You are on page 1of 7

Med Oncol (2013) 30:740

DOI 10.1007/s12032-013-0740-3

REVIEW ARTICLE

Hemophagocytic lymphohistiocytosis (HLH): a review


of literature
Rohtesh S. Mehta • Roy E. Smith

Received: 28 August 2013 / Accepted: 25 September 2013 / Published online: 9 October 2013
Ó Springer Science+Business Media New York 2013

Abstract Hemophagocytic lymphohistiocytosis (HLH) is Background


a rare disease in children and an exceptionally rare
occurrence in adults. It is categorized broadly into primary Hemophagocytic lymphohistiocytosis (HLH) is an inap-
(familial) or the secondary types; the latter being associated propriate immune activation syndrome manifesting as
most commonly with an underlying malignancy. HLH hematological disorder encompassed by an array of patho-
carries a high rate of mortality, and the treatment itself is logical findings and various physical and laboratory abnor-
associated with significant morbidity and risk of mortality. malities. It is broadly classified as either primary or
A high degree of suspicion for the diagnosis, early treat- secondary form. Primary HLH is autosomal recessive/
ment and aggressive supportive care is critical for man- familial (F-HLH), which is usually diagnosed within the first
agement. We present a comprehensive review of literature 2 years of life [1], although adult onset primary HLH has also
describing the clinical features, diagnosis, management and been reported rarely. On paper described two siblings in their
outcome of HLH. 20 s diagnosed with HLH with perforin (PRF1) mutations
[2], while another case reported HLH with PRF1 mutation in
Keywords Hemophagocytic lymphohistiocytosis a 62-year-old Japanese male [3]. Secondary HLH may
(HLH)  Fever of unknown origin (FUO)  Cytopenia develop anytime during life as a consequence of a ‘‘strong
 Hyperferritinemia  Soluble CD25  activation of the immune system,’’ which may occur with
Hypofibrinogenemia various malignancies (malignancy-associated hemophago-
cytic syndrome, MAHS), infections (infection-associated
Abbreviations hemophagocytic syndrome, IAHS)—most commonly with
HLH Hemophagocytic lymphohistiocytosis viruses such as Epstein-Barr virus (EBV), respiratory syn-
F-HLH Familial hemophagocytic lymphohistiocytosis cytial virus (RSV), rotavirus, adenovirus (VAHS), ‘‘fol-
HSCT Hematopoietic stem cell transplant lowing prolonged intravenous nutrition including
MAS Macrophage activation syndrome administration of soluble lipids (fat overload syndrome)’’ or
autoimmune disorders such as systemic lupus erythematosus
(SLE) [4], adult onset Still’s disease, mixed connective tis-
sue diseases and other rheumatic disease [1, 5, 6]. In the
setting of rheumatological diseases, this disorder is often
referred to as macrophage activation syndrome (MAS). HLH
has also been reported after kidney [7], liver [8] and hema-
topoietic stem cell transplants [9].
With the identification of several molecular defects
R. S. Mehta (&)  R. E. Smith associated with the primary HLH, this category is now
Division of Hematology-Oncology, University of Pittsburgh
subdivided into F-HLH types 1–5, associated respectively
Medical Center, UPMC Cancer Pavilion, Room 463,
5150 Centre Avenue, Pittsburgh, PA 15232, USA with (a) an unidentified defect on chromosome 9q21.3-22,
e-mail: mehtars@upmc.edu (b) perforin gene (PRF1) mutation on 10q22, found in

123
Page 2 of 7 Med Oncol (2013) 30:740

6–58 % of patients with F-HLH2, (c) UNC13D gene muta- an underlying malignancy (MAHS), 28 % were due to an
tion on 17q25.1, which encodes for the Munc13-4 protein, infection and the rest were idiopathic. Of all the MAHS
found in 8–13 % of patients with F-HLH3, (d) syntaxin 11 cases, 60 % were associated with non-Hodgkin’s lym-
(STX11) gene mutation on 6q24, found in about 20 % of phoma—primarily T-cell type, followed by other malig-
F-HLH4 cases and (e) syntaxin-binding protein-2 (STXBP2) nancies such as acute leukemias, MDS, Langerhans cell
gene mutation, which encodes for syntaxin-binding protein- histiocytosis and histiocytic sarcoma [24]. Similarly, in
2 or Munc18-2 protein [10]. adults, 55 % of the cases of secondary HLH are associated
Certain rare hereditary disorders are closely related to with an underlying lymphoma (LAHS). Most of them are
HLH. These include X-linked lymphoproliferative (XLP) due to an aggressive NK/T-cell leukemia (38 %), periph-
syndromes types 1 (XLP1) and 2 (XLP2), Griscelli syndrome eral T-cell lymphoma (28 %), diffuse large B-cell lym-
type 2, Chédiak–Higashi syndrome and Hermansky-Pudlak phoma (18 %), extranodal NK/T-cell lymphoma (10 %)
syndrome type 2 caused by mutations in SH2D1A, XIAP and anaplastic large cell lymphoma (7 %) [25]. Other
[11], RAB27A [12], LYST [13] and AP3B1 [14], respec- malignancies associated with HLH include ALL, AML,
tively. XLP is associated with an unusual susceptibility to rhabdomyosarcoma, neuroblastoma, Hodgkin disease and
Epstein-Barr virus (EBV) infection. HLH can occur in both Langerhans cell histiocytosis [26].
XLP-1 (55 %) and XLP-2 (76 %), but recurrent spleno-
megaly, cytopenias and fever are more common in XLP-2
(87 %) than XLP-1 (7 %) [15]. Chédiak–Higashi syndrome Pathophysiology
is an autosomal recessive disorder that presents in children
with hair and skin hypopigmentation, recurrent pyogenic The basic pathophysiological mechanism underlying HLH
infections, mild coagulation defects and varying degree of is an inappropriate activation of T-lymphocytes and mac-
neurological problems [16]. It may mimic HLH in its rophages resulting in phagocytosis of other blood cells,
accelerated phase [17]. Griscelli syndrome is also an auto- excessive cytokine production (interferon-c, TNF-a, IL-2,
somal recessive disorder that presents in children with partial IL-6, IL-8, IL-10, IL-12 and IL-4), resulting in fever and
albinism, neutropenia, thrombocytopenia, neurological other clinical manifestations [27–30].
involvement, immunodeficiency and may also mimic HLH
in its accelerated phase [18, 19]. Hermansky-Pudlak syn-
drome type II primarily presents as a platelet defect-related Clinical features
bleeding disorder, neutropenia, recurrent respiratory ill-
nesses and oculocutaneous albinism [20], but may progress In cases of primary HLH, fever (91 %), hepatomegaly
to fulminant HLH [21]. (90–95 %) and splenomegaly (84 %) are common occur-
rences, while neurological symptoms (47 %), rash (43 %)
and lymphadenopathy (42 %) are seen less frequently.
Epidemiology Common laboratory abnormalities include thrombocyto-
penia (97 %), anemia (88 %), neutropenia (69 %) [31],
HLH is a rare disease. The incidence of the familial type is hypertriglyceridemia (68 %) [32], hyperferritin-
estimated to be 1.2 per 1,000,000 children per year [22]. A emia [ 10,000 lg/L (60 %) [31], hypofibrinogenemia,
retrospective review of 2306 bone marrow biopsies con- transaminitis, hyperbilirubinemia and hypoalbuminemia
ducted between 1987 and 1990 found 23 cases of sec- [22]. Neurological symptoms (33 %) [31] with or without
ondary HLH. Of these, 17 (73 %) had lymphoma- CSF abnormalities such as spinal fluid mononuclear pleo-
associated hemophagocytic syndrome (LAHS). The cytosis and/or elevated spinal fluid protein may also be
remaining cases were associated with acute myelomono- present. Conversely, CSF abnormalities may be present in
cytic leukemia (AMML), myelodysplastic syndrome— the absence of neurological symptoms. Among children
refractory anemia with excess blasts (MDS-RAEB), dia- enrolled in the HLH-94 trial who had CSF analysis done at
betes mellitus and SLE, and a few were of unknown eti- the time of diagnosis, 52 % (101/193) had CSF abnor-
ology [23]. Of all the LAHS cases, HLH was the presenting mality and 37 % had neurological symptoms (72/193).
feature of lymphoma in approximately 40 % (9/23), while Seizure was the most common symptom (11 %) [33], fol-
30 % (7/23) had a preexisting diagnosis of lymphoma, lowed by a diverse variety of neurological symptoms
which was diagnosed within the previous 6 months to ranging from irritability to coma [34, 35].
6 years. Further, there was only a single case of HLH In secondary HLH, fever, anemia and hypertriglyceri-
diagnosed with PTCL-cd type found on splenectomy. demia are seen almost universally, while neutropenia
Another retrospective study of 52 children with sec- (55–100 %), thrombocytopenia (70–80 %), hepatomegaly
ondary HLH found that almost half of the cases were due to (60–90 %), splenomegaly (13–90 %), hyperferritinemia

123
Med Oncol (2013) 30:740 Page 3 of 7

Table 1 Diagnostic criteria for HLH consolidation, pleural effusions, pulmonary edema with
The diagnosis HLH can be established if either I or II is fulfilled
airspace disease, pneumothorax or mediastinal lymphade-
(I) A molecular diagnosis consistent with HLH
nopathy. Abdominal imaging (ultrasound or CT) may
reveal hepatosplenomegaly, lymphadenopathy or ascites.
(II) Diagnostic criteria for HLH fulfilled (five out of the eight
criteria below) Brain CT or MRI in patients with neurological symptoms
(A) Initial diagnostic criteria (to be evaluated in all patients with may show nonspecific white-matter signal abnormalities,
HLH) cerebral volume loss, hydrocephalus, enhancing hemi-
1. Fever spheric lesions, edema, parenchymal hemorrhage, hemor-
2. Splenomegaly rhagic infarct, sinus thrombosis or parenchymal
3. Cytopenias (affectingC 2 of 3 lineages in the peripheral calcifications [40].
blood):
Hemoglobin \90 g/L (in infants \4 weeks:
hemoglobin \100 g/L) Diagnosis (Table 1) [1, 6]
Platelets \100 9 109/L
Neutrophils \1.0 9 109/L In the absence of family history or above-mentioned
4. Hypertriglyceridemia and/or hypofibrinogenemia: molecular defects consistent with F-HLH, five out of the
Fasting triglycerides C3.0 mmol/L (i.e., C265 mg/dL) eight criteria must be met to make a diagnosis of HLH.
Fibrinogen B1.5 g/L These include (a) fever, (b) splenomegaly, (c) cytopenias
5. Hemophagocytosis in bone marrow or spleen or lymph affecting at least two of three lineages, (d) hypertriglyceri-
nodes demia and/or hypofibrinogenemia, (e) hemophagocytosis,
No evidence of malignancy (f) low/absent NK-cell activity, (g) hyperferritinemia and
(B) New diagnostic criteria (h) high-soluble interleukin-2-receptor (soluble CD25)
6. Low or absent NK-cell activity (according to local levels. Once the diagnosis of F-HLH is established, genetic
laboratory reference)
counseling to all siblings should be offered [41].
7. Ferritin C500 lg/L
8. Soluble CD25 (i.e., soluble IL-2 receptor) C2,400 U/mL
Comments:
Treatment
(1) If hemophagocytic activity is not proven at the time of
presentation, further search for hemophagocytic activity is
encouraged. If the bone marrow specimen is not conclusive, In general, the treatment backbone for the primary HLH is
material may be obtained from other organs. Serial marrow constituted by chemotherapy combined with immunother-
aspirates overtime may also be helpful apy. The most common chemotherapy class of drugs used
(2) The following findings may provide strong supportive includes vinca alkaloids—primarily etoposide (VP-16) [42,
evidence for the diagnosis: (a) spinal fluid pleocytosis
43] and rarely teniposide [44]—and the immunotherapy
(mononuclear cells) and/or elevated spinal fluid protein,
(b) histological picture in the liver resembling chronic persistent agents include cyclosporin A, antithymocyte globulin
hepatitis (biopsy) (ATG) and steroids [45]. This framework of chemo-
(3) Other abnormal clinical and laboratory findings consistent immunotherapy was laid by a landmark trial—HLH-94 [1],
with the diagnosis are as follows: cerebromeningeal symptoms, which was later modified to the HLH-2004 [6] regimen. It
lymph node enlargement, jaundice, edema, skin rash. Hepatic should, however, be noted that both these trials included
enzyme abnormalities, hypoproteinemia, hyponatremia, VLDL
:, HDL ; only children; HLH-94 enrolled patients aged 15 or
younger; and the HLH-2004 enrolled patients aged 18 or
Reproduced with written permission from Henter et al. [1, 6]
younger.
Both these trials included two treatment phases—(a) an
(55–100 %), hypofibrinogenemia (14–100 %), hypertri- induction phase and (b) continuation phase—which was
glyceridemia (43–79 %) and elevated LDH (50–100 %) continued until a suitable donor was available for HSCT,
occur variably depending upon the underlying etiology [4, which is the only curative treatment for F-HLH. All
36–39]. patients received induction therapy for the first 8 weeks,
starting with dexamethasone 10 mg/m2 daily, tapered over
8 weeks (5 mg/m2 for 2 weeks, 2.5 mg/m2 for 2 weeks,
Radiographic abnormalities 1.25 mg/m2 for 1 week, and 1 week of tapering) and eto-
poside 150 mg/m2 twice weekly for 2 weeks and then
Over two-third of patients may have nonspecific abnor- weekly.
malities detected on a chest X-ray or a CT scan. These The use of continuation therapy beyond 8 weeks was
include atelectasis (25 %), interstitial opacities (28 %), limited to those with F-HLH, genetically proven HLH,

123
Page 4 of 7 Med Oncol (2013) 30:740

persistent secondary HLH or those who relapse after the sibling donor were offered hematopoietic stem cell trans-
discontinuation of induction therapy. Continuation therapy plantation (HSCT). Response was evaluated after every
included dexamethasone 10 mg/m2 for 3 days at two two cycles. Complete response (CR) was defined as
weekly intervals alternating every week with VP-16 ‘‘unequivocal resolution of clinical signs and symptoms, as
150 mg/m2 at two weekly intervals. In HLH-94, daily oral well as normalization of the laboratory findings, particu-
cyclosporine (target trough levels of 200 lg/L) was also larly the serum level of ferritin.’’ Partial response (PR) was
used in the continuation phase. In HLH-2004, on the other defined as ‘‘a persistent fever and other symptoms of HLH
hand, cyclosporine was used upfront and continued in both or an abnormally high level of serum ferritin in the absence
the phases to intensify the induction regimen. Intrathecal of definitive symptoms.’’ The rest of the cases were clas-
chemotherapy is used in selected patients. In one study, sified as non-responders. After two cycles, CR and PR were
posterior reversible encephalopathy syndrome (PRES) attained in 41 and 18 %, respectively. The overall response
developed in 5 out of 17 patients during the course of rate was 70 % in LAHS, 60 % in EBV–HLH and 40 % in
treatment, which was presumed to be related to cyclo- idiopathic cases. The median follow-up was 100 weeks,
sporine and other potential contributors such as the ele- median overall survival (OS) was 18 weeks (95 % CI
vated blood pressure, liver dysfunction and inflammation. 6–30 weeks) and the 2-year OS rate was roughly 44 %.
Of note, cyclosporine levels in all of them were greater Salvage treatment with DHAP (dexamethasone, highdose
than 200 ng/mL (range 204–532) [46]. cytarabine and cisplatin) was used in 2/17; one patient
received ESHAP (etoposide, methylprednisolone, highdose
cytarabine and cisplatin), and two patients underwent
Intrathecal therapy allogeneic SCT. All of the patients who received salvage
chemotherapy or HSCT died.
HLH-2004 recommends CSF analysis at every 4 weeks in
all children or CSF analysis along with brain MRI at the Anti-CD52 monoclonal antibody
time of new onset of CNS symptoms or re-activation of
systemic or CNS disease. CSF analysis should include at One retrospective review [47] explored the outcome with
least proteins and cell count. Cytological analysis should be alemtuzumab as a salvage therapy in patients refractory to
conducted using cytocentrifugation method (‘‘Cytospin’’) if prior treatment, including a variety of agents such as dexa-
pleocytosis is noted. methasone, etoposide, cyclosporine, intrathecal hydrocorti-
Both the HLH protocols used intrathecal treatment sone, intrathecal methotrexate, methylprednisolone and
weekly during the induction phase from weeks 3–6 in rituximab. Although nobody achieved a CR, 14/22 (63 %)
patients who had an evidence of CNS disease progression did achieve a PR, defined as ‘‘at least a 25 % improvement in
after 2 weeks of systemic treatment, or in those with two or more quantifiable symptoms or laboratory markers of
worsening or unimproved CSF pleocytosis. The rationale HLH 2 weeks following alemtuzumab.’’ Long-term outcome
of not using upfront intrathecal chemotherapy in everybody is unknown.
who had CNS involvement was that the CNS symptoms One case report described the use of alemtuzumab as a
improved with systemic therapy alone in most of the cases. bridge to allogeneic-matched unrelated donor HSCT,
HLH-94 protocol used intrathecal methotrexate alone, which resulted in a complete remission with a normal bone
while HLH-2004 used intrathecal prednisone in addition. marrow biopsy at 1 year with full donor chimerism [48].

Immunotherapy
Other treatment options
In a case series, six children with F-HLH [45], with an age
Chemotherapy regimens range from 3 weeks to 4.5 years, were treated with intra-
venous methylprednisolone starting at 2–5 mg/kg/day,
In a Korean study [39], seventeen HLH adults were treated along with rabbit ATG (10 mg/kg/day) for 5 days, after
with standard CHOP regimen, which included cyclophos- which steroids were tapered and patients were started on
phamide 750 mg/m2, doxorubicin 50 mg/m2, vincristine continuous infusion of cyclosporine A (CSA) (3–5 mg/kg/
1.4 mg/m2 (maximum dose 2 mg) intravenously on day 1 day) with a target trough level between 150 and 200 ng/
and prednisone 40 mg/m2 orally for 5 days, repeated mL, followed by oral CSA (8–10 mg/kg/day). Intrathecal
3–4 weeks for a total of 6–8 cycles. Seventy percent of the methotrexate and corticosteroids were also administered.
patients were younger than 60 years of age and approxi- One patient had received chemotherapy with etoposide,
mately 40 % had an underlying lymphoma. At any point highdose intravenous methotrexate and cyclophosphamide
during the treatment, patients with available HLA-matched about 3 weeks prior to receiving ATG and steroids. All

123
Med Oncol (2013) 30:740 Page 5 of 7

other children received ATG and steroids as the initial suggest that the overall survival may be better in EBV–
therapy. Two children underwent T-cell-depleted HSCT, HLH compared to the F-HLH case (median 10-year OS
conditioned with VP-16, busulfan and cyclophosphamide. 85.7 ± 9.4 vs. 65 ± 7.9 %). However, caution is war-
One of them died at around day 100, while the other one ranted while interpreting these results as the patient pop-
was alive 2 years after the diagnosis with full donor ulations were not directly comparable. Few isolated reports
engraftment. Three out of other four children who did not have suggested the potential role of HSCT in LAHS, based
undergo transplant were well and alive, on maintenance on the underlying type of lymphoma [39, 58].
treatment with CSA (with or without prednisone) at the
follow-up period ranging from 3 to 22 months. One died
within 30 days from sepsis. Survival
Another retrospective review [49] from the same insti-
tute presented the outcome of the same regimen in 38 Untreated HLH is universally fatal with a median survival
patients with F-HLH. Seventy-three percent (28/38) of about 2 months [22]. With treatment and HSCT, the
received ATG as the first line treatment—82 % (23/28) of 5-year survival probability ranges from 45 to 75 % based
those attained CR and 18 % (5/28) achieved PR. The on the study. For instance, in the HLH-94 trial, with a
remaining ten patients received ATG as the second line, median follow-up of 6.2 years, the cumulative 5-year sur-
resulting in CR in 50 % (5/10) and PR in 40 % (4/10). vival was 54 % (95 % CI ± 6 %); 66 ± 14 % after HSCT
Overall, the toxicity profile was ‘‘acceptable.’’ and 0 % without HSCT [31]. There was no survival dif-
ference between matched related (74 ± 16 %) and mat-
Other immune modulators ched unrelated donor transplantations (76 ± 12 %).
Patients with neurological symptoms and CSF abnormality
Some of the immune modulating drugs such as anti-TNF-a at the time of diagnosis had poorer outcome (40 ± 14 %).
(Infliximab, 10 mg/kg once per week) and IL-1 receptor
antagonist anakinra [50] have also been used to treat HLH/
MAS with ‘‘marked clinical and laboratory improvements.’’ Supportive care

As most patients have fever, broad spectrum antimicrobials


Hematopoietic stem cell transplant (HSCT) should be used presumptively until the culture results are
available. Prophylactic use of cotrimoxazole three times a
HSCT is the only curative treatment for F-HLH. However, week, oral antimycotic therapy during initial dexametha-
even after the transplant, mortality remains high, which is sone phase and IVIG (0.5 g/kg) every 4 weeks are also
primarily treatment related (TRM).This is reflected by a recommended [1, 6]. Gastrointestinal prophylaxis with a
majority (50–100 %) of deaths occurring within the first proton pump inhibitor or an H-2 blocker should be used
100 days of HSCT [51]. Some of the most common with steroids. Granulocyte colony-stimulating factor (G-
treatment-related complications include acute infections CSF) and blood transfusions should be used as needed.
(bacterial and fungal), infections caused by CMV re-acti- However, the use of GM-CSF (granulocyte–macrophage
vation, veno-occlusive disease, graft-versus-host disease colony-stimulating factor) is associated with worse out-
and hemorrhage [51]. In addition to these, progression of come manifested by worsening of thrombocytopenia,
HLH is one of the causes of deaths occurring beyond splenomegaly and even death [59]. Early involvement of
100 days of HSCT, which accounts for as high as 50 % of multidisciplinary care teams including a nutritionist, social
the late deaths in some series [51] to as little as 0–10 % in worker, psychologist or a palliative care specialist may be
others [52–54]. Reduced intensity conditioning (RIC) reg- helpful in providing psychological and symptomatic sup-
imens are proposed to be associated with lower TRM as port to the patient as well as family.
compared to the myeloablative conditioning regimens
(MAC), perhaps at the expense of mixed donor chimerism
[55]. On the other hand, a few non-comparative studies Conclusion
using MAC and RIC in selected patients with EBV–HLH
did not show any statistically significant difference in HLH is a rare disease in children and an extremely rare
overall survival [53]. occurrence in adults, which is universally fatal without
The role of HSCT in secondary HLH is not clearly treatment. A majority (50-75 %) of the cases of secondary
defined, as most of the trials involved patients primarily HLH may have an underlying malignancy—most com-
with F-HLH. A few studies that included EBV-associated monly lymphoma. For familial HLH, steroids and etopo-
cases either alone [56] or along with F-HLH cases [53, 57] side-based regimens constitute the induction phase of

123
Page 6 of 7 Med Oncol (2013) 30:740

treatment, and HSCT is the only curative treatment, which 11. Coffey AJ, Brooksbank RA, Brandau O, Oohashi T, Howell GR,
itself is associated with high morbidity and mortality. Bye JM, et al. Host response to EBV infection in X-linked
lymphoproliferative disease results from mutations in an SH2-
Reduced intensity conditioning regimens may be better domain encoding gene. Nat Genet. 1998;20:129–35.
than the traditional myeloablative regimens in terms of 12. Menasche G, Pastural E, Feldmann J, Certain S, Ersoy F, Dupuis
toxicity as well as overall survival, although comparative S, et al. Mutations in RAB27A cause Griscelli syndrome asso-
trials are lacking. Some cases of secondary HLH may ciated with haemophagocytic syndrome. Nat Genet. 2000;25:
173–6.
respond to the treatment for the underlying disease, but 13. Barrat FJ, Auloge L, Pastural E, Lagelouse RD, Vilmer E, Cant
there is a paucity of data, suggesting that the risk of AJ, et al. Genetic and physical mapping of the Chediak-Higashi
recurrence is poorly defined. Most patients with secondary syndrome on chromosome 1q42-43. Am J Hum Genet. 1996;59:
HLH require treatment with chemo-immunotherapy (same 625–32.
14. Dell’Angelica EC, Shotelersuk V, Aguilar RC, Gahl WA, Bo-
as for F-HLH). Some authors suggest using chemo- nifacino JS. Altered trafficking of lysosomal proteins in Her-
immunotherapy as an initial treatment for LAHS followed mansky-Pudlak syndrome due to mutations in the beta 3A subunit
by the specific treatment for the lymphoma, with a goal of of the AP-3 adaptor. Mol Cell. 1999;3:11–21.
pacifying the heightened inflammatory response first [60]. 15. Pachlopnik Schmid J, Canioni D, Moshous D, Touzot F, Mah-
laoui N, Hauck F, et al. Clinical similarities and differences of
The role of HSCT in secondary forms is less clearly patients with X-linked lymphoproliferative syndrome type 1
defined. A high degree of clinical suspicion for diagnosis is (XLP-1/SAP deficiency) versus type 2 (XLP-2/XIAP deficiency).
required; immediate referral to a tertiary care center and Blood. 2011;117:1522–9.
early treatment are of paramount importance. 16. Kaplan J, De Domenico I, Ward DM. Chediak-Higashi syn-
drome. Curr Opin Hematol. 2008;15:22–9.
17. Rubin CM, Burke BA, McKenna RW, McClain KL, White JG,
Conflict of interest None. Nesbit ME Jr, et al. The accelerated phase of Chediak-Higashi
syndrome. An expression of the virus-associated hemophagocytic
syndrome? Cancer. 1985;56:524–30.
18. Griscelli C, Durandy A, Guy-Grand D, Daguillard F, Herzog C,
References Prunieras M. A syndrome associating partial albinism and
immunodeficiency. Am J Med. 1978;65:691–702.
1. Henter JI, Arico M, Egeler RM, Elinder G, Favara BE, Filipovich 19. Mancini AJ, Chan LS, Paller AS. Partial albinism with immu-
AH, et al. HLH-94: a treatment protocol for hemophagocytic nodeficiency: Griscelli syndrome: report of a case and review of
lymphohistiocytosis. HLH study Group of the Histiocyte Society. the literature. J Am Acad Dermatol. 1998;38:295–300.
Med Pediatr Oncol. 1997;28:342–7. 20. Wei ML. Hermansky-Pudlak syndrome: a disease of protein
2. Clementi R, Emmi L, Maccario R, Liotta F, Moretta L, Danesino trafficking and organelle function. Pigment Cell Res. 2006;19:
C, et al. Adult onset and atypical presentation of hemophagocytic 19–42.
lymphohistiocytosis in siblings carrying PRF1 mutations. Blood. 21. Enders A, Zieger B, Schwarz K, Yoshimi A, Speckmann C,
2002;100:2266–7. Knoepfle EM, et al. Lethal hemophagocytic lymphohistiocytosis
3. Nagafuji K, Nonami A, Kumano T, Kikushige Y, Yoshimoto G, in Hermansky-Pudlak syndrome type II. Blood. 2006;108:81–7.
Takenaka K, et al. Perforin gene mutations in adult-onset he- 22. Henter JI, Elinder G, Soder O, Ost A. Incidence in Sweden and
mophagocytic lymphohistiocytosis. Haematologica. 2007;92: clinical features of familial hemophagocytic lymphohistiocytosis.
978–81. Acta Paediatr Scand. 1991;80:428–35.
4. Fukaya S, Yasuda S, Hashimoto T, Oku K, Kataoka H, Horita T, 23. Chang CS, Wang CH, Su IJ, Chen YC, Shen MC. Hematophagic
et al. Clinical features of haemophagocytic syndrome in patients histiocytosis: a clinicopathologic analysis of 23 cases with special
with systemic autoimmune diseases: analysis of 30 cases. reference to the association with peripheral T-cell lymphoma.
Rheumatology (Oxford). 2008;47:1686–91. J Formos Med Assoc. 1994;93:421–8.
5. Kuzmanova SI. The macrophage activation syndrome: a new 24. Veerakul G, Sanpakit K, Tanphaichitr VS, Mahasandana C, Jir-
entity, a potentially fatal complication of rheumatic disorders. arattanasopa N. Secondary hemophagocytic lymphohistiocytosis
Folia Med (Plovdiv). 2005;47:21–5. in children: an analysis of etiology and outcome. J Med Assoc
6. Henter JI, Horne A, Arico M, Egeler RM, Filipovich AH, Thai. 2002;85(Suppl 2):S530–41.
Imashuku S, et al. HLH-2004: diagnostic and therapeutic 25. Han AR, Lee HR, Park BB, Hwang IG, Park S, Lee SC, et al.
guidelines for hemophagocytic lymphohistiocytosis. Pediatr Lymphoma-associated hemophagocytic syndrome: clinical fea-
Blood Cancer. 2007;48:124–31. tures and treatment outcome. Ann Hematol. 2007;86:493–8.
7. Karras A, Thervet E, Legendre C. Hemophagocytic syndrome in 26. Celkan T, Berrak S, Kazanci E, Ozyurek E, Unal S, Ucar C, et al.
renal transplant recipients: report of 17 cases and review of lit- Malignancy-associated hemophagocytic lymphohistiocytosis in
erature. Transplantation. 2004;77:238–43. pediatric cases: a multicenter study from Turkey. Turk J Pediatr.
8. Taniai N, Akimaru K, Kawano Y, Mizuguchi Y, Shimizu T, 2009;51:207–13.
Takahashi T, et al. Hemophagocytic syndrome after living-donor 27. Osugi Y, Hara J, Tagawa S, Takai K, Hosoi G, Matsuda Y, et al.
liver transplantation for fulminant liver failure: a case report. Cytokine production regulating Th1 and Th2 cytokines in he-
Hepatogastroenterology. 2005;52:923–6. mophagocytic lymphohistiocytosis. Blood. 1997;89:4100–3.
9. Boelens JJ, Lazo G, Gaiser JF, Wulffraat NM. Epstein-Barr virus- 28. Takada H, Ohga S, Mizuno Y, Nomura A, Hara T. Increased IL-
associated haemophagocytic lympho-histiocytosis after stem cell 16 levels in hemophagocytic lymphohistiocytosis. J Pediatr
transplantation. Bone Marrow Transpl. 2006;38:709–10. Hematol Oncol. 2004;26:567–73.
10. Cetica V, Pende D, Griffiths GM, Arico M. Molecular basis of 29. Takada H, Nomura A, Ohga S, Hara T. Interleukin-18 in hemo-
familial hemophagocytic lymphohistiocytosis. Haematologica. phagocytic lymphohistiocytosis. Leuk Lymphoma. 2001;42:
2010;95:538–41. 21–8.

123
Med Oncol (2013) 30:740 Page 7 of 7

30. Takada H, Takahata Y, Nomura A, Ohga S, Mizuno Y, Hara T. 46. Thompson PA, Allen CE, Horton T, Jones JY, Vinks AA,
Increased serum levels of interferon-gamma-inducible protein 10 McClain KL. Severe neurologic side effects in patients being
and monokine induced by gamma interferon in patients with treated for hemophagocytic lymphohistiocytosis. Pediatr Blood
haemophagocytic lymphohistiocytosis. Clin Exp Immunol. Cancer. 2009;52:621–5.
2003;133:448–53. 47. Marsh RA, Allen CE, McClain KL, Weinstein JL, Kanter J,
31. Trottestam H, Horne A, Arico M, Egeler RM, Filipovich AH, Skiles J, et al. Salvage therapy of refractory hemophagocytic
Gadner H, et al. Chemoimmunotherapy for hemophagocytic lymphohistiocytosis with alemtuzumab. Pediatr Blood Cancer.
lymphohistiocytosis: long-term results of the HLH-94 treatment 2013;60:101–9.
protocol. Blood. 2011;118:4577–84. 48. Strout MP, Seropian S, Berliner N. Alemtuzumab as a bridge to
32. Okamoto M, Yamaguchi H, Isobe Y, Yokose N, Mizuki T, Tajika allogeneic SCT in atypical hemophagocytic lymphohistiocytosis.
K, et al. Analysis of triglyceride value in the diagnosis and Nat Rev Clin Oncol. 2010;7:415–20.
treatment response of secondary hemophagocytic syndrome. 49. Mahlaoui N, Ouachee-Chardin M, de Saint Basile G, Neven B,
Intern Med. 2009;48:775–81. Picard C, Blanche S, et al. Immunotherapy of familial hemo-
33. Horne A, Trottestam H, Arico M, Egeler RM, Filipovich AH, phagocytic lymphohistiocytosis with antithymocyte globulins: a
Gadner H, et al. Frequency and spectrum of central nervous single-center retrospective report of 38 patients. Pediatrics.
system involvement in 193 children with haemophagocytic 2007;120:e622–8.
lymphohistiocytosis. Br J Haematol. 2008;140:327–35. 50. Verbsky JW, Grossman WJ. Hemophagocytic lymphohistiocy-
34. Hallahan AR, Carpenter PA, O’Gorman-Hughes DW, Vowels tosis: diagnosis, pathophysiology, treatment, and future perspec-
MR, Marshall GM. Haemophagocytic lymphohistiocytosis in tives. Ann Med. 2006;38:20–31.
children. J Paediatr Child Health. 1999;35:55–9. 51. Henter JI, Samuelsson-Horne A, Aricò M, Egeler RM, Elinder G,
35. Henter JI, Elinder G. Cerebromeningeal haemophagocytic lym- Filipovich AH, et al. Treatment ofhemophagocytic lymphohisti-
phohistiocytosis. Lancet. 1992;339:104–7. ocytosis with HLH-94 immunochemotherapy and bone marrow
36. Tong H, Ren Y, Liu H, Xiao F, Mai W, Meng H, et al. Clinical transplantation. Blood. 2002;100:2367–73.
characteristics of T-cell lymphoma associated with hemophago- 52. Yoon HS, Im HJ, Moon HN, Lee JH, Kim HJ, Yoo KH, et al. The
cytic syndrome: comparison of T-cell lymphoma with and outcome of hematopoietic stem cell transplantation in Korean
without hemophagocytic syndrome. Leuk Lymphoma. 2008;49: children with hemophagocytic lymphohistiocytosis. Pediatr
81–7. Transpl. 2010;14:735–40.
37. Risdall RJ, McKenna RW, Nesbit ME, Krivit W, Balfour HH Jr, 53. Ohga S, Kudo K, Ishii E, Honjo S, Morimoto A, Osugi Y, et al.
Simmons RL, et al. Virus-associated hemophagocytic syndrome: Hematopoietic stem cell transplantation for familial hemophag-
a benign histiocytic proliferation distinct from malignant histio- ocytic lymphohistiocytosis and Epstein-Barr virus-associated
cytosis. Cancer. 1979;44:993–1002. hemophagocytic lymphohistiocytosis in Japan. Pediatr Blood
38. Takahashi N, Miura I, Chubachi A, Miura AB, Nakamura S. A Cancer. 2010;54:299–306.
clinicopathological study of 20 patients with T/natural killer 54. Sparber-Sauer M, Honig M, Schulz AS, zur Stadt U, Schutz C,
(NK)-cell lymphoma-associated hemophagocytic syndrome with Debatin KM, et al. Patients with early relapse of primary he-
special reference to nasal and nasal-type NK/T-cell lymphoma. mophagocytic syndromes or with persistent CNS involvement
Int J Hematol. 2001;74:303–8. may benefit from immediate hematopoietic stem cell transplan-
39. Shin HJ, Chung JS, Lee JJ, Sohn SK, Choi YJ, Kim YK, et al. tation. Bone Marrow Transpl. 2009;44:333–8.
Treatment outcomes with CHOP chemotherapy in adult patients 55. Marsh RA, Jordan MB, Filipovich AH. Reduced-intensity con-
with hemophagocytic lymphohistiocytosis. J Korean Med Sci. ditioning haematopoietic cell transplantation for haemophago-
2008;23:439–44. cytic lymphohistiocytosis: an important step forward. Br J
40. Fitzgerald NE, MacClain KL. Imaging characteristics of hemo- Haematol. 2011;154:556–63.
phagocytic lymphohistiocytosis. Pediatr Radiol. 2003;33: 56. Imashuku S, Teramura T, Tauchi H, Ishida Y, Otoh Y, Sawada
392–401. M, et al. Longitudinal follow-up of patients with Epstein-Barr
41. Zhang K, Filipovich AH, Johnson J, Marsh RA, Villanueva J. virus-associated hemophagocytic lymphohistiocytosis. Haemato-
Hemophagocytic lymphohistiocytosis, familial. In: Pagon RA, logica. 2004;89:183–8.
Bird TD, Dolan CR, Stephens K, Adam MP, editors. SourceGe- 57. Imashuku S, Hibi S, Todo S, Sako M, Inoue M, Kawa K, et al.
neReviewsTM [Internet]. Seattle, WA: University of Washington, Allogeneic hematopoietic stem cell transplantation for patients
Seattle; 1993–2006 [updated 2010 Sep 07]. with hemophagocytic syndrome (HPS) in Japan. Bone Marrow
42. Ambruso DR, Hays T, Zwartjes WJ, Tubergen DG, Favara BE. Transpl. 1999;23:569–72.
Successful treatment of lymphohistiocytic reticulosis with 58. Yu JT, Hwang WL, Wang RC, Teng CL. Reduced intensity
phagocytosis with epipodophyllotoxin VP 16-213. Cancer. conditioning allogeneic hematopoietic stem cell transplant could
1980;45:2516–20. be beneficial to angioimmunoblastic T-cell lymphoma patients
43. Janka GE. Familial hemophagocytic lymphohistiocytosis: therapy with hemophagocytic lymphohistiocytosis. Ann Hematol.
in the German experience. Pediatr Hematol Oncol. 1989;6: 2012;91:805–7.
227–31. 59. Wang S, Degar BA, Zieske A, Shafi NQ, Rose MG. Hemo-
44. Henter JI, Elinder G, Finkel Y, Soder O. Successful induction phagocytosis exacerbated by G-CSF/GM-CSF treatment in a
with chemotherapy including teniposide in familial erythro- patient with myelodysplasia. Am J Hematol. 2004;77:391–6.
phagocytic lymphohistiocytosis. Lancet. 1986;2:1402. 60. Jordan MB, Allen CE, Weitzman S, Filipovich AH, McClain KL.
45. Stephan JL, Donadieu J, Ledeist F, Blanche S, Griscelli C, How I treat hemophagocytic lymphohistiocytosis. Blood. 2011;
Fischer A. Treatment of familial hemophagocytic lymphohistio- 118:4041–52.
cytosis with antithymocyte globulins, steroids, and cyclosporin A.
Blood. 1993;82:2319–23.

123

You might also like