kir–hla receptor-ligand mismatch associated with a graft-versus-tumor effect in haploidentical...

5
HNPCC-associated cancers, but without hematological malig- nancy and brain tumor. Eur J Hum Genet 2006;14:561–566. 16. Scott RH, Mansour S, Pritchard-Jones K, et al. Medulloblastoma, acute myelocytic leukemia and colonic carcinomas in a child with biallelic MSH6 mutations. Nat Clin Pract Oncol 2007;4:130 – 134. 17. Rahner N, Hoefler G, Hogenauer C, et al. Compound hetero- zygosity for two MSH6 mutations in a patient with early onset colorectal cancer, vitiligo and systemic lupus erythematosus. Am J Med Genet A 2008;146A:1314 – 1319. 18. Etzler J, Peyrl A, Zatkova A, et al. RNA-based mutation analysis identifies an unusual MSH6 splicing defect and circumvents PMS2 pseudogene interference. Hum Mutat 2008;29:299–305. 19. Hayward BE, De Vos M, Valleley EM, et al. Extensive gene conversion at the PMS2 DNA mismatch repair locus. Hum Mutat 2007;28:424–430. 20. Fedier A, Fink D. Mutations in DNA mismatch repair genes: Implications for DNA damage signaling and drug sensitivity (review). Int J Oncol 2004;24:1039–1047. KIR–HLA Receptor-Ligand Mismatch Associated With a Graft-Versus-Tumor Effect in Haploidentical Stem Cell Transplantation for Pediatric Metastatic Solid Tumors Antonio Pe ´rez-Martı ´nez, MD, PhD, 1 * Wing Leung, MD, PhD, 2 Evangelina Mun ˜oz, 1 Rekha Iyengar, PhD, 2 Manuel Ramı ´rez, MD, PhD, 1 Jose ´ Luis Vicario, PhD, 3 A ´ lvaro Lassaletta, MD, 1 Julia ´n Sevilla, MD, PhD, 1 Marta Gonza ´lez-Vicent, MD, PhD, 1 Luis Madero, MD, PhD, 1 and Miguel A ´ ngel Dı ´az-Pe ´rez, MD, PhD 1 INTRODUCTION Little progress has been made in the survival of children with metastatic and refractory solid tumors [1]. Allogeneic hemato- poietic stem cell transplantation (HSCT) has been proposed as a potential curative alternative in patients with refractory malignan- cies [2]. A possible graft-versus-tumor (GVT) effect has been documented in children with metastatic and relapsed Ewing sarcoma [3], neuroblastoma [4], melanoma [5], and hepatoblastoma [6]. However, the immunological mechanisms that mediate the GVT effect have not been elucidated. Early immune reconstitution after a conditioning regimen is led by natural killer (NK) cells, and for several weeks, they represent the only detectable lymphoid population. NK cell alloreactivity in vitro may be predicted by the lack of ligands for inhibitory killer immunoglobulin-like receptors (KIRs) [7]. Inhibitory KIRs bind with four specificities for polymorphic human leukocyte antigen (HLA) class I molecules. NK cell reactivity may be further enhanced by a Th1 cytokine environment and by T-cell lymphopenia [8,9]. To evaluate the potential of NK cell-mediated GVT effect and its immunological mechanisms, we performed a pilot study of haploidentical HSCT in three children with refractory tumors with no known cure. METHODS Patients and Transplantation The study involved three patients with metastatic solid tumors that were refractory to chemotherapy (Table I). This study was approved by the Ethical Committee of Hospital Nin ˜o Jesu ´s. Informed consent was obtained in accordance with the Helsinki Declaration. HSCT was performed using parental CD3/ CD19 depleted G-CSF mobilized peripheral blood. Reduced- intensity conditioning consisted of busulfan (4 mg/kg/day, 2 days), fludarabine (30 mg/m 2 /day, 5 days), thiotepa (5 mg/kg/day, 2 days), and methylprednisolone (4 mg/kg/day, 5 days). Methotrexate (15 mg/m 2 /day, day þ1, and 10 mg/m 2 /day, days þ3 and þ6) and cyclosporine (3 mg/kg/day, day 1 until 1 month after the infusion) were also administered. Immune reconstitution and hematopoietic chimerism were assessed monthly after HSCT. Computed tomo- graphy and magnetic resonance imaging were performed before and 3 months after HSCT. HLA Typing and KIR Genotyping, Phenotyping, and Cytotoxicity HLA-C allotypes (C1 and C2) and HLA-B allotypes (Bw4) were determined using high-resolution polymerase chain reaction- Killer immunoglobulin-like receptors (KIRs) on natural killer cells (NKs) recognize groups of human leukocyte antigen (HLA) class I alleles. Cells without an inhibitory HLA ligand may trigger NK activation. Reduced risk of relapse has been reported in malignant hematologic diseases after haploidentical transplantation when HLA ligands against the inhibitory KIRs present in the donor were absent in the recipient. We performed haploidentical transplant in three children with refractory solid tumors. Our results showed that beneficial antitumor effects could be observed in the presence of inhibitory KIR–HLA mismatch. These preliminary results suggest a possible association between disease control and NK cell allor- eactivity. Pediatr Blood Cancer 2009;53:120–124. ß 2009 Wiley-Liss, Inc. Key words: haploidentical stem cell transplantation; KIR-HLA mismatch; NK cells; pediatric solid tumors —————— 1 Department of Pediatric Hematology and Oncology, Hospital Nin ˜o Jesu ´s, Universidad Auto ´noma de Madrid, Madrid, Spain; 2 Department of Oncology, St. Jude Children’s Research Hospital, Memphis, Tennessee; 3 Laboratorio de Histocompatibilidad y Biologı ´a Molecular, Centro de Transfusio ´n de Madrid, Madrid, Spain Grant sponsor: Spanish National Health Service; Grant number: FIS CM-04/00011. *Correspondence to: Antonio Pe ´rez-Martı ´nez, Servicio de Oncologı ´a y Trasplante del Hospital Infantil Universitario Nin ˜o Jesu ´s, Mene ´ndez Pelayo 69, Madrid 28009, Spain. E-mail: [email protected] Received 7 November 2008; Accepted 6 January 2009 ß 2009 Wiley-Liss, Inc. DOI 10.1002/pbc.21955 Published online 12 February 2009 in Wiley InterScience (www.interscience.wiley.com) 120 Brief Reports

Upload: antonio-perez-martinez

Post on 06-Jul-2016

212 views

Category:

Documents


0 download

TRANSCRIPT

HNPCC-associated cancers, but without hematological malig-

nancy and brain tumor. Eur J Hum Genet 2006;14:561–566.

16. Scott RH, Mansour S, Pritchard-Jones K, et al. Medulloblastoma,

acute myelocytic leukemia and colonic carcinomas in a child with

biallelic MSH6 mutations. Nat Clin Pract Oncol 2007;4:130–134.

17. Rahner N, Hoefler G, Hogenauer C, et al. Compound hetero-

zygosity for two MSH6 mutations in a patient with early onset

colorectal cancer, vitiligo and systemic lupus erythematosus. Am J

Med Genet A 2008;146A:1314–1319.

18. Etzler J, Peyrl A, Zatkova A, et al. RNA-based mutation analysis

identifies an unusual MSH6 splicing defect and circumvents PMS2

pseudogene interference. Hum Mutat 2008;29:299–305.

19. Hayward BE, De Vos M, Valleley EM, et al. Extensive gene

conversion at the PMS2 DNA mismatch repair locus. Hum Mutat

2007;28:424–430.

20. Fedier A, Fink D. Mutations in DNA mismatch repair genes:

Implications for DNA damage signaling and drug sensitivity

(review). Int J Oncol 2004;24:1039–1047.

KIR–HLA Receptor-Ligand Mismatch Associated With a Graft-Versus-Tumor Effectin Haploidentical Stem Cell Transplantation for Pediatric Metastatic Solid Tumors

Antonio Perez-Martınez, MD, PhD,1* Wing Leung, MD, PhD,2 Evangelina Munoz,1 Rekha Iyengar, PhD,2

Manuel Ramırez, MD, PhD,1 Jose Luis Vicario, PhD,3 Alvaro Lassaletta, MD,1 Julian Sevilla, MD, PhD,1

Marta Gonzalez-Vicent, MD, PhD,1 Luis Madero, MD, PhD,1 and Miguel Angel Dıaz-Perez, MD, PhD1

INTRODUCTION

Little progress has been made in the survival of children with

metastatic and refractory solid tumors [1]. Allogeneic hemato-

poietic stem cell transplantation (HSCT) has been proposed as a

potential curative alternative in patients with refractory malignan-

cies [2]. A possible graft-versus-tumor (GVT) effect has been

documented in children with metastatic and relapsed Ewing

sarcoma [3], neuroblastoma [4], melanoma [5], and hepatoblastoma

[6]. However, the immunological mechanisms that mediate the

GVT effect have not been elucidated. Early immune reconstitution

after a conditioning regimen is led by natural killer (NK) cells, and

for several weeks, they represent the only detectable lymphoid

population. NK cell alloreactivity in vitro may be predicted by the

lack of ligands for inhibitory killer immunoglobulin-like receptors

(KIRs) [7]. Inhibitory KIRs bind with four specificities for

polymorphic human leukocyte antigen (HLA) class I molecules.

NK cell reactivity may be further enhanced by a Th1 cytokine

environment and by T-cell lymphopenia [8,9].

To evaluate the potential of NK cell-mediated GVT effect and

its immunological mechanisms, we performed a pilot study of

haploidentical HSCT in three children with refractory tumors with

no known cure.

METHODS

Patients and Transplantation

The study involved three patients with metastatic solid

tumors that were refractory to chemotherapy (Table I). This

study was approved by the Ethical Committee of Hospital Nino

Jesus. Informed consent was obtained in accordance with the

Helsinki Declaration. HSCT was performed using parental CD3/

CD19 depleted G-CSF mobilized peripheral blood. Reduced-

intensity conditioning consisted of busulfan (4 mg/kg/day, 2 days),

fludarabine (30 mg/m2/day, 5 days), thiotepa (5 mg/kg/day, 2 days),

and methylprednisolone (4 mg/kg/day, 5 days). Methotrexate

(15 mg/m2/day, day þ1, and 10 mg/m2/day, days þ3 and þ6) and

cyclosporine (3 mg/kg/day, day�1 until 1 month after the infusion)

were also administered. Immune reconstitution and hematopoietic

chimerism were assessed monthly after HSCT. Computed tomo-

graphy andmagnetic resonance imagingwere performed before and

3 months after HSCT.

HLA Typing and KIR Genotyping, Phenotyping,and Cytotoxicity

HLA-C allotypes (C1 andC2) andHLA-B allotypes (Bw4) were

determined using high-resolution polymerase chain reaction-

Killer immunoglobulin-like receptors (KIRs) on natural killer cells(NKs) recognize groups of human leukocyte antigen (HLA) class Ialleles. Cells without an inhibitory HLA ligand may trigger NKactivation. Reduced risk of relapse has been reported in malignanthematologic diseases after haploidentical transplantation when HLAligands against the inhibitory KIRs present in the donor were absentin the recipient. We performed haploidentical transplant in three

children with refractory solid tumors. Our results showed thatbeneficial antitumor effects could be observed in the presence ofinhibitory KIR–HLA mismatch. These preliminary results suggest apossible association between disease control and NK cell allor-eactivity. Pediatr Blood Cancer 2009;53:120–124.� 2009 Wiley-Liss, Inc.

Key words: haploidentical stem cell transplantation; KIR-HLA mismatch; NK cells; pediatric solid tumors

——————1Department of Pediatric Hematology and Oncology, Hospital Nino

Jesus, Universidad Autonoma de Madrid, Madrid, Spain; 2Department

of Oncology, St. Jude Children’s Research Hospital, Memphis,

Tennessee; 3Laboratorio de Histocompatibilidad y Biologıa

Molecular, Centro de Transfusion de Madrid, Madrid, Spain

Grant sponsor: Spanish National Health Service; Grant number: FIS

CM-04/00011.

*Correspondence to: Antonio Perez-Martınez, Servicio de Oncologıa y

Trasplante del Hospital Infantil Universitario Nino Jesus, Menendez

Pelayo 69, Madrid 28009, Spain.

E-mail: [email protected]

Received 7 November 2008; Accepted 6 January 2009

� 2009 Wiley-Liss, Inc.DOI 10.1002/pbc.21955Published online 12 February 2009 in Wiley InterScience(www.interscience.wiley.com)

120 Brief Reports

sequence-based typing. Fifteen human KIR genes and two

pseudogenes were analyzed by polymerase chain reaction with a

KIR typing kit (MiltenyiBiotec, Inc., Auburn,CA).Antibodies used

to identify KIR phenotypes by flow cytometry were CD45-FITC,

CD19/CD20-PE, CD56-APC, and CD158a,h (EB6)-PE (Beckman

Coulter, Fullerton, CA); CD3-PECY7 and CD158b1/b2,j (CHL)-

FITC (Becton Dickinson, Franklin Lakes, NJ); and CD158e1

(DX9)-FITC, CD158e1 (DX9)-PE, and CD158a,h (EB6)-FITC

(Miltenyi Biotec, Inc.). Natural cytotoxicity of donor unstimulated

fresh NK cells against K562 leukemia cells was assessed in a

conventional 2-hr europium-TDA release assay (Perkin-Elmer

Wallac, Turku, Finland). NK cells were magnetically isolated from

peripheral blood of donors using standard density gradient

centrifugation over Ficoll-Paque Plus (Pharmacia, Uppsala,

Sweden) and NK cell isolation kit from Miltenyi Biotec, Inc.,

following the manufacturer’s specifications. Labeled cells were

processed on a QuadroMACS separator with LS columns (Miltenyi

Biotec, Inc.). Purity was higher than 95% of CD56þCD3� cells.

RESULTS

Patient 1

A 4-year-old male with an embryonal rhabdomyosarcoma of

the right testicle and lung metastasis underwent surgery and

chemotherapy according to the Malignant Mesenchymal Tumor

Committee MMT 98 protocol [10]. Because lung metastasis

persisted, he received second-line chemotherapy with topotecan/

cyclophosphamide and then third-line chemotherapy with irinote-

can/temozolomide, resulting in partial response. HSCT was then

performed using a parental donor mismatched at KIR2DL1 with

missing HLA-CLys80 in the recipient. In addition, donor NK cells

showed high lysis to K562 cells in vitro and possessed three

activating genes KIR2DS1, KIR2DS3, and KIR3DS1 (Fig. 1A–C);

the latter twomay interact with the HLA-CAsn80 and Bw4 present in

the recipient. The patient achieved full donor chimerism 1 month

after transplantation, and cyclosporine was withdrawn. Ten days

later, the patient presented with herpes zoster infection and

pancytopenia. Bone marrow chimerism was 98% recipient.

Secondary graft failure was diagnosed and treated with steroids,

cyclosporine, and granulocyte colony-stimulating factor. Because

no responsewas observed, a secondHSCT from the same donor was

performed 3 months later, adding muromonab (an anti-CD3

monoclonal, 0.1 mg/kg/d, 7 days) to the conditioning regimen

described above. Full donor chimerism was reached 1 month after

the second HSCT, and no toxicities were observed. Since then, NK

cells were the predominant lymphocytes throughout the first year

after transplantation and lung metastasis no longer appeared in

computed tomography evaluations (Fig. 1D). Ayear and a half after

Pediatr Blood Cancer DOI 10.1002/pbc

TABLE I. Patients Characteristics and Outcome

Subject Patient 1 Patient 2 Patient 3

Tumor Embryonal rhabdomyosarcoma Neuroblastoma Ewing sarcoma

Metastasis Lung Bone/BM Bone /BM

Chemotherapy MMT IV 98.11 HR-NBL’1/ESIOP SEOP 2001

Surgery Yes Yes Yes

Radiotherapy No Yes No

Second lines chemotherapies TPT-CCF/IRT-TMZ TVD/IRT-TMZ ICE/TPT-CCF/IRT-TMZ

High dose chemotherapy No Yes No

Pre-HSCT status Partial remission Metastatic relapse Metastatic relapse

Graft content: CD34/CD3/CD56 (�106/kg) 5.7/0.2/9 5.2/0.34/60 5.8/0.8/17

Toxicities Zoster None None

NE/PE (day) 12/15 13/14 15/16

Post-HSCT response Complete remission No response Partial response for 5 months

Outcome/time after HSCT Alive/21þ months DOPD/10 months DOPD/9 months

HLA-B

Patient 44/52 (Bw4/4) 49/55 (Bw4/6) 51/41 (Bw4/6)

Donor 44/49 (Bw4/4) 44/49 (Bw4/4) 41/41 (Bw6/6)

HLA-Cw

Patient 12/16 (Asn80/Asn80) 3/6 (Asn80/Lys80) 15/17 (Lys80/Lys80)

Donor 7/16 (Asn80/Asn80) 6/6 (Lys80/Lys80) 7/17 (Asn80/Lys80)

KIR2DL1 (%)

Patient — — —

Donor 12.6 6 10.9

KIR2DL2/3 (%)

Patient — — —

Donor 28.2 25.2 33.8

KIR3DL1 (%)

Patient — — —

Donor 2.5 3.4 22.3

KIR–HLA

Receptor-ligand No Lys80 All ligands present No Asn80

Mismatch KIR2DL1 mismatch No KIR–HLA mismatch KIR2DL2/3 mismatch

BM, bone marrow; NE, neutrophil engraftment; PE, platelet engraftment; TPT-CCF, topotecan cyclophosphamide; IRT-TMZ irinotecan-

temozolomide; TVD, topotecan-vincristine-doxorubicin; ICE, ifosfamide-carboplatin-etoposide; DOPD, died of progressive disease; HSCT,

haploidentical stem cell transplantation.

Brief Reports 121

Pediatr Blood Cancer DOI 10.1002/pbc

Fig. 1. KIR NK cell profile, immune reconstitution and clinical response after haploidentical stem cell transplantation. A: KIR inhibitory donor

phenotype and KIR donor genotyping. B: Patient’s immune reconstitution after haploidentical HSCT. C: Donor’s NK cytotoxicity against K562

cells.D: Lung computed tomography of Patient 1withmetastatic rhabdomyosarcoma at diagnosis and after chemotherapy. The far right panel shows

the absence of metastatic lesions in the lung 1 year after HSCT.E: Short tau inversion recovery saggital magnetic resonance images of lumbar spine

of Patient 3. The left panel, before HSCT, shows several focal bone marrow lesions, most pronounced at L2–L3. The right panel shows complete

resolution of the lesions and fatty replacement of the vertebral bone marrow, indicating a positive response.

122 Brief Reports

the second HSCT, the patient was still alive with full donor

chimerism, donor NK genotype, high lysis to K562 cells and

complete remission.

Patient 2

A 16-year-old female with a stage IV suprarenal neuroblastoma

and metastasis to bone and marrow was treated according to the

Societe Internationale d’Oncologie Pediatrique HR-NBL-1/ESIOP

protocol [11]. Chemotherapy was followed by primary tumor

resection. Evaluation after treatment showed progressive disease in

bone marrow and suprarenal gland. Because first-line treatment

failed, three cycles of topotecan/vincristine/doxorubicin (TVD) and

high-dose chemotherapy (busulfan/melphalan) with autologous

stem cell rescuewere administered. Partial remission was achieved,

but 3 months later, progressive disease was observed in the dorsal

column, bone marrow, and right suprarenal gland. Three cycles of

TVD plus third-line chemotherapy (irinotecan/temozolomide)

were given with minimal response. HSCT was then perform-

ed with a parental donor with no inhibitory KIR–HLA

mismatch. Furthermore, the donor NK cells had very poor lytic

activity against K562 cells in vitro and possessed only two

activating genesKIR2DS2 and 2DS3 but notKIR2DS1orKIR3DS1

(Fig. 1A–C). Full donor chimerism was reached 1 month

after HSCT, and no toxicities were observed. NK cells were the

most numerous during the first 3 months after transplantation

with rapid reconstitution of CD3þ lymphocytes thereafter. Her

tumors had no objective response after HSCT and progressed at

6 months in the dorsal and lumbar regions associated with medullar

compression. The patient died of neuroblastoma 10 months

after HSCT.

Patient 3

A 17-year-old male received chemotherapy according to the

Spanish Society of Pediatric Oncology SEOP-2001 protocol for

group 1 right fibula Ewing sarcoma. The regimen consisted of an

induction phase of vincristine/ifosfamide/etoposide/doxorubicin,

right lower extremity amputation, and a consolidation phase of

vincristine/actinomycin/cyclophosphamide. Seven months after

treatment, relapsed disease was found in bone marrow and bone

(lumbar spine and humerus). Second-line chemotherapy (ifosfa-

mide/carboplatin/etoposide) was given without response. He then

received third-line chemotherapy with two cycles of topotecan/

cyclophosphamide and two cycles of irinotecan/temozolomide.

Despite persistent disease, HSCT was performed using a parental

donor mismatched at KIR2DL2/3 with missing HLA-CAsn80 in the

recipient. Donor NK cells showed intermediate natural cytotoxicity

against K562 cells and possessed three activating genes KIR2DS1,

KIR2DS2, and KIR3DS1 (Fig. 1A,C); the latter two may interact

with the HLA-CLys80 and Bw4 present in the recipient. Full donor

chimerism was reached 1 month after HSCT, and no toxicities

were observed. In the first 3 months after transplantation, NK cells

were the predominant lymphocyte population and a total response in

the vertebral bones were observed (Fig. 1E). CD3þ lymphocytes

then rapidly reconstituted in the following 2 months, with falling

NK cell counts and progressive tumors in lumbar spine, humerus,

and lung appeared 5 months after transplantation. The patient died

of progressive disease 9 months after HSCT.

DISCUSSION

Clinical response has been observed in previous pediatric solid

tumor allogeneic transplantations. Although a GVT effect was

suggested (in addition to the conditioning chemotherapy effect), the

actual immune mechanisms have never been elucidated. For

instance, graft-versus-neuroblastoma effect has been suggested

in haploidentical transplantation for advanced disease [5,12].

Refractory metastatic Ewing sarcoma and rhabdomyosarcoma has

been treated with allogeneic transplant [4,13,14]. An immuno-

logical effect associatedwith graft-versus-host disease (GVHD) has

been described in hepatoblastoma [6], and a rapid recovery of NK

cytotoxic activity was describe in successful cases of metastatic

osteosarcoma after allogeneic bonemarrow transplantation [15,16].

The results of our preliminary study suggest that a clinically

beneficial GVT effect might be mediated by donor–recipient

inhibitory KIR–HLA mismatched NK cells. Clinical response was

observed in the two patients with a KIR–HLA mismatched donor

during the time when NK cells were the major lymphocyte

population. In addition, the degree of tumor response appeared to

correlate with the donor NK natural cytotoxicity [17], the disease

status at the time of transplantation, the number of KIR activating

receptors (2DS1, 2DS3, 3DS1), and possibly with the presence of

activating KIR3DS1–Bw4 interaction. These findings raise the

possibility that KIR is a major biological determinant of antitumor

effect in pediatric cancer, in addition to hematologic malignancies

as demonstrated in adult AML by the Perugia group [18,19] and in

pediatric AML and ALL by the Memphis group [20].

Our observation that tumor responses were only seen in those

patients with inhibitory KIR–HLA mismatched NK cells and

provide suggestive evidence that allogeneic NK cells might be

the effector cells that mediated GVT effects in the setting of a

mismatched allogeneic transplant. Although our results are

preliminary, this report provides novel data to support further

investigation in the use of KIR–HLA mismatched HSCT for the

treatment of refractory childhood solid tumors.

ACKNOWLEDGMENT

The authors want to thank Dr. Sara Sirvent and Dr. Gustavo

Albi for computed tomography andmagnetic resonance images, and

Dr. David Galloway for scientific editing. The authors also want

to thank Fundacion Inocente for supporting the project ‘‘Allogeneic

Stem Cell Transplantation in Pediatric Solid Tumors.’’ A.

Perez-Martınez was supported by Spanish National Health Service

grant FIS CM-04/00011.

REFERENCES

1. Krishnan K, Khanna C, Helman LJ. The biology of metastases in

pediatric sarcomas. Cancer J 2005;11:306–333.

2. Childs R, Chernoff A, Contentin N, et al. Regression of metastatic

renal-cell carcinoma after nonmyeloablative allogeneic peripheral-

blood stem-cell transplantation. N Engl JMed 2000;343:750–758.

3. Koscielniak E, Gross-Wietsch U, Treuner J, et al. Graft-versus-

Ewing sarcoma effect and long-term remission induced by

haploidentical stem-cell transplantation in a patient with relapse

of metastatic disease. J Clin Oncol 2005;23:242–244.

4. Lang P, Pfeiffer M, Muller I, et al. Haploidentical stem cell

transplantation in patients with pediatric solid tumors: Preliminary

Pediatr Blood Cancer DOI 10.1002/pbc

Brief Reports 123

results of a pilot study and analysis of graft versus tumor effects.

Klin Padiatr 2006;218:321–326.

5. Kasow KA, Handgretinger R, Krasin MJ, et al. Possible allogeneic

graft-versus-tumor effect in childhood melanoma. J Pediatr

Hematol Oncol 2003;25:982–986.

6. Inaba H, Handgretinger R, Furman W, et al. Allogeneic graft-

versus-hepatoblastoma effect. Pediatr BloodCancer 2006;46:501–

505.

7. Re F, Staudacher C, Zamai L, et al. Killer cell Ig-like receptors

ligand-mismatched, alloreactive natural killer cells lyse primary

solid tumors. Cancer 2006;107:640–648.

8. Lowe EJ, Turner V, Handgretinger R, et al. T-cell alloreactivity

dominates natural killer cell alloreactivity in minimally T-cell-

depleted HLA-non-identical paediatric bone marrow transplanta-

tion. Br J Haematol 2003;123:323–326.

9. Leung W, Handgretinger R, Iyengar R, et al. Inhibitory KIR-HLA

receptor-ligand mismatch in autologous haematopoietic stem cell

transplantation for solid tumor and lymphoma. Br J Cancer 2007;

97:539–542.

10. OrbachD,ReyA,Oberlin O, et al. Soft tissue sarcoma ormalignant

mesenchymal tumors in the first year of life: Experience of the

International Society of Pediatric Oncology (SIOP) Malignant

Mesenchymal Tumor Committee. J Clin Oncol 2005;23:4363–

4371.

11. Ladenstein R, Philip T, Lasset C. Multivariate analysis of risk

factors in stage 4 neuroblastoma patients over the age of one year

treated with megatherapy and stem-cell transplantation: A report

from the European Bone Marrow Transplantation Solid Tumor

Registry. J Clin Oncol 1998;16:953–965.

12. InoueM, Nakano T, Yoneda A, et al. Graft-versus-tumor effect in a

patient with advanced neuroblastoma who received HLA haplo-

identical bone marrow transplantation. Bone Marrow Transplant

2003;32:103–106.

13. Lucas KG, Schwartz C, Kaplan J. Allogeneic stem cell trans-

plantation in a patient with relapsed Ewing sarcoma. Pediatr Blood

Cancer 2008;51:142–144.

14. Misawa A, Hosoi H, Tsuchiya K, et al. Regression of refractory

rhabdomyosarcoma after allogeneic stem-cell transplantation.

Pediatr Hematol Oncol 2003;20:151–155.

15. GoiK,SugitaK,TezukaT, et al.A successful caseof allogeneic bone

marrow transplantation for osteosarcoma with multiple metastases

of lung and bone. Bone Marrow Transplant 2006;37: 115–116.

16. Esartia PT, Deichman GI, Kluchareva TE, et al. Allogenic bone-

marrow transfusion suppresses development of lung metastases in

osteogenic sarcoma patients after radical surgery. Int J Cancer

1993;54:907–910.

17. Imai K,Matsuyama S,Miyake S, et al. Natural cytotoxic activity of

peripheral-blood lymphocytes and cancer incidence: An 11-year

follow-up study of a general population. Lancet 2000;356:1795–

1799.

18. Ruggeri L, Mancusi A, Perruccio K, et al. Natural killer cell

alloreactivity for leukemia therapy. J Immunother 2005;28:175–

182.

19. Ruggeri L, Capanni M, Urbani E, et al. Effectiveness of donor

natural killer cell alloreactivity in mismatched hematopoietic

transplants. Science 2002;15:2097–2100.

20. LeungW, Iyengar R, Turner V, et al. Determinants of antileukemia

effects of allogeneic NK cells. J Immunol 2004;172:644–650.

Congenital Glioblastoma

G.M. Milano, MD, PhD,1* C. Cerri, MD,1 V. Ferruzzi,1 I. Capolsini, MD,1 E. Mastrodicasa, MD,1

L. Genitori, MD,2 and F. Aversa, MD1

INTRODUCTION

First described by Holt [1], congenital brain tumors, which

account from 0.3% to 4% of CNS tumors in children are extremely

rare forms of childhood tumors. They often arise during pregnancy

and are identified only at autopsy [2,3]. In the century since the first

report, the entire cohort comprises nearly 600 cases, all of which

had a very poor prognosis [4]. Characteristics and outcomes of

200 congenital brain tumors were extensively discussed in a review

by Wakai et al. [7].

The Solitare and Krigman classification groups congenital

brain tumors according to time of diagnosis as: definitely congenital

(present at birth), probably congenital (detected within the 1st week

of life), and possibly congenital, that is, detected within the first

2months of life [6]. Other classifications take symptoms and time of

diagnosis into account and extend congenital tumor criteria to cover

the first year of life [6,7]. Although congenital growth does not

seem to be correlated with any particular tumor, glioblastoma,

and teratoma are most often diagnosed, with astrocytoma,

We describe the case a 2-day-old female with congenitalglioblastoma. Total resection was followed by adjuvant and highdose chemotherapy, as indicated by the current Italian infantprotocol. The child is alive and well 18 months after diagnosis. Areview of 67 selected congenital brain tumors showed the mortalityrate was 82%. Even though the majority of patients had glioblastoma,only 5/67 had received adjuvant therapy. To ensure optimal

outcomes, we recommend total or subtotal surgical resection,followed by adjuvant and high dose chemotherapy. Given the lackspecific protocols for congenital brain tumors an internationalconsensus seems to be needed, starting with congenital glioblas-toma. Pediatr Blood Cancer 2009;53:124–126.� 2009 Wiley-Liss, Inc.

Key words: chemotherapy; congenital brain tumors; high dose chemotherapy; infant

——————Additional Supporting Information may be found in the online version

of this article.

1Unit of Pediatric Oncology and Haematology, Ospedale ‘‘S. Maria

della Misericordia’’, A.O. di Perugia, Italy; 2Division of Neurosurgery,

Meyer Children’s Hospital, Florence, Italy

*Correspondence to: G.M. Milano, Division of Pediatric Oncology and

Haematology, Ospedale ‘‘S. Maria della Misericordia,’’ A.O. di

Perugia Sant’Andrea delle Fratte, 06156, Perugia, Italy.

E-mail: [email protected]

Received 27 December 2008; Accepted 10 February 2009

� 2009 Wiley-Liss, Inc.DOI 10.1002/pbc.22008Published online 23 March 2009 in Wiley InterScience(www.interscience.wiley.com)

124 Brief Reports