bright in 98%), as well as CD11c (bright in 96%). HCL cells from all classic patients expressed CD25, which was bright in 44 (71%), positive in 17 (27%), and in 1 (2%) patient varied from bright to negative. In contrast, the HCLv cells were dim to negative or negative for CD25. CD123 was expressed homogenously in 31 (100%) of 31 classic patients and in 3 (38%) of 8 HCLv patients evaluated, and 1 HCLv patient had partial CD123 expression. CD103 was expressed homogenously in 77 (94%) of the 82 patients. Two patients with classic HCL had partial CD103 expression, 1 of whom had previously documented homogenous CD103 expression. The 3 CD103 patients also were CD25 but had either TRAP+ or CD11c bright-positive cells consistent with HCLv rather than SMZL. Patient BL26 had partial positive CD11c expression with lack of CD25 and CD103, suggesting SMZL. However, the splenic pathology showed red pulp rather than a white pulp infiltration, and positivity for both TRAP and DBA.44 were more consistent with HCLv than with SMZL.2,5,30  Lymphoplasmacytic lymphoma was considered but the morphology, the bright expression of CD22 and CD20, and the lack of CD25 were against this diagnosis.31  One patient previously diagnosed as HCLv was diagnosed with SMZL on the basis of flowcytometry and morphology and was therefore not included in the 82 patients reported. The male/female ratio was 74:8 and not different with respect to variant status. Age from diagnosis was older for HCLv than for classic HCL (P = .02), with median age 55 versus 46 years.

Table 1

Immunophenotypic and molecular characteristics of patients with VH4-34 or HCLv (n = 26)

Patient no.AgeSexCD19CD20CD22CD25CD11CCD5CD10CD23FMC7CD103CD123LCTRDxVH
BH18 57 ++ +++ +++ +++ +++ − − − ++ ++ NA κ HCL VH4-34 
BH32 58 ++ +++ +++ +++ +++ NA NA NA NA ++ ++ λ NA HCL VH4-34 
BH34 56 ++ +++ +++ P+ +++ − − − +++ NA λ HCL VH4-34 
BN27 38 ++ +++ +++ +++ +++ − − − ++ ++ NA κ HCL VH4-34 
BN29 55 ++ +++ +++ +++ +++ − − − ++ ++ NA λ NA HCL VH4-34 
HH14 50 ++ +++ +++ ++ +++ − − − ++ ++ ++ κ − HCL VH4-34 
BH16 52 ++ +++ +++ − +++ − − − ++ NA κ HCLv VH4-34 
BH28 42 ++ +++ +++ − +++ − − − ++ ++ NA λ HCLv VH4-34 
BH29 55 ++ +++ +++ +/− +++ − − − ++ ++ ++ λ HCLv VH4-34 
BL18 69 ++ +++ +++ − +++ − − − ++ ++ NA κ HCLv VH4-34 
BL26 55 ++ +++ ++ − P+ − − − ++ − NA κ HCLv VH4-34 
BN15 61 ++ +++ +++ − +++ − − ++ ++ − κ NA HCLv VH4-34 
BN21 71 ++ +++ +++ − +++ − − − ++ − NA κ NA HCLv VH4-34 
BN35 55 ++ +++ +++ − +++ − − − ++ ++ − κ NA HCLv VH4-34 
HH10 40 ++ +++ +++ − +++ − − − ++ ++ − κ HCLv VH1-18 
BN17 72 +++ +++ +++ − +++ − − − ++ ++ NA λ NA HCLv VH3-23 
BN20 50 ++ +++ +++ − +++ − − − ++ ++ NA κ HCLv VH3-48 
BN23 35 ++ +++ +++ − +++ − − − ++ ++ P+ λ NA HCLv VH3-66 
BL14 38 ++ +++ +++ +/− +++ − − − ++ ++ κ HCLv VH4-31 
BN16 63 ++ +++ +++ − +++ − − − ++ ++ NA λ HCLv VH4-31 
BN22 55 ++ +++ ++ +/− ++ − − − ++ ++ NA λ HCLv VH4-39 
BN39 77 ++ +++ +++ − +++ − − − ++ ++ κ NA HCLv VH4-39 
BN24 73 ++ +++ +++ − ++ − − − ++ ++ NA − NA HCLv VH4-61 
BN04 57 ++ +++ +++ − +++ P+ − − ++ − NA κ HCLv VH4-61 
HH19 44 ++ +++ +++ − +++ − − − ++ ++ NA κ NA HCLv VH5-a 
BN19 37 ++ +++ +++ − +++ − − − ++ ++ − κ NA HCLv VH6-1 
Patient no.AgeSexCD19CD20CD22CD25CD11CCD5CD10CD23FMC7CD103CD123LCTRDxVH
BH18 57 ++ +++ +++ +++ +++ − − − ++ ++ NA κ HCL VH4-34 
BH32 58 ++ +++ +++ +++ +++ NA NA NA NA ++ ++ λ NA HCL VH4-34 
BH34 56 ++ +++ +++ P+ +++ − − − +++ NA λ HCL VH4-34 
BN27 38 ++ +++ +++ +++ +++ − − − ++ ++ NA κ HCL VH4-34 
BN29 55 ++ +++ +++ +++ +++ − − − ++ ++ NA λ NA HCL VH4-34 
HH14 50 ++ +++ +++ ++ +++ − − − ++ ++ ++ κ − HCL VH4-34 
BH16 52 ++ +++ +++ − +++ − − − ++ NA κ HCLv VH4-34 
BH28 42 ++ +++ +++ − +++ − − − ++ ++ NA λ HCLv VH4-34 
BH29 55 ++ +++ +++ +/− +++ − − − ++ ++ ++ λ HCLv VH4-34 
BL18 69 ++ +++ +++ − +++ − − − ++ ++ NA κ HCLv VH4-34 
BL26 55 ++ +++ ++ − P+ − − − ++ − NA κ HCLv VH4-34 
BN15 61 ++ +++ +++ − +++ − − ++ ++ − κ NA HCLv VH4-34 
BN21 71 ++ +++ +++ − +++ − − − ++ − NA κ NA HCLv VH4-34 
BN35 55 ++ +++ +++ − +++ − − − ++ ++ − κ NA HCLv VH4-34 
HH10 40 ++ +++ +++ − +++ − − − ++ ++ − κ HCLv VH1-18 
BN17 72 +++ +++ +++ − +++ − − − ++ ++ NA λ NA HCLv VH3-23 
BN20 50 ++ +++ +++ − +++ − − − ++ ++ NA κ HCLv VH3-48 
BN23 35 ++ +++ +++ − +++ − − − ++ ++ P+ λ NA HCLv VH3-66 
BL14 38 ++ +++ +++ +/− +++ − − − ++ ++ κ HCLv VH4-31 
BN16 63 ++ +++ +++ − +++ − − − ++ ++ NA λ HCLv VH4-31 
BN22 55 ++ +++ ++ +/− ++ − − − ++ ++ NA λ HCLv VH4-39 
BN39 77 ++ +++ +++ − +++ − − − ++ ++ κ NA HCLv VH4-39 
BN24 73 ++ +++ +++ − ++ − − − ++ ++ NA − NA HCLv VH4-61 
BN04 57 ++ +++ +++ − +++ P+ − − ++ − NA κ HCLv VH4-61 
HH19 44 ++ +++ +++ − +++ − − − ++ ++ NA κ NA HCLv VH5-a 
BN19 37 ++ +++ +++ − +++ − − − ++ ++ − κ NA HCLv VH6-1 

Dx indicates diagnosis; HCL, classic hairy-cell leukemia; HCLv, variant HCL; LC, light chain; NA, not available; P+, part positive; and TR, tartrate-resistant acid phosphatase stain.

Table 2

Immunophenotypic and molecular characteristics of patients with non–VH4-34 HCL (n = 56)

CD19CD20CD22CD25CD11CCD5CD10CD23FMC7CD103CD123
Evaluable 56 56 56 56 56 50 49 52 52 56 29 
Bright + (+++) 56 56 40 56      
Positive (++) 54   16   50 54 27 
Dim + (+)         
Dim to negative (+/−)          
Part positive (P+)        
Negative (−)      44 44 46    
CD19CD20CD22CD25CD11CCD5CD10CD23FMC7CD103CD123
Evaluable 56 56 56 56 56 50 49 52 52 56 29 
Bright + (+++) 56 56 40 56      
Positive (++) 54   16   50 54 27 
Dim + (+)         
Dim to negative (+/−)          
Part positive (P+)        
Negative (−)      44 44 46    

One patient had monophasic light chain. Median patient age = 45 years; male/female = 50:6; κ/λ light chain = 25:31; TRAP +:− = 38:1.

HCL indicates classic hairy cell leukemia; and TRAP, tartrate-resistant acid phosphatase stain.

VH usage

To investigate immunoglobulin VH usage in classic and variant HCL, a total of 63 rearrangements in 62 classic HCL and 22 rearrangements in 20 HCLv patients were amplified, sequenced, and analyzed. As shown in Figure 1A, VH3-family VH chains were more common in classic HCL than HCLv (57% vs 23%, P = .007), and VH4 was more common in HCLv than in classic disease (64% vs 25%, P = .002). The usage in classic HCL is similar to the reported32  usage of VH families 1 to 7 in 206 rearrangements from normal (CD5/IgM+) B cells, namely 13.1%, 1.9%, 53.9%, 24.8%, 2.9%, 2.4%, and 1%, respectively. However, in HCLv, VH3 usage was lower (P = .007) and VH4 usage was greater (P < .001) compared with normal B cells.

Figure 1

VH use frequency in HCL. A, V families; B, VH gene usage. Patients had classic (back bars) or variant (shaded bars) HCL.

Figure 1

VH use frequency in HCL. A, V families; B, VH gene usage. Patients had classic (back bars) or variant (shaded bars) HCL.

Close modal

VH gene usage

When further subdivided into individual genes within each family, a total of 30 different VH genes were cloned. Half (15/30) of these occurred in more than 1 patient, and their frequencies are shown in Figure 1B. The most frequently used classic VH genes were VH3-23 (n = 13, 21%), VH4-34 (n = 6, 10%), and VH3-30 (n = 5, 8%). Of the 3 patients with 2 rearrangements each, 1 with HCLv expressed both VH3-66 and VH3-30-3, 1 with HCLv expressed both VH4-39 and VH3-30, and 1 with classic HCL expressed both VH1-2 and VH3-74. The most frequently used variant VH gene was VH4-34 (n = 8, 36%), and the largest difference between classic and variant rearrangements was observed with incidence of VH4-34 (10% vs 36%, P = .007). Thus, VH4-34 was more common in but not confined to HCLv.

Somatic hypermutation

To determine whether the frequency of somatic mutations in the VH rearrangements differed with respect to variant disease or VH4-34 expression, they were cloned and compared with the IMGT database. Figure 2 shows homology to germline sequence for the 11 different rearrangements observed in at least 3 patients each. All but 1 VH4-34+ rearrangement was unmutated compared with 15% of the others (P < .001). Homology to germline sequences was greater for VH4-34+ compared with VH4-34 rearrangements (median homology 99.4% vs 95.6%, P < .001). In contrast, there was no relationship between homology and variant versus classic phenotype (97.6% vs 95.9%, P = .12), despite the relationship between variant phenotype and VH4-34 positivity. In fact, when considering the 22 variant and 63 classic rearrangements separately, VH4-34 positivity in each group is still associated with increased homology (Table 3, P = .006-.02). Thus, VH4-34 expression is associated with high homology to germline sequence unrelated to diagnosis of HCLv.

Figure 2

Homology to germline sequence for rearrangements with the indicated VH gene usage and combined data from 71 non–VH4-34–expressing rearrangements. Error bars indicate SDs from the means of the points shown. Patients with classic (○) or variant (●) hairy cells are shown.

Figure 2

Homology to germline sequence for rearrangements with the indicated VH gene usage and combined data from 71 non–VH4-34–expressing rearrangements. Error bars indicate SDs from the means of the points shown. Patients with classic (○) or variant (●) hairy cells are shown.

Close modal
Table 3

Characteristics at diagnosis

VH4-34 status and variant phenotypeVH4-34+VH4-34PHCLvHCLP
Median age at diagnosis (n) 55.0 (14) 45.6 (68) .011 54.8 (20) 45.7 (62) .016 
Median, homology, % (n) 99.4 (14) 95.6 (71) < .0001 97.6 (22) 95.9 (63) .12 
Median homology, %, classic HCL only (n) 98.7 (6) 95.6 (57) .006    
Median homology, %, HCLv only (n) 99.6 (8) 95.7 (14) .02    
White blood cell count, > 5 × 109/L (%) 13/14 (93) 23/57 (40) .0006 15/18 (83) 21/53 (40) .002 
White blood cell count, > 5 × 109/L, classic HCL only (%) 6/6 (100%) 15/47 (32%) .002    
White blood cell count > 5 × 109/L, VH4-34 only (%)    8/10 (80%) 15/47 (32%) .01 
Median white blood cell count, ×109/L (n) 9.0 (14) 3.8 (57) .002 11.3 (18) 3.5 (53) < .0001 
Median white blood cell count, ×109/L, classic HCL only (n) 10.5 (6) 2.9 (47) .003    
Median white blood cell count, ×109/L, VH4-34 only (n)    13.8 (10) 2.9 (47) .0003 
Median neutrophils, ×109/L (n) 2.23 (12) 0.82 (49) .004 3.01 (14) 0.77 (47) < .0001 
Median hemoglobin, g/dL 13.2 (13) 11.6 (52) .12 13.2 (17) 11.4 (48) .006 
Median platelets, ×109/L (n) 105 (13) 68 (54) .014 135 (16) 65 (51) .0001 
VH4-34 status and variant phenotypeVH4-34+VH4-34PHCLvHCLP
Median age at diagnosis (n) 55.0 (14) 45.6 (68) .011 54.8 (20) 45.7 (62) .016 
Median, homology, % (n) 99.4 (14) 95.6 (71) < .0001 97.6 (22) 95.9 (63) .12 
Median homology, %, classic HCL only (n) 98.7 (6) 95.6 (57) .006    
Median homology, %, HCLv only (n) 99.6 (8) 95.7 (14) .02    
White blood cell count, > 5 × 109/L (%) 13/14 (93) 23/57 (40) .0006 15/18 (83) 21/53 (40) .002 
White blood cell count, > 5 × 109/L, classic HCL only (%) 6/6 (100%) 15/47 (32%) .002    
White blood cell count > 5 × 109/L, VH4-34 only (%)    8/10 (80%) 15/47 (32%) .01 
Median white blood cell count, ×109/L (n) 9.0 (14) 3.8 (57) .002 11.3 (18) 3.5 (53) < .0001 
Median white blood cell count, ×109/L, classic HCL only (n) 10.5 (6) 2.9 (47) .003    
Median white blood cell count, ×109/L, VH4-34 only (n)    13.8 (10) 2.9 (47) .0003 
Median neutrophils, ×109/L (n) 2.23 (12) 0.82 (49) .004 3.01 (14) 0.77 (47) < .0001 
Median hemoglobin, g/dL 13.2 (13) 11.6 (52) .12 13.2 (17) 11.4 (48) .006 
Median platelets, ×109/L (n) 105 (13) 68 (54) .014 135 (16) 65 (51) .0001 

HCL indicates classic hairy-cell leukemia; and HCLv, variant HCL.

Leukopenia in VH4-34+ and VH4-34 HCL

To determine whether VH4-34 positivity was associated with adverse clinical factors, those clinical factors associated with HCLv were investigated. Leukocytosis at presentation, a known characteristic of HCLv, is attributed to both absence of neutropenia and increase in circulating malignant cells.3,7  Absence of leukopenia, specifically a presenting white blood cell count of at least 5 × 109/L, was more frequently observed in VH4-34+ than VH4-34 patients (93% vs 40%, P < .001; Table 3). Although a greater proportion of variant than classic patients reached this white blood cell count at diagnosis (83% vs 40%, P = .002), VH4-34 positivity was independently related because among just classic patients a greater percentage had white blood cell counts of at least 5 × 109/L (100% vs 32%, P = .002, Table 3). As shown in Figure 3, by rank order analysis, presenting white blood cell counts were greater for VH4-34+ versus negative patients (median 9.0 vs 3.8 × 109/L, P = .002). Although the same was true of variant versus classic patients (median 11.3 vs 3.5 × 109/L, P < .001), among just classic patients, VH4-34 positivity correlated with presenting white blood cell count (P = .003). Thus, absence of leukopenia is a feature that correlates independently with both VH4-34 positivity and with HCLv. As shown in Table 3, presenting neutrophils, hemoglobin, and platelets were significantly greater with variant compared with classic disease, and although neutrophil and platelet counts were significantly greater with VH4-34+ than VH4-34 patients (P = .004-.014), VH4-34 status was not independently related to levels of these normal cells. Thus, VH4-34 positivity was correlated with lack of leukopenia, probably because of increased malignant rather than increased normal cells.

Figure 3

White blood cells counts at diagnosis in patients with or without VH4-34 expression, whose hairy cells are classic (○) or variant (●) HCL.

Figure 3

White blood cells counts at diagnosis in patients with or without VH4-34 expression, whose hairy cells are classic (○) or variant (●) HCL.

Close modal

VH4-34 positivity and failure of initial cladribine therapy in patients with HCL

To determine whether VH4-34 positivity was associated with the failure of initial cladribine to achieve remission, an important characteristic of HCLv,3  we studied HCL patients who were being screened for recombinant immunotoxin trials that required inadequate response to initial or most recent purine analog therapy (Table 4). Historical data on each patient were analyzed to determine complete, hematologic, or partial remission on the basis of published criteria.9  Hematologic remission, which requires all the criteria of complete remission except for bone marrow biopsy, was important to consider because many patients did not get bone-marrow studies after cladribine. As shown in Table 5, complete, hematologic, and partial remission rates were significantly lower with VH4-34+ compared with VH4-34 patients (P = .03, .004, and < .001, respectively).

Table 4

Outcome after initial cladribine for HCL (patient numbers)

TotalVH4-34+, HCLvVH4-34+, HCLVH4-34, HCLvVH4-34, HCL
Patients evaluable for survival 82 12 56 
Patients evaulable for response 61 40 
Complete remission 14 13 
Hematologic remission 18 14 
Partial response 10 
No response 19 
TotalVH4-34+, HCLvVH4-34+, HCLVH4-34, HCLvVH4-34, HCL
Patients evaluable for survival 82 12 56 
Patients evaulable for response 61 40 
Complete remission 14 13 
Hematologic remission 18 14 
Partial response 10 
No response 19 

HCL indicates classic hairy-cell leukemia; and HCLv, variant HCL.

Table 5

Outcome after initial cladribine for HCL (response rates, progression-free, and overall survival)

Response comparisonsVH4-34+VH4-34PHCLvHCLP
Complete remission rate (%) 0/13 (0) 14/48 (29) .03 1/15 (7) 13/46 (28) .15 
Complete/hematologic remission rate (%) 2/13 (15) 30/48 (63) .004 3/15 (20) 29/46 (63) .006 
Major response rate (%) 2/13 (15) 40/48 (83) .00001 4/15 (27) 38/46 (83) .0001 
Median progression-free survival, mo (n) 5.1 (13) 22.8 (48) .007 5.1 (15) 23.1 (46) .02 
Median overall survival, y (n) 8.63 (14) 26.22 (68) < .0001 >8.79 (20) 26.22 (62) .02 
Major response rate for classic HCL only (%) 2/6 (33) 36/40 (90) .006    
Major response rate for those VH4-34 only (%)    4/8 (50) 36/40 (90) .02 
Median overall survival, y, classic HCL only (n) 4.70 (6) 26.64 (56) < .0001    
Median overall survival, y, VH4-34 only (n)    >4.74 (12) 26.64 (56) .22 
Median age at diagnosis, age > 50 y (n) 55.75 (12) 57.00 (24) .31 60.8 (13) 56.70 (23) .1 
Median overall survival, age > 50 y (n) 8.63 (12) 21.05 (24) .0001 8.79 (13) 16.53 (23) .11 
Median overall survival, age > 50 y, classic HCL only 4.70 (5) 17.33 (18) < .0001    
Median overall survival, age > 50 y, VH4-34 only    21.05 (6) 17.33 (18) .23 
Response comparisonsVH4-34+VH4-34PHCLvHCLP
Complete remission rate (%) 0/13 (0) 14/48 (29) .03 1/15 (7) 13/46 (28) .15 
Complete/hematologic remission rate (%) 2/13 (15) 30/48 (63) .004 3/15 (20) 29/46 (63) .006 
Major response rate (%) 2/13 (15) 40/48 (83) .00001 4/15 (27) 38/46 (83) .0001 
Median progression-free survival, mo (n) 5.1 (13) 22.8 (48) .007 5.1 (15) 23.1 (46) .02 
Median overall survival, y (n) 8.63 (14) 26.22 (68) < .0001 >8.79 (20) 26.22 (62) .02 
Major response rate for classic HCL only (%) 2/6 (33) 36/40 (90) .006    
Major response rate for those VH4-34 only (%)    4/8 (50) 36/40 (90) .02 
Median overall survival, y, classic HCL only (n) 4.70 (6) 26.64 (56) < .0001    
Median overall survival, y, VH4-34 only (n)    >4.74 (12) 26.64 (56) .22 
Median age at diagnosis, age > 50 y (n) 55.75 (12) 57.00 (24) .31 60.8 (13) 56.70 (23) .1 
Median overall survival, age > 50 y (n) 8.63 (12) 21.05 (24) .0001 8.79 (13) 16.53 (23) .11 
Median overall survival, age > 50 y, classic HCL only 4.70 (5) 17.33 (18) < .0001    
Median overall survival, age > 50 y, VH4-34 only    21.05 (6) 17.33 (18) .23 

HCL indicates classic hairy cell leukemia; and HCLv, variant HCL.

Hematologic and partial remission rates also were lower for variant compared with classic patients. The lower-than-reported observed remission rates for classic patients in the current study were expected given the skewed patient population studied. Among just classic HCL patients, VH4-34 positivity correlated significantly with lower response (P = .006, Table 5), indicating its independence from variant versus classic phenotype. The relationship of variant phenotype to response in just VH4-34 patients had less significance (P = .02). As shown in Figure 4A and B, shorter progression-free survival was associated with VH4-34 positivity (P = .007) and HCLv (P = .02), with median progression-free survivals of 5.1 versus 22.8 months depending on VH4-34 status, and 5.1 versus 23.1 months depending on HCL variant versus classic (Table 5). Thus, response rate and duration after initial cladribine was more significantly related to VH4-34 status than to variant phenotype.

Figure 4

Progression-free survival and overall survival in patients with respect to VH4-34 status and variant phenotype. (A-B) Progression-free survival; (C-F) overall survival. Median values are listed in Table 5. Panels C and D show data for all 82 patients with respect to VH4-34 (C) or variant (D) status, whereas panel E includes only classic HCL and panel F only VH4-34 patients.

Figure 4

Progression-free survival and overall survival in patients with respect to VH4-34 status and variant phenotype. (A-B) Progression-free survival; (C-F) overall survival. Median values are listed in Table 5. Panels C and D show data for all 82 patients with respect to VH4-34 (C) or variant (D) status, whereas panel E includes only classic HCL and panel F only VH4-34 patients.

Close modal

VH4-34 positivity and overall survival

Because lower overall survival was reported in HCLv than in classic HCL,3  dates of death and diagnosis were examined to compare survival after diagnosis relative to VH4-34 status or variant phenotype. As shown in Table 5 and Figure 4C, VH4-34 positivity was significantly correlated with poorer survival, with predicted median survival 8.63 years in VH4-34+ versus 26.22 years in VH4-34 patients (P < .001). A difference of more than 8.79 versus 26.22 years was observed for variant versus classic patients (Figure 4D, P = .02). As shown in Table 5, VH4-34 status was independent of variant phenotype in correlating with poorer overall survival (P < .001). In contrast, overall survival was not significantly associated with variant phenotype in only VH4-34 patients (P = .22). Of the 8 VH4-34+ patients in this study who died, in 7 the immediate cause of death was HCL along with infectious complications. The eighth patient had significant HCL tumor burden and died of a massive stroke, despite being young in age (63 years) and having an absence of carotid atherosclerotic disease. Therefore, an association of thromboembolic disease to his HCL could not be excluded.

VH4-34 positivity and overall survival in older HCL patients

As was observed in comparing variant to classic patients, VH4-34+ patients were older than VH4-34 patients at diagnosis (median ages 55.0 vs 45.6, P = .011, Table 3). To determine whether lower overall survival in VH4-34+ patients was related to their older age at diagnosis, patients older than the age of 50 were considered separately, because in those 36 older patients, there was no significant difference in age with respect to VH4-34 status (P = .31) or variant phenotype (P = .1; Table 5). In these older patients, overall survival was still significantly shorter for VH4-34+ than VH4-34 patients (median 8.63 vs 21.05 years, P < .001) but not for HCLv compared with classic patients (median 8.79 vs 16.53 years, P = .11). As shown in Table 5, VH4-34 status was independent of variant phenotype in its association with poorer survival in older patients (P < .001), but variant phenotype was not associated with poor survival independent of VH4-34 status (P = .23). Thus, VH4-34 positivity was associated with lower survival from diagnosis, independent of age or variant phenotype. Moreover, lower survival of the HCLv patients was the result of VH4-34+ patients with HCLv, rather than to an independent association between HCLv and survival.

Our original goal was to study molecular characteristics of HCLv to determine whether expressed immunoglobulin rearrangements have homology to germline and gene usage different from the “classic” or typical form of the disease. We found that VH4-34, the most common VH gene used in HCLv, had very high homology to its germline sequence and was associated with variant features such as advanced age, lack of leukopenia, poor response to initial cladribine treatment, and poor progression-free and overall survival. Moreover, these associations were observed even in patients with VH4-34+ hairy cells who are considered classic HCL rather than HCLv. Conversely, comparisons of response and overall survival between classic and variant patients lost significance when only the VH4-34 population was examined. Thus, VH4-34+ HCL is a disorder, overlapping only partially with HCLv, associated with variant features and poor prognosis. It has long been thought that patients with HCL not responding to initial purine analog treatment actually have a different disease. These results indicate that VH4-34+ HCL is a disorder accounting for many of these patients and may account for a component of the poor outcome previously reported in HCLv.

Molecular versus immunophenotype in HCL

Each of the 82 patients in this study was carefully examined to verify the diagnosis of HCL or HCLv by the use of flow cytometry, immunohistochemistry, and morphology of blood, bone marrow, spleen, and lymph nodes when available. Nevertheless, because of significant overlap in criteria, in some patients a perfect distinction between SMZL, HCLv, and HCL may not be possible. The frequent loss of antigen expression over time, as observed in this study, may add to the unreliability of available methods to classify patients with HCL and predict response to standard cladribine therapy. Our results show that unmutated VH4-34 positivity is a fixed marker in HCL which predicts for poor outcome, both response to initial therapy and survival, and this appears independent of the exact diagnosis within HCL.

VH4-34 expression in malignant lymphocytes

VH4-34 is expressed in approximately 5% to 10% of adult normal B cells,33-36  65% of diffuse large-cell lymphoma,37  25% of duodenal follicular lymphoma,38  50% of primary central nervous system lymphoma,39  4% to 22% of chronic lymphocytic leukemia (CLL),40,41  9% to 33% of acute lymphoblastic leukemia,40,42  and almost never in multiple myeloma.43-45  We and others have reported VH4-34 expression in isolated cases of HCLv and classic HCL,18,24,25  but the 2 to 5 cases per study were insufficient for understanding the impact of VH4-34 in this disease.

Relation between VH4-34 and poor prognosis

VH4-34 is reported to be overexpressed in autoimmune disease, including 55% of systemic lupus erythematosus,46  virtually all cases of cold agglutinin disease,47-52  and 89% of multiple sclerosis.53  Monoclonal antibodies produced by VH4-34–expressing B cells are reported to react to a variety of normal antigens, including red blood cells,46-49  normal and malignant B cells,50-52,54,55  and single-stranded DNA.56  Although percent homology to germline is rarely determined for VH4-34 clones binding to autoantigens, mutated and less-commonly unmutated VH4-34 rearrangements are reported, the latter from either human fetal spleen or cerebral spinal fluid in multiple sclerosis.49,57-60  It is proposed that VH4-34–expressing B cells may recognize antigen, undergo somatic mutation, and then either mediate autoimmune disease or transform to CLL or diffuse large-cell lymphoma, both known for frequent autoimmune complications.

In contrast, malignant transformation of such cells at a much later point of differentiation to multiple myeloma does not occur because of eventual clonal deletion or anergy.43  We and others18  recently reported similarities between classic HCL and SMZL regarding VH usage and rates of somatic mutation, suggesting that classic hairy cells undergo malignant transformation in the marginal zone of the germinal center. The unmutated status of VH4-34+ hairy cells in all but 1 of our 14 patients suggests transformation before the germinal center and is consistent with the complete lack of autoimmune complications in our patients. We therefore speculate that VH4-34+ lymphocytes in general may be less susceptible to checkpoints leading to clonal deletion, which leads to aggressive and resistant disease in hairy cell patients, or to the autoimmune complications seen in patients with other disorders.

Other factors potentially affecting prognosis

It has been reported that unmutated rearrangements in CLL correlate with poor prognosis.61  To determine whether poor prognosis in HCL is more closely related to unmutated than to VH4-34+ rearrangements, we compared just the VH4-34 rearrangements for progression free (n = 50) and overall (n = 71) survival but found no significant difference whether they were mutated or unmutated (P = .3-.6). A total of 36 patients had abdominal imaging data before the initial cladribine, and enlarged lymph nodes, associated with lower repose to purine analog,62-64  were observed in 2 of 2 VH4-34+/HCL classic, 0 of 5 VH4-34+/HCLv, 2 of 9 VH4-34/HCLv, and 3 of 20 VH4-34/HCL classic patients. These data are insufficient to suggest that VH4-34 positivity is associated with the adenopathy observed in patients with poor prognosis for response.

Clinical implications of VH4-34+ HCL

Except for 1 VH4-34+ HCL patient who had a mutated rearrangement and hematologic remission, the other patients had unmutated VH4-34 rearrangements and poorer response to initial cladribine therapy (Table 2, Figure 2). Like HCLv, VH4-34+ HCL may represent a disorder distinct from classic HCL and overlapping with HCLv. Of those VH4-34 patients who lived to try alternative treatments, 3 had complete remission to recombinant anti-CD22 immunotoxin BL22,17,65  and 2 patients had hematologic remission to combined pentostatin and rituximab. HCL patients sometimes receive pentostatin alone after failure of cladribine because of anecdotal reports of complete remission to cladribine64,66-68  or pentostatin69  alone after failure of the other purine analog. However, of 4 VH4-34+ patients in this study who received pentostatin alone after cladribine, none responded. In view of the observation that the remission rate decreases with each course of purine analog equally whether patients receive the same or the other agent,70  it is not clear whether lack of cross resistance between cladribine and pentostatin is the exception rather than the rule. We believe that newly diagnosed patients with HCL and leukocytosis, regardless of diagnosis of HCLv, should be tested for VH4-34 positivity either by PCR cloning, by flow cytometry, or by enzyme-linked immunoabsorbent assay using an anti-idiotype antibody such as 9G4.44,46,56  In our study the incidence of VH4-34 positivity was 36% in HCL patients presenting with white blood cells greater than or equal to 5 × 109/L (Figure 3). Patients with VH4-34+ HCL could be considered for alternative approaches, such as therapy with rituximab combined with purine analog, rather than with purine analog alone.71,72 

An Inside Blood analysis of this article appears at the front of this issue.

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 our clinical staff Rita Mincemoyer, Linda Ellison, Elizabeth Maestri, Barbara Debrah, and Sonya Duke for facilitating sample acquisitions and managing results. We recognize Dr Constance Yuan and Marlene Gronberg for flow cytometric analysis and Dr Seth Steinberg for statistical advice. We wish to dedicate this manuscript to the memory of Anna Orthwein, who performed research for this work.

This work was supported by the intramural program, NCI, NIH.

National Institutes of Health

Contributions: E.A. designed and performed research, analyzed data, and wrote the paper; T.S. and M.S.-S. performed research and analyzed data; and R.J.K. designed research, analyzed data, and wrote the paper.

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

Correspondence: Robert J. Kreitman, National Cancer Institute, Laboratory of Molecular Biology, National Institutes of Health, 37/5124b, 37 Convent Dr, MSC 4255 Bethesda, MD; e-mail kreitmar@mail.nih.gov.

1
Cawley
 
JC
Burns
 
GF
Hayhoe
 
FG
A chronic lymphoproliferative disorder with distinctive features: a distinct variant of hairy-cell leukaemia.
Leuk Res
1980
, vol. 
4
 
6
(pg. 
547
-
559
)
2
Matutes
 
E
Wotherspoon
 
A
Catovsky
 
D
The variant form of hairy-cell leukaemia.
Best Pract Res Clin Haematol
2003
, vol. 
16
 
1
(pg. 
41
-
56
)
3
Matutes
 
E
Wotherspoon
 
A
Brito-Babapulle
 
V
Catovsky
 
D
The natural history and clinico-pathological features of the variant form of hairy-cell leukemia.
Leukemia
2001
, vol. 
15
 
1
(pg. 
184
-
186
)
4
Sainati
 
L
Matutes
 
E
Mulligan
 
S
, et al. 
A variant form of hairy-cell leukemia resistant to alpha-interferon: clinical and phenotypic characteristics of 17 patients.
Blood
1990
, vol. 
76
 
1
(pg. 
157
-
162
)
5
Matutes
 
E
Immunophenotyping and differential diagnosis of hairy-cell leukemia.
Hematol Oncol Clin North Am
2006
, vol. 
20
 
5
(pg. 
1051
-
1063
)
6
Matutes
 
E
Oscier
 
D
Montalban
 
C
, et al. 
Splenic marginal zone lymphoma proposals for a revision of diagnostic, staging and therapeutic criteria.
Leukemia
2008
, vol. 
22
 
3
(pg. 
487
-
495
)
7
Cessna
 
MH
Hartung
 
L
Tripp
 
S
Perkins
 
SL
Bahler
 
DW
Hairy-cell leukemia variant—fact or fiction
Am J Clin Pathol
2005
, vol. 
123
 
1
(pg. 
132
-
138
)
8
Grever
 
M
Kopecky
 
K
Foucar
 
MK
, et al. 
Randomized comparison of pentostatin versus interferon alfa-2a in previously untreated patients with hairy-cell leukemia: an intergroup study.
J Clin Oncol
1995
, vol. 
13
 
4
(pg. 
974
-
982
)
9
Saven
 
A
Burian
 
C
Koziol
 
JA
Piro
 
LD
Long-term follow-up of patients with hairy-cell leukemia after cladribine treatment.
Blood
1998
, vol. 
92
 
6
(pg. 
1918
-
1926
)
10
Goodman
 
GR
Burian
 
C
Koziol
 
JA
Saven
 
A
Extended follow-up of patients with hairy-cell leukemia after treatment with cladribine.
J Clin Oncol
2003
, vol. 
21
 
5
(pg. 
891
-
896
)
11
Blasińska-Morawiec
 
M
Robak
 
T
Krykowski
 
E
Hellmann
 
A
Urbanska-Rys
 
H
Hairy-cell leukemia-variant treated with 2-chlorodeoxyadenosine—a report of three cases.
Leuk Lymphoma
1997
, vol. 
25
 
3–4
(pg. 
381
-
385
)
12
Tetreault
 
SA
Robbins
 
BA
Saven
 
A
Treatment of hairy-cell leukemia-variant with cladribine.
Leuk Lymphoma
1999
, vol. 
35
 
3–4
(pg. 
347
-
354
)
13
Quach
 
H
Januszewicz
 
H
Westerman
 
D
Complete remission of hairy-cell leukemia variant (HCL-v) complicated by red cell aplasia post treatment with rituximab.
Haematologica
2005
, vol. 
90
 
Suppl
pg. 
ECR26
 
14
Imamura
 
T
Ohtsuka
 
E
Ogata
 
M
, et al. 
Successful induction of long-term remission using rituximab in a patient with refractory hairy-cell leukemia–Japanese variant.
Int J Hematol
2004
, vol. 
80
 
5
(pg. 
432
-
434
)
15
Goldaniga
 
M
Guffanti
 
A
Gianelli
 
U
Magni
 
M
Lambertenghi Deliliers
 
G
Baldini
 
L
Clinical and molecular complete remission in a case of variant hairy-cell leukemia treated with DHAP followed by high-dose chemotherapy plus rituximab.
Haematologica
2004
, vol. 
89
 
11
pg. 
138e
 
16
Narat
 
S
Gandla
 
J
Dogan
 
A
Mehta
 
A
Successful treatment of hairy-cell leukemia variant with rituximab.
Leuk Lymphoma
2005
, vol. 
46
 
8
(pg. 
1229
-
1232
)
17
Kreitman
 
RJ
Wilson
 
WH
Bergeron
 
K
, et al. 
Efficacy of the anti-CD22 recombinant immunotoxin BL22 in chemotherapy-resistant hairy-cell leukemia.
N Engl J Med
2001
, vol. 
345
 
4
(pg. 
241
-
247
)
18
Arons
 
E
Sunshine
 
J
Suntum
 
T
Kreitman
 
RJ
Somatic hypermutation and VH gene usage in hairy-cell leukemia.
Br J Haematol
2006
, vol. 
133
 
5
(pg. 
504
-
512
)
19
Thorsélius
 
M
Walsh
 
SH
Thunberg
 
U
Hagberg
 
H
Sundstrom
 
C
Rosenquist
 
R
Heterogeneous somatic hypermutation status confounds the cell of origin in hairy-cell leukemia.
Leuk Res
2005
, vol. 
29
 
2
(pg. 
153
-
158
)
20
Vanhentenrijk
 
V
Tierens
 
A
Wlodarska
 
I
Verhoef
 
G
Wolf-Peeters
 
CD
V(H) gene analysis of hairy-cell leukemia reveals a homogeneous mutation status and suggests its marginal zone B-cell origin.
Leukemia
2004
, vol. 
18
 
10
(pg. 
1729
-
1732
)
21
Maloum
 
K
Magnac
 
C
Azgui
 
Z
, et al. 
VH gene expression in hairy-cell leukaemia.
Br J Haematol
1998
, vol. 
101
 
1
(pg. 
171
-
178
)
22
Miranda
 
RN
Cousar
 
JB
Hammer
 
RD
Collins
 
RD
Vnencak-Jones
 
CL
Somatic mutation analysis of IgH variable regions reveals that tumor cells of most parafollicular (monocytoid) B-cell lymphoma, splenic marginal zone B-cell lymphoma, and some hairy-cell leukemia are composed of memory B lymphocytes.
Hum Pathol
1999
, vol. 
30
 
3
(pg. 
306
-
312
)
23
Forconi
 
F
Sahota
 
SS
Raspadori
 
D
Mockridge
 
CI
Lauria
 
F
Stevenson
 
FK
Tumor cells of hairy-cell leukemia express multiple clonally related immunoglobulin isotypes via RNA splicing.
Blood
2001
, vol. 
98
 
4
(pg. 
1174
-
1181
)
24
Martín-Jiménez
 
P
Garcia-Sanz
 
R
Gonzalez
 
D
, et al. 
Molecular characterization of complete and incomplete immunoglobulin heavy chain gene rearrangements in hairy-cell leukemia.
Clin Lymphoma Myeloma
2007
, vol. 
7
 
9
(pg. 
573
-
579
)
25
Forconi
 
F
Sozzi
 
E
Rossi
 
D
, et al. 
Selective influences in the expressed immunoglobulin heavy and light chain gene repertoire in hairy-cell leukemia.
Haematologica
2008
, vol. 
93
 
5
(pg. 
697
-
705
)
26
Swerdlow
 
SH
Campo
 
E
Harris
 
NL
, et al. 
WHO Classification of Tumours of Haematopoietic and Lymphoid Tissues.
2008
, vol. 
Vol 2
 4th ed.
Lyon, France
WHO Press
27
van Dongen
 
JJ
Langerak
 
AW
Bruggemann
 
M
, et al. 
Design and standardization of PCR primers and protocols for detection of clonal immunoglobulin and T-cell receptor gene recombinations in suspect lymphoproliferations: report of the BIOMED-2 Concerted Action BMH4-CT98-3936.
Leukemia
2003
, vol. 
17
 
12
(pg. 
2257
-
2317
)
28
IMGT database
The International Immunogenetics Information System
Accessed September 22, 2009 
29
VBASE2 database
Accessed September 22, 2009 
30
Dunphy
 
CH
Reaction patterns of TRAP and DBA. 44 in hairy-cell leukemia, hairy-cell variant, and nodal and extranodal marginal zone B-cell lymphomas.
Appl Immunohistochem Mol Morphol
2008
, vol. 
16
 
2
(pg. 
135
-
139
)
31
Konoplev
 
S
Medeiros
 
LJ
Bueso-Ramos
 
CE
Jorgensen
 
JL
Lin
 
P
Immunophenotypic profile of lymphoplasmacytic lymphoma/Waldenstrom macroglobulinemia.
Am J Clin Pathol
2005
, vol. 
124
 
3
(pg. 
414
-
420
)
32
Brezinschek
 
HP
Foster
 
SJ
Brezinschek
 
RI
Dorner
 
T
Domiati-Saad
 
R
Lipsky
 
PE
Analysis of the human VH gene repertoire. Differential effects of selection and somatic hypermutation on human peripheral CD5(+)/IgM+ and CD5(−)/IgM+ B cells.
J Clin Invest
1997
, vol. 
99
 
10
(pg. 
2488
-
2501
)
33
Logtenberg
 
T
Schutte
 
ME
Inghirami
 
G
, et al. 
Immunoglobulin VH gene expression in human B cell lines and tumors: biased VH gene expression in chronic lymphocytic leukemia.
Int Immunol
1989
, vol. 
1
 
4
(pg. 
362
-
366
)
34
Stewart
 
AK
Huang
 
C
Stollar
 
BD
Schwartz
 
RS
High-frequency representation of a single VH gene in the expressed human B-cell repertoire.
J Exp Med
1993
, vol. 
177
 
2
(pg. 
409
-
418
)
35
Kraj
 
P
Friedman
 
DF
Stevenson
 
F
Silberstein
 
LE
Evidence for the overexpression of the VH4-34 (VH4.21) Ig gene segment in the normal adult human peripheral blood B-cell repertoire.
J Immunol
1995
, vol. 
154
 
12
(pg. 
6406
-
6420
)
36
Stevenson
 
FK
Smith
 
GJ
North
 
J
Hamblin
 
TJ
Glennie
 
MJ
Identification of normal B-cell counterparts of neoplastic cells which secrete cold agglutinins of anti-I and anti-i specificity.
Br J Haematol
1989
, vol. 
72
 
1
(pg. 
9
-
15
)
37
Hsu
 
FJ
Levy
 
R
Preferential use of the VH4 Ig gene family by diffuse large-cell lymphoma.
Blood
1995
, vol. 
86
 
8
(pg. 
3072
-
3082
)
38
Sato
 
Y
Ichimura
 
K
Tanaka
 
T
, et al. 
Duodenal follicular lymphomas share common characteristics with mucosa-associated lymphoid tissue lymphomas.
J Clin Pathol
2008
, vol. 
61
 
3
(pg. 
377
-
381
)
39
Montesinos-Rongen
 
M
Kuppers
 
R
Schluter
 
D
, et al. 
Primary central nervous system lymphomas are derived from germinal-center B cells and show a preferential usage of the V4-34 gene segment.
Am J Pathol
1999
, vol. 
155
 
6
(pg. 
2077
-
2086
)
40
Deane
 
M
Baker
 
BW
Norton
 
JD
Immunoglobulin VH4 gene usage in B lymphoid leukaemias.
Br J Haematol
1993
, vol. 
84
 
2
(pg. 
242
-
249
)
41
Pritsch
 
O
Magnac
 
C
Dumas
 
G
Egile
 
C
Dighiero
 
G
V gene usage by seven hybrids derived from CD5+ B-cell chronic lymphocytic leukemia and displaying autoantibody activity.
Blood
1993
, vol. 
82
 
10
(pg. 
3103
-
3112
)
42
Maloum
 
K
Magnac
 
C
Pritsch
 
O
Binet
 
JL
Merle-Beral
 
H
Dighiero
 
G
Skewed rearrangement of the VH4-21 gene during pre-B acute lymphoblastic leukemia.
Leuk Lymphoma
1995
, vol. 
17
 
5–6
(pg. 
435
-
441
)
43
Rettig
 
MB
Vescio
 
RA
Cao
 
J
, et al. 
VH gene usage in multiple myeloma: complete absence of the VH4.21 (VH4-34) gene.
Blood
1996
, vol. 
87
 
7
(pg. 
2846
-
2852
)
44
Frøyland
 
M
Thompson
 
KM
Thorpe
 
SJ
Sahota
 
SS
Gedde-Dahl
 
T
Bogen
 
B
A VH4-34+ myeloma protein with weak autoreactivity.
Haematologica
2007
, vol. 
92
 
5
(pg. 
690
-
693
)
45
González
 
D
González
 
M
Balanzategui
 
A
, et al. 
Molecular characteristics and gene segment usage in IGH gene rearrangements in multiple myeloma.
Haematologica
2005
, vol. 
90
 
7
(pg. 
906
-
913
)
46
Bhat
 
NM
Lee
 
LM
van Vollenhoven
 
RF
Teng
 
NN
Bieber
 
MM
VH4-34 encoded antibody in systemic lupus erythematosus: effect of isotype.
J Rheumatol
2002
, vol. 
29
 
10
(pg. 
2114
-
2121
)
47
Pascual
 
V
Victor
 
K
Spellerberg
 
M
Hamblin
 
TJ
Stevenson
 
FK
Capra
 
JD
VH restriction among human cold agglutinins. The VH4-21 gene segment is required to encode anti-I and anti-i specificities.
J Immunol
1992
, vol. 
149
 
7
(pg. 
2337
-
2344
)
48
Riboldi
 
P
Gaidano
 
G
Schettino
 
EW
, et al. 
Two acquired immunodeficiency syndrome-associated Burkitt's lymphomas produce specific anti-i IgM cold agglutinins using somatically mutated VH4-21 segments.
Blood
1994
, vol. 
83
 
10
(pg. 
2952
-
2961
)
49
Silberstein
 
LE
Jefferies
 
LC
Goldman
 
J
, et al. 
Variable region gene analysis of pathologic human autoantibodies to the related i and I red blood cell antigens.
Blood
1991
, vol. 
78
 
9
(pg. 
2372
-
2386
)
50
Bhat
 
NM
Bieber
 
MM
Hsu
 
FJ
, et al. 
Rapid cytotoxicity of human B lymphocytes induced by VH4-34 (VH4.21) gene-encoded monoclonal antibodies, II.
Clin Exp Immunol
1997
, vol. 
108
 
1
(pg. 
151
-
159
)
51
Bhat
 
NM
Bieber
 
MM
Stevenson
 
FK
Teng
 
NN
Rapid cytotoxicity of human B lymphocytes induced by VH4-34 (VH4.21) gene-encoded monoclonal antibodies.
Clin Exp Immunol
1996
, vol. 
105
 
1
(pg. 
183
-
190
)
52
Bhat
 
NM
Bieber
 
MM
Teng
 
NN
Cytotoxicity of murine B lymphocytes induced by human VH4-34 (VH4.21) gene-encoded monoclonal antibodies.
Clin Immunol Immunopathol
1997
, vol. 
84
 
3
(pg. 
283
-
289
)
53
Baranzini
 
SE
Jeong
 
MC
Butunoi
 
C
Murray
 
RS
Bernard
 
CC
Oksenberg
 
JR
B cell repertoire diversity and clonal expansion in multiple sclerosis brain lesions.
J Immunol
1999
, vol. 
163
 
9
(pg. 
5133
-
5144
)
54
Childs
 
RA
Dalchau
 
R
Scudder
 
P
Hounsell
 
EF
Fabre
 
JW
Feizi
 
T
Evidence for the occurrence of O-glycosidically linked oligosaccharides of poly-N-acetyllactosamine type on the human leukocyte common antigen.
Biochem Biophys Res Commun
1983
, vol. 
110
 
2
(pg. 
424
-
431
)
55
Bhat
 
NM
Bieber
 
MM
Chapman
 
CJ
Stevenson
 
FK
Teng
 
NN
Human antilipid A monoclonal antibodies bind to human B cells and the i antigen on cord red blood cells.
J Immunol
1993
, vol. 
151
 
9
(pg. 
5011
-
5021
)
56
van Vollenhoven
 
RF
Bieber
 
MM
Powell
 
MJ
, et al. 
VH4-34 encoded antibodies in systemic lupus erythematosus: a specific diagnostic marker that correlates with clinical disease characteristics.
J Rheumatol
1999
, vol. 
26
 
8
(pg. 
1727
-
1733
)
57
Owens
 
GP
Burgoon
 
MP
Anthony
 
J
Kleinschmidt-DeMasters
 
BK
Gilden
 
DH
The immunoglobulin G heavy chain repertoire in multiple sclerosis plaques is distinct from the heavy chain repertoire in peripheral blood lymphocytes.
Clin Immunol
2001
, vol. 
98
 
2
(pg. 
258
-
263
)
58
Smith-Jensen
 
T
Burgoon
 
MP
Anthony
 
J
Kraus
 
H
Gilden
 
DH
Owens
 
GP
Comparison of immunoglobulin G heavy-chain sequences in MS and SSPE brains reveals an antigen-driven response.
Neurology
2000
, vol. 
54
 
6
(pg. 
1227
-
1232
)
59
Colombo
 
M
Dono
 
M
Gazzola
 
P
, et al. 
Accumulation of clonally related B lymphocytes in the cerebrospinal fluid of multiple sclerosis patients.
J Immunol
2000
, vol. 
164
 
5
(pg. 
2782
-
2789
)
60
Parr
 
TB
Johnson
 
TA
Silberstein
 
LE
Kipps
 
TJ
Anti-B–cell autoantibodies encoded by VH 4-21 genes in human fetal spleen do not require in vivo somatic selection.
Eur J Immunol
1994
, vol. 
24
 
12
(pg. 
2941
-
2949
)
61
Hamblin
 
TJ
Davis
 
Z
Gardiner
 
A
Oscier
 
DG
Stevenson
 
FK
Unmutated Ig V(H) genes are associated with a more aggressive form of chronic lymphocytic leukemia.
Blood
1999
, vol. 
94
 
6
(pg. 
1848
-
1854
)
62
Mercieca
 
J
Matutes
 
E
Moskovic
 
E
, et al. 
Massive abdominal lymphadenopathy in hairy-cell leukaemia: a report of 12 cases.
Br J Haematol
1992
, vol. 
82
 
3
(pg. 
547
-
554
)
63
Mercieca
 
J
Puga
 
M
Matutes
 
E
Moskovic
 
E
Salim
 
S
Catovsky
 
D
Incidence and significance of abdominal lymphadenopathy in hairy-cell leukaemia.
Leuk Lymphoma
1994
, vol. 
14
 
Suppl 1
(pg. 
79
-
83
)
64
Mercieca
 
J
Matutes
 
E
Emmett
 
E
Coles
 
H
Catovsky
 
D
2-Chlorodeoxyadenosine in the treatment of hairy-cell leukaemia: differences in response in patients with and without abdominal lymphadenopathy.
Br J Haematol
1996
, vol. 
93
 
2
(pg. 
409
-
411
)
65
Kreitman
 
RJ
Squires
 
DR
Stetler-Stevenson
 
M
, et al. 
Phase I trial of recombinant immunotoxin RFB4(dsFv)-PE38 (BL22) in patients with B-cell malignancies.
J Clin Oncol
2005
, vol. 
23
 
27
(pg. 
6719
-
6729
)
66
Saven
 
A
Piro
 
LD
Complete remissions in hairy-cell leukemia with 2-chlorodeoxyadenosine after failure with 2′-deoxycoformycin.
Ann Intern Med
1993
, vol. 
119
 
4
(pg. 
278
-
283
)
67
Piro
 
LD
Ellison
 
DJ
Saven
 
A
The Scripps Clinic experience with 2-chlorodeoxyadenosine in the treatment of hairy-cell leukemia.
Leuk Lymphoma
1994
, vol. 
14
 
Suppl 1
(pg. 
121
-
125
)
68
Maloisel
 
F
Benboubker
 
L
Gardembas
 
M
, et al. 
Long-term outcome with pentostatin treatment in hairy-cell leukemia patients. A French retrospective study of 238 patients.
Leukemia
2003
, vol. 
17
 
1
(pg. 
45
-
51
)
69
Chadha
 
P
Rademaker
 
AW
Mendiratta
 
P
, et al. 
Treatment of hairy-cell leukemia with 2-chlorodeoxyadenosine (2-CdA): long-term follow-up of the Northwestern University experience.
Blood
2005
, vol. 
106
 
1
(pg. 
241
-
246
)
70
Else
 
M
Ruchlemer
 
R
Osuji
 
N
, et al. 
Long remissions in hairy-cell leukemia with purine analogs—A report of 219 patients with a median follow-up of 12.5 years.
Cancer
2005
, vol. 
104
 
11
(pg. 
2442
-
2448
)
71
Ravandi
 
F
Jorgensen
 
JL
O'Brien
 
SM
, et al. 
Eradication of minimal residual disease in hairy-cell leukemia.
Blood
2006
, vol. 
107
 
12
(pg. 
4658
-
4662
)
72
Else
 
M
Osuji
 
N
Forconi
 
F
, et al. 
The role of rituximab in combination with pentostatin or cladribine for the treatment of recurrent/refractory hairy-cell leukemia.
Cancer
2007
, vol. 
110
 
10
(pg. 
2240
-
2247
)
Sign in via your Institution