Skip to main content
Cureus logoLink to Cureus
. 2023 Oct 15;15(10):e47085. doi: 10.7759/cureus.47085

A Case of Hairy Cell Leukemia Variant: Literature Analysis With Focus on Unmet Needs

Amitabh Kumar Upadhyay 1,, Manoj Kumar 1, Anil Prasad 2, Shashank Shekhar 3, Reetal Singh 3
Editors: Alexander Muacevic, John R Adler
PMCID: PMC10651060  PMID: 38022018

Abstract

Hairy cell leukemia variant (HCLv) is a sporadic, B-cell non-Hodgkin lymphoma classified under chronic lymphoproliferative disorders. HCLv usually presents with easy fatigue, dragging pain abdomen, anemia, splenomegaly, hepatomegaly, initially leukocytosis followed by leucopenia, hairy cells in the smear and bone marrow, and an increased risk of infections. There is hypercellular bone marrow, and cytopenias are secondary to hypersplenism. It is essential to differentiate HCL from disorders like classic hairy cell leukemia (HCLc), splenic marginal zone lymphoma, and splenic diffuse red pulp lymphoma, as these are biologically different, with divergent approaches and outcomes. HCLv is poorly responsive or primary refractory to standard purine analogs cladribine or pentostatin. It has lower response rates to even cladribine and rituximab combination, a standard of care for classic HCL with very good response rates. Here, we present a case of an elderly male who presented with splenomegaly and leukocytosis, diagnosed as HCLv, and was treated with a cladribine and rituximab-based regime but showed residual cells in bone marrow on flow cytometry at six months post-treatment. There were no residual cells in peripheral blood in flow cytometry. Various aspects of the disease are discussed here with a detailed literature analysis. There is a definite unmet need for research on better treatment options in HCLv to improve its overall outcome.

Keywords: cladribine, massive splenomegaly, lymphoma, hairy cell leukaemia, hairy cell leukemia variant

Introduction

Hairy cell leukemia (HCL) is a sporadic, indolent, B-cell non-Hodgkin lymphoma (NHL) that is classified under chronic lymphoproliferative disorders and comprises classical HCL (HCLc) and HCL-like disorders [1,2]. Hairy cell leukemia variant (HCLv) is a provisional entity in the most recent revision of the WHO 2016 classification [1,2]. It presents with anemia, thrombocytopenia, splenomegaly, hepatomegaly, and leukocytosis initially followed by leucopenia, increased risk of infections, and easy fatigue. It is essential to differentiate HCL from HCL-like disorders as these are biologically different, with divergent treatment approaches and prognoses. Morphologically, these disorders look similar, and immunophenotyping helps to distinguish between these entities. In 2008, the World Health Organization (WHO) reclassified the disorders into separate categories of lymphoproliferative disorders [3]. The four immunophenotyping markers which help in differentiation are CD11C, CD103, CD25, and CD123. Each marker is assigned a score of 1 if expressed and 0 if not expressed. Classical HCL scores 3 to 4, whereas the HCL-like disorders have low immunological scores of 0 to 1 [1-3]. HCLv is a mature lymphoid B-cell disorder characterized by the identification of hairy cells, a specific immunophenotypic profile, and a different clinical course than other disorders of a similar spectrum like HCLc, splenic marginal zone lymphoma (SMZL), and splenic diffuse red pulp lymphoma (SDRPL). HCLv patients do present with splenomegaly, leukocytosis, or infections. The circulating lymphoid cells have a morphology intermediate between prolymphocytes and hairy cells. The HCL immunological score is low (0 or 1), with no expression of CD25 and inconstant or weak CD123 expression [1-3]. 

HCLv is a more aggressive disease than HCLc, with a different clinical course and usually poorly responsive or primary refractory to standard treatment options. The response rate with purine analogs cladribine or pentostatin is greater than 90% for HCLc but in the 40-50% range for HCLv, with most responses being partial [4]. In HCLv, the abnormal lymphoid cells do not usually express CD25, CD200, and CD123. The proto-oncogene B-Raf (BRAFV600E) gene mutation is not present in HCLv but is a hallmark of HCLc [1-4]. Similarly, mitogen-activated protein kinase kinase 1(MAP2K1) gene mutations are present in about 30% of HCLv cases [1-4]. HCLv is treated with more aggressive therapies with purine analogs and rituximab. The clinical course of HCLv is more aggressive, with a median overall survival (OS) of 9 years [1-4]. Here, we present a case of HCLv who presented with splenomegaly and leukocytosis and was treated with a cladribine and rituximab-based regime. 

Case presentation

A 68-year-old male patient with Eastern Cooperative Oncology Group (ECOG) performance status of one presented with complaints of heaviness in the left upper side of the abdomen for one year, which was gradually progressive and worsened in the last four months. He also had B symptoms of fever and significant weight loss of more than 10% in the previous six months. His physical examination revealed massive splenomegaly, palpable 3 cm below the umbilicus. His hemogram showed a white blood cell (WBC) count at 25.8 x109/L, hemoglobin (Hb) level at 10.1 gm/dl, and platelet (PLT) count at 92 x 109/L. Differential counts showed an absolute neutrophil count of 1290 (N), 23994 lymphocytes (L), 516 monocytes (M), no eosinophils (E), and no basophils (B). Peripheral smear showed atypical lymphoid cells, round to oval nuclei with exemplary hairy processes, which are larger than the size of mature lymphocytes (Figure 1A). His bone marrow aspiration showed diluted marrow with suppression of erythroid and myeloid series, proliferation of lymphoid cells, mostly atypical lymphoid cells (68%), and absolute lymphocytosis. Lymphoid series accounted for 82% of all nucleated cells (Figure 1B). Bone marrow biopsy showed bony trabeculae with normocellular marrow, trilineage differentiation with predominantly normoblastic to micronormoblastic erythropoiesis, normal myelopoiesis, and adequate megakaryocytes. Focal marrow areas showed lymphoid cell clusters, intermediate size with coarse chromatin and perinuclear halo. There was no evidence of granulomatous inflammation (Figure 1C). Flow cytometry was done on bone marrow aspirate and showed negative for CD5, CD10, CD25, CD123, and CD200 and positive for CD103 and CD11c, suggesting the variant form of hairy cell leukemia. B-RAF mutation analysis by polymerase chain reaction (PCR) was negative. Contrast-enhanced computed tomography (CECT) of the neck, thorax, and abdomen revealed no significant abnormality or lymphadenopathy in the neck or thorax region. There was gross splenomegaly (21 cm) and hepatomegaly (19 cm). 

Figure 1. (A) Peripheral smear Leishman staining in 100X magnification showing atypical lymphoid cells with fine hairy processes (blue arrow); (B) bone marrow aspiration Leishman 400X magnification showing hairy cells having round to oval nucleus with moderate pale grey cytoplasm (blue arrow); (C) bone marrow biopsy hematoxylin and eosin (H&E) staining in 100X magnification showing cellular marrow, medium-sized lymphoid cells with oval nuclei, open chromatin, absent nucleoli, and a characteristic serrated cytoplasmic border.

Figure 1

This patient received systemic therapy with one cycle of cladribine in a dose of 0.15 mg/Kg on days 1-5, with eight weekly doses of rituximab 375 mg/m2 beginning day one. The hemogram findings during treatment and at six months are shown in Table 1. Follow-up hemogram at six months showed a WBC count of 5.4 x109/L, a Hb level of 12.9 gm/dl, and a platelet count of 105 x 109/L. Differential counts showed 71% neutrophils (N), 17% lymphocytes (L), 9% monocytes (M), 03% eosinophils (E), and 00% basophils (B). Peripheral smear showed normocytic normochromic RBC (Figure 2A). Marrow aspiration findings showed cellular reactive marrow with mild erythroid hyperplasia, lymphoid series accounting for 12%, and no evidence of granulomas/residual malignancy (Figure 2B). Bone marrow biopsy findings showed bony trabeculae with normocellular marrow, trilineage differentiation with predominantly normoblastic to micronormoblastic erythropoiesis, normal myelopoiesis, and adequate megakaryocytes. Focal areas (1%) show lymphoid cell clusters, intermediate size with coarse chromatin, and perinuclear halo without any evidence of granulomatous inflammation (Figure 2C). CECT Abdomen shows hepatomegaly and splenomegaly (12.36cm) with standard shape, density, and enhancement. The splenomegaly had markedly reduced after treatment (Figures 3A, 3B). Flow cytometry revealed 0.5% B lymphoid cells on bone marrow aspirate with positive expression of hairy cell markers (CD103+CD11c+ CD20+ bright) (Figure 4). Flow cytometry identified 21% normal lymphocytes in peripheral blood consisting of 16% T cells, 4% NK cells, and 1% B cells. There was no evidence of any residual cells with hairy cell markers (Figure 5). 

Table 1. Hemogram at the time of chemotherapy and rituximab doses and at six months .

Drugs WBC Hb (g/dL) Platelets N (%) L (%) M (%) E (%) B (%)
Cladribine (Day1 to Day 5), Rituximab Week 1 25.8 x 109/L 10.1 92 x 109/L 5 93 2 0 0
Rituximab - Week 2 8.9 x 109/L 09.5 91 x 109/L 18 78 4 0 0
Rituximab - Week 3 2.7 x 109/L 13.2 71 x 109/L 26 65 8 1 0
Rituximab - Week 4 6.5 x 109/L 13.8 98 x 109/L 62 31 6 1 0
Rituximab - Week 5 4.8 x 109/L 12.1 73 x 109/L 79 11 6 3 1
Rituximab - Week 6 4.7 x 109/L 12.8 78 x 109/L 73 15 7 5 0
Rituximab - Week 7 5.4 x 109/L 12.4 115 x 109/L 72 14 8 6 1
Rituximab - Week 8 4.7 x 109/L 12.3 110 x 109/L 73 17 8 2 0
At six months 5.4 x 109/L 12.9 105 x 109/L 71 17 9 3 0
Normal reference range 4.0-11.0 x 109/L 11.5-16.5 g/dL 150-400 x 109/L 60-70% 30-40% 2-8% 1-6% 0-1%

Figure 2. (A) Peripheral smear Leishman staining in 10X magnification showing normal leukocyte count and distribution, (B) bone marrow aspiration Leishman 40X magnification showing normal hematopoietic cells and lymphocytes, and (C) bone marrow biopsy post-chemotherapy Leishman H&E 10X magnification showing remission of hairy cell leukemia.

Figure 2

Figure 3. (A) Axial plane of CECT abdomen showing the presence of splenomegaly (upward red arrow) and hepatomegaly (downward red arrow) and (B) axial plane of CECT abdomen at six months after treatment showing significant resolution of splenomegaly (upward red arrow) and hepatomegaly (downward red arrow).

Figure 3

CECT: Contrast-enhanced computerized tomography 

Figure 4. Bone marrow flow cytometric findings show 0.5% B lymphoid cells with positive expression of B cell markers (CD103, CD11c, and CD20).

Figure 4

Figure 5. Peripheral blood flow cytometric findings identify 21% lymphocytes (16% of total events are T cells, <1% B cells and 4% NK cells), and hairy cell markers are negative.

Figure 5

NK: Natural killer

The patient became asymptomatic at six months. Due to discrepant results for residual cells in the bone marrow and peripheral blood, he has been counseled for a second course of rituximab for eight doses to have undetected residual cells in the bone marrow. 

Discussion

HCL-v, first described by Cawley et al. [5], occurs at an incidence of 0.03 per 100,000 persons per year, has a male predominance (ratio of 6:1), and constitutes around 0.4% of all chronic lymphoid malignancies [1-5]. HCL-v is a disease of the elderly, with a median age of 71 [1-4]. Initial symptoms are abdominal discomfort or pain, usually secondary to splenomegaly, hepatomegaly, and symptoms derived from cytopenias such as anemia, bleeding, and/or recurrent infections. Usually, diagnosis is delayed for many years due to nonspecific symptoms of weakness, easy fatigue, and dragging pain in the abdomen, which is attributed to advanced age. Sometimes, we find a history of blood transfusions because of anemia. The diagnosis warrants a detailed clinical history, physical examination, and investigations, including peripheral blood smear, bone marrow morphology, immunophenotyping, and molecular diagnostics. Peripheral blood morphology of HCLv typically shows small-to-medium lymphocytes with circumferential cytoplasmic projections. Patients usually present with massive splenomegaly, leukocytosis without monocytopenia, and a hypercellular bone marrow that can be easily aspirated. Patients develop cytopenias with time because of hypersplenism [1-4]. 

SMZL with villous lymphocytes is another indolent B-cell lymphoma that may be mistaken for HCL. However, there are unipolar or bipolar cytoplasmic projections rather than circumferential projections, as seen in HCL [1-4]. SMZL is characterized by abnormal lymphoid cells with round nuclei, condensed chromatin, and basophilic cytoplasm with polar short villi (villous lymphocytes) in the peripheral blood [1-4]. SDRPL is a provisional entity, very close if not identical to HCL-V. SDRPL could be the first step before the occurrence of HCLv and is characterized by the presence of a large proportion of small to medium-sized villous lymphoid cells in peripheral blood [1-4]. The abnormal lymphoid cells have a polar distribution of the villi, and the nucleolus is small or invisible. The characteristics of HCLc, HCLv, SMZL, and SDRPL are further described in Table 2 [1-4].

Table 2. Analysis of disease characteristics of HCLc, HCLv, SMZL, and SDRPL.

TRAP: Tartrate-resistant acid phosphatase, IGHV: immunoglobulin heavy chain, CCND3: cyclin D3, del: deletion; HCLc: classic hairy cell leukemia; HCLv: hairy cell leukemia variant; SMZL: splenic marginal zone lymphoma; SDRPL: splenic diffuse red pulp lymphoma

Data from [1-4]

Table creation: AK Upadhyay

                                   - HCLc HCLv SMZL SDRPL
B symptoms Rare Rare Approx 1/4th Approx 1/3rd
Blood smear Reniform or oval nuclei, circumferential long villi, inconspicuous nucleoli Abundant circumferential villi and prominent nucleoli Polar villi and inconspicuous nucleoli Broad-based polar cytoplasmic extensions
Leukocyte count Low High High High
Median age 59 71 69 66.5
TRAP activity   Positive Negative Weak Negative
Surface Immunoglobulin   IgM IgG IgM Biclonal or monoclonal
Immunophenotype CD11c+, CD25+, CD123+, CD 200+ CD11c+, CD25-, CD123-, CD200-, CD103+ CD11c+, CD25±, CD103- CD11c-, CD25-, CD123-, CD103-, DBA.44+
Annexin A1 Positive Negative Negative Negative
Cytogenetics del 13q, del 7q del 17p del 7q, trisomy 3 del 7q, trisomy 18, del 17p
IGHV mutation Mutated Unmutated Usually mutated Positive
BRAF mutation Mutated Unmutated Not reported Negative
MAP2K1 positivity Less common More common Not reported Positive
CCND3 Wild type Mutated Unreported Mutated
Spleen involvement Red pulp Red pulp White pulp Red pulp
Bone marrow infiltration Inter-sinusoidal Mostly intra-sinusoidal, rarely inter-sinusoidal Paratrabecular and predominantly inter-sinusoidal Interstitial, rarely nodular
Treatment Purine analogs, rituximab Purine analogs and rituximab, Splenectomy Splenectomy, rituximab Splenectomy  
Median overall survival from diagnosis (years)   20 9 10 5-year survival of more than 90%

HCLv is inherently aggressive and does not respond well to single-agent purine analogs. Cladribine given as a single five-day daily, the two-hour intravenous infusion has shown poorer response rates than HCLc [1-4]. The combination therapy of purine analogs with an anti-CD20 monoclonal antibody, rituximab, is the first-line treatment option for HCLv [6]. In the largest phase 2 study by Chihara et al., 20 patients of HCLv received cladribine 0.15 mg/kg on days one to five with eight rituximab doses of 375 mg/m2 at weekly intervals, beginning day one [6]. Patients were eligible for a second rituximab course six months after cladribine if minimal residual disease (MRD) was detected in the blood. Lymphopenia and neutropenia are seen in 65-75% of patients by week two, which gradually improves by week four to six. The complete remission (CR) rate from the cladribine plus rituximab regime was 95%, and the median duration of CR was 70.1 months. The patients with TP53 mutations had a shorter progression-free survival (PFS) (median, 36.4 months vs. unreached; P = .0024) and OS (median, 52.4 months vs. unreached; P = .032). Similarly, subsets with MRD-negative CR at six months were showing a longer PFS (unreached vs. 17.4 months; P < .0001) and OS (unreached vs 38.2 months; P < .0001) [6]. Very limited data is available regarding the median overall survival of patients in CR and patients without CR for other cases or case series since no follow-up treatment details are available. 

Many treatment modalities have been attempted to treat HCLv with gradual evolution. Splenectomy is a potential treatment option shown in a study by Matutes et al. [7]. It showed remission in 13 out of 19 patients lasting one to ten years, with a median of four years. Pentostatin and α-interferon, active in HCLc, have shown poor responses in HCLv [1-4,8]. Allogenic marrow transplantation has been tried in a patient with HCLv, which achieved clinical remission for only 16 months [9]. Splenic irradiation and alemtuzumab have also been attempted in HCLv with limited success [10,11]. The bendamustine plus rituximab combination was attempted in three cases with a good response in first-line treatment in HCLv [12]. Small studies showed the effect of the administration of cladribine and late rituximab after one month of cladribine [13]. It is debatable whether starting rituximab concurrent with cladribine or after one month is preferable. There is a possibility of infections and fever with cladribine infusion, which makes concurrent rituximab administration difficult.

Due to the rarity of this disease and the limited number of studies, no clear second or subsequent lines of therapy are superior to retreatment with cladribine or any other choice. Once the patient relapses, several second-line options are rituximab, a monoclonal antibody against CD52 (alemtuzumab), and combination chemotherapies such as CHOP or CHOP-R [1-4,11]. Newer therapies may include treatment with a recombinant immunotoxin with an anti-CD22 variable domain fused to a truncated Pseudomonas endotoxin, moxetumomab pasudotox, a Bruton’s tyrosine kinase inhibitor, ibrutinib, and mitogen-activated protein kinase (MEK) inhibitors if MAP2K1 mutation is present [1-4]. 

The rearrangements expressing immunoglobulin variable heavy chain gene, VH4-34, commonly used in autoimmune disorders, were found in 40% of patients with HCLv versus 10% with HCLc in a study by Arons et al. [14]. VH4-34+ patients were found to have higher white blood cell counts at diagnosis, a lower response rate and PFS, and shorter overall survival with first-line cladribine [14]. 

The cases of HCLv are scarce, leading to the publication of a limited number of case reports and small series. An extensive search for the concerned cases was done on the internet, and 28 case reports were found from 1996 to 2023, summarized in tabular form (Table 3) [3,4,9-12,15-36]. There are no phase 3 studies to date due to rare occurrences. Limited phase 2 studies and a series of cases are summarized in tabular form (Table 4) [6,8,13,37-42]. 

Table 3. Published case reports of HCLv.

Author Year Country Topic Age/Sex
Sgarabotto et al. [10] 1996 Italy Remission in Hairy Cell Leukemia-Variant Following Splenic Radiotherapy Alone 79/M
Palomera et al. [15] 2002 Spain Cladribine (2-chlorodeoxyadenosine) therapy in hairy cell leukemia variant. A report of three cases NA
Goldaniga et al. [16] 2004 Italy Clinical and molecular complete remission in a case of variant hairy cell leukemia treated with DHAP followed by high-dose chemotherapy plus rituximab NA
Quach et al. [17] 2005 Australia Complete remission of hairy cell leukemia variant (HCL-v) complicated by red cell aplasia post treatment with rituximab NA
Narat et al. [18] 2005 UK Successful treatment of hairy cell leukemia variant with rituximab 53/M
Ya-In et al. [19] 2005 Canada Hairy Cell Leukemia Variant With Features of Intrasinusoidal Bone Marrow Involvement 68/F
Gupta et al. [20] 2005 India Hairy cell leukemia-variant--a case report 66/M
Telek et al. [21] 2007 Hungary  Successful alemtuzumab treatment of a patient with atypical hairy cell leukaemia variant 58/M
Hadzi-Pecova et al. [22] 2008 Republic of Macedonia Rituximab in the treatment of the variant of hairy cell leukaemia: a case report 57/F
Sasaki et al. [11] 2008 Japan Effective Treatment of a Refractory Hairy Cell Leukemia Variant with Splenic Pre-Irradiation and Alemtuzumab 72/M
Busemann et al. [9] 2010 Germany Late extramedullary relapse after allogeneic transplantation in a case of variant hairy cell leukaemia 60/M
Hsieh et al. [23] 2011 Taiwan Hairy cell leukemia and variant in Taiwan: report of a variant case and literature review 67/M
Kanellis et al. [24] 2011 Greece Hairy cell leukemia variant A description of the spleen morphology and immunophenotype of an archetypical case 79/M
Pande et al. [25] 2013 India A Hairy Cell Leukaemia Variant – A Rare Case Report 58/F
Amelia et al. [26] 2015 Romania A case of hairy cell leukemia variant 52/M
Rudolf-Oliveira et al. [27] 2015 Brazil Hairy cell leukemia variant: the importance of differential diagnosis 74/M
Jian et al. [28] 2016 Canada A Unique Hairy Cell Leukemia Variant 65/F
Kapoor et al. [29] 2017 India Hairy Cell Leukemia – Variant (HCL-V): A Separate Entity 65/F
Visentin et al. [12] 2017 Italy Bendamustine plus rituximab is an effective first-line treatment in hairy cell leukemia variant: a report of three cases 77/F, 83/F, 90/M
 McKay et al. [30] 2017 Australia Hairy cell leukaemia variant with periarticular joint infiltration and excellent radiotherapy response 73/M
Bohn et al. [31] 2017 Austria  Ibrutinib for relapsed refractory hairy cell leukemia variant 82/M
Jain et al. [32] 2018 USA Biclonal IGHV-4-34 hairy cell leukemia variant and CLL - successful treatment with ibrutinib and venetoclax 79/M
Andritsos et al. [33] 2018 USA Trametinib for the treatment of IGHV4-34, MAP2K1-mutant variant hairy cell leukemia 52/M
Wiber et al. [34] 2019 France Variant form of hairy cell leukemia 64, 72/M
Visentin et al. [3] 2020 Italy Ibrutinib in relapsed hairy cell leukemia variant: A case report and review of the literature 48/F, 90/F
Otieno et al. [4] 2022 USA Remarkable Response of Hairy Cell Leukemia Variant to Single-Agent Cladribine 68Y/M
Passucci et al. [35] 2022 Italy High Curative Potential of Ibrutinib in Hairy Cell Leukemia Variant Refractory to Conventional Chemotherapy: A Case Report 62/F
Khan et al. [36] 2023 India A Case of Variant Hairy Cell Leukaemia in Durable Complete Remission on Combination Therapy 50Y/F

Table 4. Published larger case series and studies related to HCLv.

Author Year Country Topic Number of cases
Sainati et al. [8] 1990 UK A Variant Form of Hairy Cell Leukemia Resistant to α-Interferon: Clinical and Phenotypic Characteristics of 17 Patients 17
Zinzani et al. [37] 1990 Italy Hairy cell leukemia variant: a morphologic, immunologic and clinical study of 7 cases. 7
Robak et al. [38] 1999 Poland 2-chlorodeoxyadenosine (cladribine) in the treatment of hairy cell leukemia and hairy cell leukemia variant: 7-year experience in Poland 6
Ravandi et al. [13] 2011 USA Phase 2 study of cladribine followed by rituximab in patients with hairy cell leukemia 5
Kreitman et al. [39] 2013 USA Cladribine with Immediate Rituximab for the Treatment of Patients with Variant Hairy Cell Leukemia 10
Angelova et al. [40] 2018 USA Clinicopathologic and molecular features in hairy cell leukemia variant:single institutional experience 23
Chihara et al. [6] 2021 USA Long term follow-up of a phase II study of cladribine with concurrent rituximab with hairy cell leukemia variant 20
Rogers et al. [41] 2021 USA Phase 2 study of ibrutinib in classic and variant hairy cell leukemia 7
Wei et al. [42] 2023 China Clinical and molecular characteristics and prognosis of classical hairy cell leukemia and hairy cell leukemia variant 9

MRD measurements can be done with IHC, multicolor flow cytometry, or PCR methods. Our case showed a discrepancy in residual disease with 0.5% abnormal cells in bone marrow flow cytometry. However, no abnormal population of cells in flow cytometry was done on peripheral blood six months after therapy with cladribine and rituximab. Bone marrow examination is required to document remission post-treatment, and so it was performed, and flow was sent on the bone marrow sample. Flow cytometry or polymerase chain reaction on bone marrow is a better modality to detect MRD or early relapse in leukemias. We also wanted to check the residual disease status on blood as the largest study on HCLv by Chihara et al. had done flow on blood for remission status post-treatment [6]. So, in our case, this discrepancy was picked up where bone marrow showed 0.5% residual cells while peripheral blood showed no residual cells. There is no consensus in the cases where the MRD discrepancy is noted. We have advised a second course of Rituximab to our case as bone marrow showed 0.5% residual disease and is presumed to be an early site of relapse even before the obvious signs of failure in peripheral blood. There are no standard comparisons for various methods of MRD assessment [43]. There is no role of second rituximab in MRD-negative cases as of now, as MRD-negative patients were excluded from further doses of rituximab in the phase II study. Since HCLv differs from HCLc, we cannot use the same guidelines for both diseases. There are many dilemmas for a clinician which are yet to be answered. 

Which one is preferred out of concurrent and late rituximab with cladribine infusion? This also needs further research and consensus on whether to include rituximab maintenance in MRD-negative patients. Since it is an aggressive, poorly responsive disease, rituximab is a well-tolerated drug. Theoretically, there is no harm in adding rituximab maintenance in such cases to compensate for the poor disease biology. The role of adding Ibrutinib over second rituximab in MRD-positive cases at six months after cladribine and rituximab therapy needs further research and consensus. In MRD discrepant cases in bone marrow and blood, like in our case, what is ideal treatment is yet to be answered. There is no consensus on the preferred second-line therapy. To conclude, there are many unmet needs in the treatment part of HCLv, which warrants further research. 

Conclusions

This case report summarizes the related scarce literature in one place and will likely help clinicians in decision-making and literature review for future cases. This case also highlights and replicates the poor response to the standard cladribine plus rituximab regime in such cases. This case reinforces the unmet need for future research in therapeutics for this aggressive disease. The role of MRD positivity, the timing of testing, the method of testing, and treatment implications are not standard at present, and this case reinforces the need for a clearer picture. 

The authors have declared that no competing interests exist.

Human Ethics

Consent was obtained or waived by all participants in this study

References

  • 1.Updates in hairy cell leukemia (HCL) and variant-type HCL (HCL-V): rationale for targeted treatments with a focus on ibrutinib. Paillassa J, Safa F, Troussard X. Ther Adv Hematol. 2022;13 doi: 10.1177/20406207221090886. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Current and emerging therapeutic options for hairy cell leukemia variant. Liu Q, Harris N, Epperla N, Andritsos LA. Onco Targets Ther. 2021;14:1797–1805. doi: 10.2147/OTT.S242247. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Ibrutinib in relapsed hairy cell leukemia variant: a case report and review of the literature. Visentin A, Imbergamo S, Trimarco V, et al. Hematol Oncol. 2020;38:823–826. doi: 10.1002/hon.2810. [DOI] [PubMed] [Google Scholar]
  • 4.Remarkable response of hairy cell leukemia variant to single-agent cladribine. Otieno SB, Ogbeide O. Cureus. 2022;14:0. doi: 10.7759/cureus.24976. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.A chronic lymphoproliferative disorder with distinctive features: a distinct variant of hairy-cell leukaemia. Cawley JC, Burns GF, Hayhoe FG. Leuk Res. 1980:547–559. doi: 10.1016/0145-2126(80)90066-1. [DOI] [PubMed] [Google Scholar]
  • 6.Long term follow-up of a phase II study of cladribine with concurrent rituximab with hairy cell leukemia variant. Chihara D, Arons E, Stetler-Stevenson M, et al. Blood Adv. 2021;5:4807–4816. doi: 10.1182/bloodadvances.2021005039. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.The natural history and clinico-pathological features of the variant form of hairy cell leukemia. Matutes E, Wotherspoon A, Brito-Babapulle V, Catovsky D. Leukemia. 2001;15:184–186. doi: 10.1038/sj.leu.2401999. [DOI] [PubMed] [Google Scholar]
  • 8.A variant form of hairy cell leukemia resistant to alpha-interferon: clinical and phenotypic characteristics of 17 patients. Sainati L, Matutes E, Mulligan S, de Oliveira MP, Rani S, Lampert IA, Catovsky D. Blood. 1990;76:157–162. [PubMed] [Google Scholar]
  • 9.Late extramedullary relapse after allogeneic transplantation in a case of variant hairy cell leukaemia. Busemann C, Schüler F, Krüger W, Kiefer T, Wuppermann M, Androshchuk M, Dölken G. Bone Marrow Transplant. 2010;45:1117–1118. doi: 10.1038/bmt.2009.293. [DOI] [PubMed] [Google Scholar]
  • 10.Remission in hairy cell leukemia-variant following splenic radiotherapy alone. Sgarabotto D, Vianello F, Radossi P, et al. Leuk Lymphoma. 1997;26:395–398. doi: 10.3109/10428199709051790. [DOI] [PubMed] [Google Scholar]
  • 11.Effective treatment of a refractory hairy cell leukemia variant with splenic pre-irradiation and alemtuzumab. Sasaki M, Sugimoto K, Mori T, Karasawa K, Oshimi K. Acta Haematol. 2008;119:48–53. doi: 10.1159/000115785. [DOI] [PubMed] [Google Scholar]
  • 12.Bendamustine plus rituximab is an effective first-line treatment in hairy cell leukemia variant: a report of three cases. Visentin A, Imbergamo S, Frezzato F, et al. Oncotarget. 2017;8:110727–110731. doi: 10.18632/oncotarget.21304. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Phase 2 study of cladribine followed by rituximab in patients with hairy cell leukemia. Ravandi F, O'Brien S, Jorgensen J, et al. Blood. 2011;118:3818–3823. doi: 10.1182/blood-2011-04-351502. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Molecular variant of hairy cell leukemia with poor prognosis. Arons E, Kreitman RJ. Leuk Lymphoma. 2011;52 Suppl 2:99–102. doi: 10.3109/10428194.2011.565841. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Cladribine (2-chlorodeoxyadenosine) therapy in hairy cell leukemia variant. A report of three cases. Palomera L, Domingo JM, Sola C, Azaceta G, Calvo MT, Gutierrez M. https://www.haematologica.org/article/view/2228. Haematologica. 2002;87:107–108. [PubMed] [Google Scholar]
  • 16.Clinical and molecular complete remission in a case of variant hairy cell leukemia treated with DHAP followed by high-dose chemotherapy plus rituximab. Goldaniga M, Guffanti A, Gianelli U, Magni M, Deliliers GL, Baldini L. https://www.haematologica.org/article/view/3291. Haematologica. 2004;89:0. [PubMed] [Google Scholar]
  • 17.Complete remission of hairy cell leukemia variant (HCL-v) complicated by red cell aplasia post treatment with rituximab. Quach H, Januszewicz H, Westerman D. https://pubmed.ncbi.nlm.nih.gov/16266917/ Haematologica. 2005;90 Suppl:0. [PubMed] [Google Scholar]
  • 18.Successful treatment of hairy cell leukemia variant with rituximab. Narat S, Gandla J, Dogan A, Mehta A. Leuk Lymphoma. 2005;46:1229–1232. doi: 10.1080/10428190500083433. [DOI] [PubMed] [Google Scholar]
  • 19.Hairy cell leukemia variant with features of intrasinusoidal bone marrow involvement. Ya-In C, Brandwein J, Pantalony D, Chang H. Arch Pathol Lab Med. 2005;129:395–398. doi: 10.5858/2005-129-395-HCLVWF. [DOI] [PubMed] [Google Scholar]
  • 20.Hairy cell leukemia-variant--a case report. Gupta K, Jasmina A, Malhotra P, et al. https://pubmed.ncbi.nlm.nih.gov/16761764/ Indian J Pathol Microbiol. 2005;48:387–389. [PubMed] [Google Scholar]
  • 21.Successful alemtuzumab treatment of a patient with atypical hairy cell leukaemia variant (Artice in Hungarian) Telek B, Batár P, Udvardy M. Orv Hetil. 2007;148:1805–1807. doi: 10.1556/OH.2007.28169. [DOI] [PubMed] [Google Scholar]
  • 22.Rituximab in the treatment of the variant of hairy cell leukaemia: a case report. Hadzi-Pecova L, Stojanovik A, Petrusevska G, Panovska I. https://pubmed.ncbi.nlm.nih.gov/19259059/ Prilozi. 2008;29:355–360. [PubMed] [Google Scholar]
  • 23.Hairy cell leukemia and variant in Taiwan: report of a variant case and literature review. Hsieh YC, Chang ST, Chuang SS, Lu CL, Tsao CJ, Lin CN, Li CY. https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3037205/ Int J Clin Exp Pathol. 2011;4:183–189. [PMC free article] [PubMed] [Google Scholar]
  • 24.Hairy cell leukemia variant. Kanellis G, Garcia-Alonso L, Camacho FI, et al. J Hematopathol. 2011;4:13–16. [Google Scholar]
  • 25.A hairy cell leukaemia variant - a rare case report. Pande P, Yelikar BR, Kumar U M. J Clin Diagn Res. 2013;7:358–360. doi: 10.7860/JCDR/2013/5169.2768. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.A case of hairy cell leukemia variant. Amelia MG, Camelia D, Mihnea AG. https://rjme.ro/archive/56/2/32/ Rom J Morphol Embryol. 2015;56:553–556. [PubMed] [Google Scholar]
  • 27.Hairy cell leukemia variant: the importance of differential diagnosis. Rudolf-Oliveira RC, Pirolli MM, de Souza FS, Michels J, Santos-Silva MC. Rev Bras Hematol Hemoter. 2015;37:132–135. doi: 10.1016/j.bjhh.2015.01.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.A unique hairy cell leukemia variant. Jian C, Hsia CC. Case Rep Oncol. 2016;9:312–316. doi: 10.1159/000446696. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Hairy cell leukemia - variant (HCL-V): a separate entity. Kapoor AK, Kamal J, Singh A, Singh P. J Med Sci Clin Res. 2017;5 [Google Scholar]
  • 30.Hairy cell leukaemia variant with periarticular joint infiltration and excellent radiotherapy response. McKay MJ, Rady KL, McKay TA. Ann Transl Med. 2017;5:162. doi: 10.21037/atm.2017.03.85. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Ibrutinib for relapsed refractory hairy cell leukemia variant. Bohn JP, Wanner D, Steurer M. Leuk Lymphoma. 2017;58:1224–1226. doi: 10.1080/10428194.2016.1239262. [DOI] [PubMed] [Google Scholar]
  • 32.Biclonal IGHV-4-34 hairy cell leukemia variant and CLL - successful treatment with ibrutinib and venetoclax. Jain P, Kanagal-Shamanna R, Konoplev S, Zuo Z, Estrov Z. Am J Hematol. 2018;93:1568–1569. doi: 10.1002/ajh.25264. [DOI] [PubMed] [Google Scholar]
  • 33.Trametinib for the treatment of IGHV4-34, MAP2K1-mutant variant hairy cell leukemia. Andritsos LA, Grieselhuber NR, Anghelina M, et al. Leuk Lymphoma. 2018;59:1008–1011. doi: 10.1080/10428194.2017.1365853. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Variant form of hairy cell leukemia. Wiber M, Maitre E, Cornet E, Salaün V, Naguib D, Troussard X. Clin Case Rep. 2019;7:1161–1166. doi: 10.1002/ccr3.2176. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.High curative potential of ibrutinib in hairy cell leukemia variant refractory to conventional chemotherapy: a case report. Passucci M, Ligia S, Giudice I, et al. https://meddocsonline.org/hematology-and-oncology-current-research/high-curative-potential-of-ibrutinib-in-hairy-cell-leukemia-variant-refractory-to-conventional-chemotherapy-a-case-report.pdf Hematol Oncol Curr Res. 2022;5:1018. [Google Scholar]
  • 36.A case of variant hairy cell leukaemia in durable complete remission on combination therapy. Khan A, Krishna V, Reddy DN. https://www.scitechnol.com/peer-review/a-case-of-variant-hairy-cell-leukaemia-in-durable-complete-remission-on-combination-therapy-P5Wj.php?article_id=21146 Clin Oncol Case Rep. 2023;6 [Google Scholar]
  • 37.Hairy cell leukemia variant: a morphologic, immunologic and clinical study of 7 cases. Zinzani PL, Lauria F, Buzzi M, et al. https://pubmed.ncbi.nlm.nih.gov/2338288/ Haematologica. 1990;75:54–57. [PubMed] [Google Scholar]
  • 38.2-Chlorodeoxyadenosine (cladribine) in the treatment of hairy cell leukemia and hairy cell leukemia variant: 7-year experience in Poland. Robak T, Błasińska-Morawiec M, Błoński J, et al. Eur J Haematol. 1999;62:49–56. doi: 10.1111/j.1600-0609.1999.tb01114.x. [DOI] [PubMed] [Google Scholar]
  • 39.Cladribine with immediate rituximab for the treatment of patients with variant hairy cell leukemia. Kreitman RJ, Wilson W, Calvo KR, et al. Clin Cancer Res. 2013;19:6873–6881. doi: 10.1158/1078-0432.CCR-13-1752. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Clinicopathologic and molecular features in hairy cell leukemia-variant: single institutional experience. Angelova EA, Medeiros LJ, Wang W, et al. Mod Pathol. 2018;31:1717–1732. doi: 10.1038/s41379-018-0093-8. [DOI] [PubMed] [Google Scholar]
  • 41.Phase 2 study of ibrutinib in classic and variant hairy cell leukemia. Rogers KA, Andritsos LA, Wei L, et al. Blood. 2021;137:3473–3483. doi: 10.1182/blood.2020009688. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Clinical and molecular characteristics and prognosis of classical hairy cell leukemia and hairy cell leukemia variant (Article in Chinese) Wei C, Jin XX, Cai H, Wang X, Zhuang JL, Zhou DB. Zhonghua Nei Ke Za Zhi. 2023;62:416–421. doi: 10.3760/cma.j.cn112138-20220414-00274. [DOI] [PubMed] [Google Scholar]
  • 43.Consensus opinion from an international group of experts on measurable residual disease in hairy cell leukemia. Ravandi F, Kreitman RJ, Tiacci E, et al. Blood Cancer J. 2022;12:165. doi: 10.1038/s41408-022-00760-z. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Cureus are provided here courtesy of Cureus Inc.

RESOURCES