You are on page 1of 33

7/4/22, 05:44 Leucemia linfoblástica aguda - ScienceDirect

Clínicas de hematología/oncología de América del Norte


Volumen 23, número 4 , agosto de 2009 , páginas 655-674

Leucemia linfoblástica aguda


Mihaela Onciu MD

Mostrar más

Esquema Cuota Citar

https://doi.org/10.1016/j.hoc.2009.04.009 Obtener derechos y contenido

Acute lymphoblastic leukemia and lymphoblastic lymphoma constitute a family of genetically


heterogeneous lymphoid neoplasms derived from B- and T-lymphoid progenitors. Diagnosis is
based on morphologic, immunophenotypic, and genetic features that allow differentiation from
normal progenitors and other hematopoietic and nonhematopoietic neoplasms. Current
intensive chemotherapy regimens have accomplished overall cure rates of 85% to 90% in children
and 40% to 50% in adults, with outcomes depending on the genetic subtype of disease and
clinical features at presentation. Therapy is optimized using minimal residual disease studies
that employ flow cytometric and molecular methodologies, and are important determinants of
prognosis. Genetic analyses currently underway are likely to provide insight into biology,
mechanisms of relapse, pharmacogenetics, and new potential therapeutic targets, which should
aid in further improvement of outcome in this disease.

Previous Next

https://www.sciencedirect.com/science/article/pii/S0889858809000859 1/33
7/4/22, 05:44 Leucemia linfoblástica aguda - ScienceDirect

Keywords
Acute leukemia; Acute lymphoblastic leukemia; Lymphoblastic lymphoma;
Cytogenetics; Genes

Acute lymphoblastic leukemia (ALL) encompasses a group of lymphoid neoplasms that


morphologically and immunophenotypically resemble B-lineage and T-lineage precursor cells.
These neoplasms may present predominantly as a leukemic process, with extensive involvement
of the bone marrow and peripheral blood or may be limited to tissue infiltration, with absent or
only limited (less than 25%) bone marrow involvement. The latter cases are typically designated
as lymphoblastic lymphomas (LBLs). ALL and LBLs appear to constitute a biologic continuum,
although they may show distinct clinical features. The current World Health Organization
Classification of hematopoietic neoplasms designates these disorders as B- or T-lymphoblastic
leukemia/lymphoma.1

Epidemiology
Most ALL cases occur in children, with an incidence of 3 to 4/100,000 in patients 0 to 14 years of
age and ∼1/100,000 in patients older than 15 years, in the United States.2 In children, ALLs
represent 75% of all acute leukemias (which in turn represent 34% of all cancers in this age
group), with a peak incidence at 2 to 5 years of age.3 This percentage is much lower in adults, in
whom acute myeloid leukemias (AMLs) and chronic lymphocytic leukemias are more common.3,
4 There is a slight male predominance in all age groups and a significant excess incidence among
white children.2

ALL presents primarily as de novo disease, with only rare cases occurring as secondary
neoplasms.5 A variety of genetic and environmental factors have been related to ALL. It occurs
with increased frequency in patients with Down syndrome, Bloom syndrome, neurofibromatosis
type I, and ataxia-telangiectasia.6 In addition, exposure in utero to ionizing radiation, pesticides,
and solvents has also been related to an increased risk for childhood leukemia.6 Leukemia-
specific fusion genes or immunoglobulin (Ig) and clonal Ig gene rearrangements have been
identified in neonatal spot (Guthrie) cards of patients who later developed ALL.7, 8

Clinical presentation
The clinical onset of ALL is most often acute, although a small percentage of cases may evolve
insidiously over several months.9 The presenting symptoms and signs correlate with the
leukemic cell burden and the degree of marrow replacement, leading to cytopenias. The most
common symptoms include fever (caused by leukemia or a secondary infection secondary to
neutropenia), fatigue and lethargy (as a result of anemia), bone and joint pain, and a bleeding
https://www.sciencedirect.com/science/article/pii/S0889858809000859 2/33
7/4/22, 05:44 Leucemia linfoblástica aguda - ScienceDirect

diathesis (related to thrombocytopenia). Patients with precursor T-cell ALL/LBL often present
with a mediastinal mass with or without associated pleural effusions, lo que puede provocar
dificultad respiratoria y otros signos del síndrome de la vena cava superior . Los sitios
extramedulares comunes de afectación incluyen ganglios linfáticos, hígado, bazo y meninges ,
mientras que, con menos frecuencia, la LLA puede infiltrar tejidos orbitarios, testículos,
amígdalas y adenoides. Los pacientes raros que presentan B-LBL pueden mostrar lesiones
cutáneas de linfadenopatía en el área de la cabeza y el cuello o lesiones óseas discretas . 10Las
anomalías de laboratorio más comunes en la LLA incluyen anemia, trombocitopenia, neutropenia
yleucopeniao leucocitosis , con hiperleucocitosis (>100 × 109/L) presente en aproximadamente el
15% de los pacientes pediátricos. 9 Otras anomalías de laboratorio comunes incluyen niveles
elevados de ácido úrico sérico y lactosa deshidrogenasa , que se correlacionan con la carga
tumoral y el grado de lisis tumoral .

Diagnóstico patológico

Morfología
Las características morfológicas clásicas de la LLA han sido bien descritas en la literatura y se
resumen mejor en las categorías delineadas por la primera clasificación de la LLA, el sistema
franco-estadounidense-británico (FAB), que se basó principalmente en la apariencia microscópica
de la leucemia. células, como se ve en frotis teñidos con Wright-Giemsa. 11 , 12 La clasificación
FAB delineó tres grupos morfológicos de LLA, designados como L1, L2 y L3. ( Fig. 1 ) Más
comúnmente, los blastos de LLA son de tamaño pequeño a intermedio, con citoplasma escaso,
cromatina nuclear condensada y nucléolo indistinto o ausente.(Subtipo FAB L1). Con menos
frecuencia, las células de ALL pueden ser más grandes, con cantidades moderadas de citoplasma
basófilo pálido, cromatina nuclear finamente dispersa y nucleolos prominentes (subtipo FAB L2).
En muy raras ocasiones, la LLA puede presentarse como el subtipo FAB L3, que consiste en
grandes blastos, con abundante citoplasma profundamente basófilo y ocasionalmente vacuolado,
cromatina nuclear agrupada en grumos gruesos y nucleolos variablemente prominentes. La
mayoría de los casos que presentan esta morfología son linfomas de Burkitt , un subtipo de
linfoma de células B maduras de alto grado. Sin embargo, un pequeño subconjunto de neoplasias
de células B precursoras, a menudo asociado con hipodiploidía, también puede presentar
características FAB L3 (ver Fig. 1 ).

https://www.sciencedirect.com/science/article/pii/S0889858809000859 3/33
7/4/22, 05:44 Leucemia linfoblástica aguda - ScienceDirect

Descargar : Descargar imagen de alta resolución (989KB)


Descargar: Descargar imagen a tamaño completo

Figura 1 . La morfología de ALL/LBL en frotis y secciones de tejido incluidas en parafina. ( A, B )


TODO L1. C–D . TODO L2. ( E, F ) Hallazgos morfológicos en un caso de LLA-B precursora con
hipodiploidía, similar a un linfoma de células B maduras de alto grado ( A, C, E, tinción de
Wright-Giemsa) ( B, D, F, hematoxilina- eosina; aumento original 60x inmersión en aceite).

Cuando se observan en secciones histológicas teñidas con hematoxilina y eosina de biopsias de


médula ósea e infiltrados tisulares, los linfoblastos neoplásicos de ALL/LBL pueden mostrar
características que corresponden a las categorías FAB. El aspecto más característico es el de una
neoplasia con patrón de crecimiento difuso, a veces con aspecto de cielo estrellado parcial,
expandiendo el área interfolicular en órganos linfoides subtotalmente reemplazados (como

https://www.sciencedirect.com/science/article/pii/S0889858809000859 4/33
7/4/22, 05:44 Leucemia linfoblástica aguda - ScienceDirect

ganglios linfáticos o amígdalas). Los blastos son pequeños, con cromatina finamente granular y
pequeños nucléolos puntiformes. En casos raros, los blastos pueden tener nucléolos prominentes
(como en el subtipo L2) o pueden parecerse a células de linfoma de células grandes (ver Fig. 1 ).).
Los casos ocasionales de LLA pueden presentarse con grados variables de necrosis medular o
fibrosis medular asociada. 13

Se han descrito varias variantes morfológicas de la LLA, ninguna de las cuales tiene importancia
pronóstica. Los blastos leucémicos pueden contener vacuolas , 14 gránulos de color rosa pálido o
azurófilos ("LLA granular") 15 , 16 , 17 o inclusiones gigantes. 18 , 19 En algunos casos, los blastos
pueden mostrar proyecciones citoplásmicas excéntricas similares a las de los urópodos ("LLA de
células en espejo de la mano"). 20 Algunos casos de LLA pueden mostrar una eosinofilia asociada
prominente (e incluso un síndrome hipereosinofílico), 21 que se relaciona con una lesión genética
específica (consulte la sección posterior).

citoquímica
La tinción citoquímica se ha utilizado con una frecuencia decreciente en el diagnóstico de la LLA
debido a la disponibilidad de la inmunofenotipificación . Las células leucémicas de ALL son
uniformemente negativas para mieloperoxidasa (MPO), Sudan Black-B, cloroacetato esterasa y
esterasas no específicas . 22 Una excepción notable es la de la LLA granular, que con frecuencia
muestra una tinción de esterasa inespecífica localizada en los gránulos y también puede mostrar
una tinción gris claro con Sudan Black-B. 1 , 23 Los blastos de la LLA a menudo (75 %) son
positivos para PAS y también pueden ser positivos para la fosfatasa ácida , que se observa con
mayor frecuencia en la LLA de células T. 22

inmunofenotipo
Los estudios inmunofenotípicos son un componente esencial de la evaluación diagnóstica de
ALL/LBL. A diferencia de las características morfológicas, el linaje de la LLA establecido de esta
manera subdivide esta enfermedad en dos categorías amplias, clínica y biológicamente
significativas: LLA de células B precursoras (LLA-B) y LLA de células T precursoras (LLA-T).

B-ALL se caracteriza por la expresión de una variedad de antígenos específicos de células B, que a
menudo incluyen PAX-5 (proteína activadora específica de células B), CD19, CD20 , CD22
(superficial y citoplasmático), CD24 y CD79a (citoplasmático). En particular, CD20, un marcador
de células B maduras, puede expresarse solo de manera parcial y débil por los linfoblastos
leucémicos o puede ser completamente negativo en B-ALL. Una gran proporción de B-ALL
también expresan CD10 (antígeno de leucemia linfocítica aguda común), un antígeno expresado
de manera consistente por progenitores de células B normales. La mayoría de las B-ALL
muestran una expresión tenue de CD45 (antígeno común leucocitario), y un subconjunto de estas
leucemias, más común en niños, puede ser CD45 negativo. Otros antígenos expresados ​a menudo
por los blastos leucémicos incluyen marcadores progenitores que a menudo se observan en

https://www.sciencedirect.com/science/article/pii/S0889858809000859 5/33
7/4/22, 05:44 Leucemia linfoblástica aguda - ScienceDirect

precursores de células B en etapa temprana, incluidos CD34 ydesoxinucleotidil transferasa


terminal (Tdt). Según su patrón de expresión de Ig, la LLA-B puede clasificarse en LLA pre-B (o
pro-B) temprana, que carece de expresión de Ig, LLA-pre-B (expresión de cadenas μ citoplásmicas,
sin expresión de cadena ligera de Ig o restricción) y pre-B-ALL de transición (con expresión
citoplásmica y superficie débil de cadenas μ, sin expresión ni restricción de cadenas ligeras de Ig).
1 Los casos raros de LLA-B precursora pueden mostrar expresión superficial de IgM completa con

restricción de cadena ligera,24,25a veces asociada con otras características de las células B
maduras (como la falta de expresión de CD34 y Tdt). 24Tales casos deben diferenciarse
cuidadosamente de las neoplasias de células B maduras. La mayoría de los casos de B-ALL
también muestran expresión de uno o varios antígenos asociados con mieloides, con mayor
frecuencia CD13 y CD33 y, con menos frecuencia, CD11b, CD15 y CD66c. El patrón de anomalías
inmunofenotípicas se correlaciona hasta cierto punto con la lesión genética subyacente en B-ALL
(véase la sección posterior).

La LLA-T se caracteriza por la expresión de antígenos asociados al linaje T (CD2, CD3, CD4, CD5,
CD7, CD8), así como CD1a, CD10, CD34, CD99, HLA-DR y Tdt. El patrón de expresión del
antígeno puede usarse para subclasificar las LLA-T de acuerdo con las etapas del desarrollo
normal de los timocitos a las que se asemejan. 1 , 26 ( Tabla 1 ) Un subconjunto de T-ALL, típico
del subtipo cortical, puede mostrar coexpresión de CD79a débil. 27 En particular, algunos T-
ALL/LBL pueden ser negativos para Tdt, 28lo que puede plantear el diagnóstico diferencial con
linfomas de células T maduras, particularmente difícil en casos que además son negativos para
HLA-DR y CD34. La T-ALL puede expresar con frecuencia antígenos mieloides, incluidos CD11b,
CD13, CD15 y CD33. Casos raros expresan CD117 (c-kit). Esta última característica se observa con
mayor frecuencia en asociación con un subconjunto de T-ALL recientemente descrito que parece
parecerse a los primeros precursores de células T que migraron recientemente de la médula ósea
al timo . 29 Estos casos genéticamente heterogéneos comparten un inmunofenotipo CD1a-, CD8-,
CD5 débilmente+, CD117+, CD34+, HLA-DR+, CD13+, CD33+, CD11b+ o CD65+ y parecen estar
invariablemente correlacionados con una mala respuesta a la terapia, incluida la inducción de la
remisión .fracaso y/o recidiva hematológica. Por lo tanto, se justifica un enfoque terapéutico
agresivo, incluido el trasplante de células madre hematopoyéticas , en estos pacientes.

Tabla 1 . Subclasificación de T-ALL según las etapas de maduración normal de los timocitos

Subtipo T-ALL CD1a CD2 cCD3 sCD3 CD4 CD5 CD7 CD8 CD34

Pro-T − − + − − − + − +

Pre-T − + + − − ± − − ±

T cortical + + + − + ± + + −

https://www.sciencedirect.com/science/article/pii/S0889858809000859 6/33
7/4/22, 05:44 Leucemia linfoblástica aguda - ScienceDirect

Subtipo T-ALL CD1a CD2 cCD3 sCD3 CD4 CD5 CD7 CD8 CD34

T medular − + + + ± un ± + ± −

a
La etapa medular T-ALL muestra expresión de CD4 o CD8 .

Genética de la LEUCEMIA LINFOBLÁSTICA AGUDA


Aunque relativamente homogéneos a nivel morfológico e inmunofenotípico, los LBL/LLA
muestran una heterogeneidad significativa a nivel genético. Sus lesiones genéticas definen
subconjuntos de enfermedades con biología y respuesta a la terapia distintas y se utilizan en los
esquemas de estratificación de riesgo para la mayoría de los protocolos de tratamiento actuales.

Genética de la leucemia linfoblástica aguda de células B precursoras

Subgrupos genéticos
Las B-ALL incluyen varios subgrupos citogenéticos con distintas características biológicas y
farmacológicas 1 que son muy importantes en la estratificación de riesgo actual de estos
pacientes. Estos subgrupos representan entre el 60 % y el 80 % de los casos en niños y adultos y
pueden identificarse mediante citogenética convencional, diagnóstico molecular (reacción en
cadena de la polimerasa con transcriptasa inversa), citometría de flujo (análisis del ciclo celular/
índice de ADN) 9 , 30 , 31 y, actualmente en ensayos preclínicos, perfiles de expresión génica
utilizando matrices de oligonucleótidos . Los restantes TODOSlos casos todavía se caracterizan
solo sobre la base de las características morfológicas e inmunofenotípicas. Los estudios de
perfiles de expresión génica han demostrado que estos subgrupos citogenéticos, aunque se
superponen ampliamente en morfología e inmunofenotipo, tienen firmas de expresión génica
distintas. 32 , 33 y sensibilidades in vitro a fármacos 34 , 35 , 36 que se correlacionan con
diferentes patrones de expresión génica de enzimas metabolizadoras de fármacos. 37 Análisis
genéticos de todo el genoma, utilizando matrices de polimorfismos de un solo nucleótido y ADN
genómicosecuenciación, han encontrado que estos subgrupos citogenéticos también contienen
alteraciones distintas en los genes que codifican los principales reguladores del desarrollo y la
diferenciación de los linfocitos B. 38 Estas metodologías de alto rendimiento han proporcionado
un punto de partida para una caracterización molecular mucho más detallada de estos subgrupos
genéticos. 39 , 40 , 41 , 42 Las características clinicopatológicas y moleculares de los principales
subtipos genéticos de B-ALL se resumen en la Tabla 2 .

Tabla 2 . Subtipos citogenéticos de LLA-B precursora y sus características clinicopatológicas

https://www.sciencedirect.com/science/article/pii/S0889858809000859 7/33
7/4/22, 05:44 Leucemia linfoblástica aguda - ScienceDirect

Subgrupo Frecuencia Anomalía Gen de Características Características Anomalías Car


citogenético (%) citogenética fusión morfológicas inmunofenotípicas moleculares farm
únicas únicas adicionales

LLA 27–29 51–65 N/A N/A N/A Poco May


hiperdiploide (PAGS) cromosomas frecuentes sens
(+4, +14, +21, mutaciones MT
6–8 (A)
+X) de BCDG
(13 %),
mutaciones
de FLT3 (21
%–25 %)

TODOS con 22–25 t(12;21) TEL/AML1 N / A Pre-B temprano Deleciones May


t(12;21) (PAGS) (p13;q22) ( ETVX / temprano, My+ monoalélicas sens
RUNX1 ) de PAX5 (28 la as
1–2 (A)
%)

TODOS con 3–6 (PAGS) t(1;19) E2A ( N/A Pre–B-ALL, N/A N/A
t(1;19) (q23;p13) TCF3 )/ CD34-/dim+,
5–7 (A)
PBX1 CD20-, CD9++

Filadelfia+ 2–3 (PAGS) t(9;22) BCR/ABL ( TODOS L1, a N/A Deleciones N/A
TODO (q34;q11.2) P190, veces IKZF1
20–30 (A)
P210 ) explosiones (Ikaros),
granulares mutaciones
BCDG en
66%

TODOS con 2–3 (PAGS) t(4;11) AF4 / N/A Pre-B temprano, Mutaciones May
t(v;11q23); (q21;q23) MLL CD10-, CD15+, FLT3 (18%) sens
5–7 (A)
MLL sCD22-, CD65+, Aumento la ci
t(19;11) ENL /
reorganizado NG2+ de la
(p13;q23) MLL
expresión
de genes
HOX

https://www.sciencedirect.com/science/article/pii/S0889858809000859 8/33
7/4/22, 05:44 Leucemia linfoblástica aguda - ScienceDirect

Subgrupo Frecuencia Anomalía Gen de Características Características Anomalías Car


citogenético (%) citogenética fusión morfológicas inmunofenotípicas moleculares farm
únicas únicas adicionales

LLA 5%– <46 N/A N/A N/A Mutaciones N / A


hipodiploide 6% (P) cromosomas de BCDG
(típicamente en el 100%
NA (A)
casi haploide
o bajo
hipodiploide)

TODOS con <1 t(5;14 IL3 / IGH Aumento de N/A N/A N/A
eosinofilia q31;q32) eosinófilos
displásicos

Abbreviations: A, % in adults; BCDG, B-cell development genes (eg, PAX5, EBF1, IKZF1, LEF1, TCF3, BLNK);
FLT3, fms-related tyrosine kinase 3; MP, mercaptopurines; MTX, methotrexate; My, myeloid antigens; NA, not
applicable, not known; P, % in pediatric patients.

Genetics of T-cell Acute Lymphoblastic Leukemia


Aunque extensos estudios moleculares combinados con perfiles de expresión génica han
descubierto una gran cantidad de información sobre la biología molecular de T-ALL, ninguno de
estos datos se usa actualmente en las decisiones de terapia en esta enfermedad, probablemente
debido a la menor cantidad de casos disponibles para evaluación en esta forma menos común de
ALL. Sin embargo, dado que algunas de estas alteraciones genéticas parecen correlacionarse con
el resultado, es probable que al menos algunas de ellas entren en el ámbito clínico en un futuro
próximo.

Las anomalías cromosómicas recurrentes en la LLA-T a menudo incluyen translocaciones


recíprocas que interrumpen genes de factores de transcripción importantes para el desarrollo,
como resultado de reordenamientos en loci para los genes del receptor de células T ( TCR ), más
comúnmente TCRα (14q11.2 ) y TCRβ (7q35) . Los ejemplos comunes incluyen el t(1;14) (p32;q11)
(3 % de los casos), que involucra al gen TAL/SCL1 (1p32), el t(10;14) (q24;q11.2) que interrumpe
latranscripción HOX11 gen del factor (10q24), 43 , 44 la translocación t(5;14) (q35;q32), con
reordenamientos delgen HOX11L2 45 , 46y t(11;14) y t(7;11) que involucran a los oncogenes LMO1
(11p15) y LMO2 (11p13), respectivamente. 47 Las translocaciones menos comunes que no
involucran los genes TCR incluyen la t(9;17) y la t(10;11) (p12;q14), que conducen al gen de fusión
AF10-CALM . 48 , 49 Este último se asocia con un fenotipo TCR γδ maduro o inmaduro y, en
estudios limitados, se asocia con un pronóstico precario para el subgrupo de pacientes con
leucemia TCR γδ inmadura. 50 , 51
https://www.sciencedirect.com/science/article/pii/S0889858809000859 9/33
7/4/22, 05:44 Leucemia linfoblástica aguda - ScienceDirect

T-ALL también puede albergar lesiones moleculares presentes sin anomalías citogenéticas
detectables. Hasta el 50% de T-ALL contiene mutaciones activadoras de TAL1/SCL ,
independientemente de las translocaciones detectables. Estas mutaciones se correlacionan con
leucemias detenidas en la etapa cortical tardía de la maduración de los timocitos , que expresan
TCRαβ y parecen tener un pronóstico inferior. 52 , 53 Aproximadamente el 30% de T-ALL
contienen HOX11 (TLX1) , más común en adultos que en niños. Estos casos corresponden a un
fenotipo de timocitos corticales tempranos que expresan TCRαβ y parecen tener una
supervivencia superior. 52 , 53 , 54 LJ1las mutaciones están presentes hasta en el 22% de la LLA-T
pediátrica, y se correlacionan con el estadio de diferenciación de timocitos tempranos doble
negativo 52 y con una supervivencia inferior. Las mutaciones del gen MLL están presentes en el
4% al 8% de las LLA-T, se asocian con la detención de la maduración en las primeras etapas de los
timocitos y la expresión de TCRγδ, y no tienen impacto en el pronóstico. 53 Las mutaciones
activadoras del gen NOTCH1 están presentes en más del 50 % de las LLA-T. 55

Estudios limitados han sugerido una correlación entre estas anomalías genéticas presentes en los
blastos T-ALL y la edad del paciente, posiblemente correspondientes a diferentes etapas de
desarrollo e involución tímica. 56

Reordenamientos del gen del receptor de células T e inmunoglobulina en la leucemia


linfoblástica aguda
Los reordenamientos de los genes Ig y/o TCR están presentes en la mayoría de ALL/LBL. Estos
reordenamientos no siempre son específicos del linaje, ya que B-ALL puede contener
reordenamientos de TCR , 57 , 58 y T-ALL puede contener reordenamientos de Ig , 59
posiblemente debido a la actividad recombinasa continua en las células hematopoyéticas
malignas . 60

Reordenamientos de Ig y TCR en la leucemia linfoblástica aguda de células B


La mayoría de las B-ALL (97 %) contienen reordenamientos del gen IG , que involucran el gen de
la cadena pesada ( IgH ) (>95 %), el gen de la cadena ligera kappa ( IgK) (30 %) o el gen de la cadena
ligera lambda ( IgL ) (20 %). ). 59 Estos reordenamientos a menudo son oligoclonales (múltiples
reordenamientos de IgH presentes en 30% a 40%, múltiples reordenamientos de IgK en 5% a
10%). 61 B-ALL también puede contener reordenamientos y/o deleciones de TCR, involucrando
los genes TCR β , γ y/o δ (en 35%, 55% y 90% de los casos, respectivamente). 58 Estos pueden ser
biclonales u oligoclonales ( TCRβ en 3%, TCRγ en 10%). 59 La oligoclonalidad observada en estos
neoplasmas puede deberse a reordenamientos continuos 60 y reordenamientos secundarios, que
pueden explicar los cambios en los patrones de reordenamientos de genes de Ig y TCR que a
menudo se observan en B-ALL en la recaída. 58

Reordenamientos de Ig y TCR en la leucemia linfoblástica aguda de células T

https://www.sciencedirect.com/science/article/pii/S0889858809000859 10/33
7/4/22, 05:44 Leucemia linfoblástica aguda - ScienceDirect

T-ALL/LBL típicamente contienen reordenamientos del gen TCR (95%–100%). Se observa una
frecuencia más baja en pro-T ALL raras, donde los genes TCR están en configuración de línea
germinal en aproximadamente el 10% de los casos. 59 Las T-ALL TCRαβ+ contienen
reordenamientos de genes TCRβ (100%) y TCRγ (100%) y tienen al menos un alelo TCRδ
delecionado , mientras que la deleción del segundo alelo se observa en el 65% de los casos. Las T-
ALL TCRγδ+ tienen reordenamientos TCRδ y TCRγ (100 % de los casos para cada uno) y
normalmente también contienen reordenamientos TCRβ (95 %). A diferencia de B-ALL, la Ig
inapropiada para el linajelos reordenamientos de genes se encuentran solo en ~20% de T-ALL,
típicamente como reordenamientos incompletos de IgH ( DH - JH ) . Además, rara vez se observa
oligoclonalidad en muestras de diagnóstico de T-ALL. 62 La LLA-T recidivante puede mostrar
reordenamientos secundarios de TCRγ y TCRβ en 15% a 20% de los casos. 62

Diagnóstico diferencial
ALL/LBL puede superponerse morfológica e inmunofenotípicamente con una variedad de
entidades benignas y malignas.

Proliferaciones benignas
Las expansiones de precursores de células B benignas ("hematogones") y los timocitos normales
pueden entrar en el diagnóstico diferencial de B-ALL/LBL y T-ALL/LBL, respectivamente, debido
a su gran parecido morfológico e inmunofenotípico con sus contrapartes neoplásicas.

Los linfocitos B precursores benignos (hematógonos) son linfocitos B precursores normales que se
encuentran principalmente en la médula ósea, pero también en pequeñas cantidades en sitios
extramedulares, como la sangre periférica, los ganglios linfáticos y las amígdalas . 63 , 64 , 65 , 66
Pueden aumentar en número (a veces hasta una proporción significativa de las células normales)
en la médula ósea de pacientes de cualquier edad, pero con mayor frecuencia en niños y adultos
jóvenes, 67 en una variedad de condiciones reactivas o regenerativas. Estos incluyen infecciones
virales y recuperación de la médula después de una infección, quimioterapia y trasplante de
médula ósea . 67 Morfológicamente, los hematógonos incluyen células que se asemejan
aLinfoblastos tipo L1 y linfocitos pequeños inmaduros y maduros. También existe un rango
inmunofenotípico de maduración, que se puede resumir en varias etapas distintas. 68,69Por lo
general, los hematogones en etapa I (células CD34+, Tdt+) representan un componente muy
pequeño de las expansiones de hematogones reactivos. Sin embargo, en ciertos entornos,
especialmente en la recuperación después de la quimioterapia, este subconjunto puede volverse
más prominente (o incluso predominante), lo que aumenta el diagnóstico diferencial con la
leucemia recurrente. 70 El análisis de citometría de flujo a menudo es esencial para esta
distinción, ya que en la mayoría de los casos B-ALL mostrará aberraciones inmunofenotípicas
que no se esperan en las expansiones de hematógonos benignos. 68

https://www.sciencedirect.com/science/article/pii/S0889858809000859 11/33
7/4/22, 05:44 Leucemia linfoblástica aguda - ScienceDirect

Los timocitos corticales normales son importantes en el diagnóstico diferencial de T-ALL/LBL


precursores, en muestras obtenidas de lesiones mediastínicas, que pueden contener corteza
tímica normal o hiperplásica , además de tejido neoplásico, y de lesiones cervicales, que pueden
contener tejido tímico ectópico. o timoma ectópico. La distinción morfológica suele ser clara, y se
basa principalmente en la presencia de la arquitectura tímica característica y las características de
los timocitos de apariencia más madura. Sin embargo, puede ocurrir una superposición
significativa a nivel inmunofenotípico. Por lo tanto, los hallazgos de células T precursoras Tdt+,
CD34+ y CD1a+ en la citometría de flujo de masas mediastínicas o cervicales (especialmente en
niños y adultos jóvenes) siempre deben correlacionarse con los hallazgos morfológicos. De
manera similar a los hematogones, cuando se examinan inmunofenotípicamente, los timocitos
normales muestran una secuencia característica de maduración, mientras que los T-ALL/LBL
muestran aberraciones inmunofenotípicas en todos los casos, lo que debería ser útil en el
diagnóstico diferencial. 68

Neoplasmas malignos
Una variedad de neoplasias malignas con características morfológicas e inmunofenotípicas
superpuestas pueden incluirse en el diagnóstico diferencial de ALL/LBL.

Otras neoplasias hematopoyéticas blásticas pueden incluir AML , leucemias de linaje mixto,
leucemias de células dendríticas plasmocitoides (DC2) y, especialmente en adultos, linfoma de
células del manto blástico . La LMA puede presentarse como un infiltrado tisular (denominado
sarcoma mieloide o monoblástico) con o sin un proceso leucémico asociado y, a menudo, se
considera en el diagnóstico diferencial de LLA/LBL, especialmente en casos con blastos más
grandes y nucleolos prominentes . En algunos casos, el sarcoma mieloide puede estar asociado
con elementos eosinofílicos inmaduros . Inmunofenotipificación extensivapor citometría de flujo
normalmente permite una fácil distinción. Sin embargo, en muestras de tejido incluidas en
parafina, solo se puede aplicar un panel más limitado de marcadores inmunohistoquímicos, lo
que dificulta este diagnóstico diferencial. Normalmente es suficiente un panel de marcadores que
incluya los de MPO , lisozima , Tdt , PAX5 y CD3. En particular, AML con t (8; 21) puede ser un
desafío ocasional en este contexto, ya que puede expresar CD19, CD79a y PAX5, además de MPO.
71
La expresión Tdt también se puede encontrar en otros tipos de AML. 72 Leucemias agudas de
linaje mixto(en particular, las leucemias bifenotípicas agudas) pueden considerarse en el
diagnóstico diferencial de la LLA en los casos que expresan varios antígenos mieloides. En tales
casos, se deben aplicar los sistemas de puntuación ideados para este propósito. La leucemia DC2 es
un tipo raro de leucemia (<1 % de todas las leucemias) 73 que se cree deriva de un subtipo de
células presentadoras de antígenos, las células plasmocitoides DC2. Desde el punto de vista
morfológico, pueden parecerse a ALL L2 o pueden mostrar un amplio citoplasma, con
pseudópodos y vacuolas citoplásmicas alineadas a lo largo de los contornos de las células como
un “collar de perlas”. Las células leucémicas expresan CD4, CD56, HLA-DR, así como los
antígenos específicos de DC2 CD123, BDCA-2 y BDCA-4. Algunos casos pueden expresar

https://www.sciencedirect.com/science/article/pii/S0889858809000859 12/33
7/4/22, 05:44 Leucemia linfoblástica aguda - ScienceDirect

antígenos linfoides y mieloides de baja especificidad (como CD2, CD22, Tdt y CD33), pero suelen
ser negativos para otros antígenos mieloides, CD3, TCR , CD79a y CD34. 73 , 74 En la actualidad,
estas leucemias se tratan de manera similar a la LLA. El linfoma de células del manto blástico rara vez
se incluye en el diagnóstico diferencial de ALL/LBL. El inmunofenotipo característico de este
linfoma, CD5+, CD10-, CD20+, Tdt- y ciclina D1+, debería permitir una fácil diferenciación. 75

Linfoma/leucemia de Burkitt
Por lo general, la distinción se basa en la apariencia morfológica distintiva del linfoma de Burkitt
, combinado con un inmunofenotipo de células B maduras, CD10+, CD20+, Ig de superficie
positiva con restricción de cadena ligera, CD34-, Tdt- y negativo para antígenos mieloides. Esta
distinción puede ser difícil en el contexto del llamado “linfoma de Burkitt atípico o precursor B”,
que contiene las translocaciones cromosómicas clásicas asociadas con Burkitt pero tiene
características inmunofenotípicas más cercanas a las neoplasias de células B precursoras. 76 , 77El
inmunofenotipo incluye características de células B precursoras (p. ej., expresión de Tdt y CD34,
dim CD20) y de células B maduras (IgM de superficie con restricción de cadena ligera). En el
contexto de tales combinaciones de características, la LLA debe diagnosticarse con precaución y
los hallazgos deben correlacionarse con la citogenética, porque en presencia de t(8;14) o sus
variantes, los pacientes se tratan de acuerdo con los protocolos de linfoma de Burkitt. 77

Linfoma periférico de células T


A variety of mature (or peripheral) T-cell lymphomas may present with leukemic involvement of
the bone marrow and /or peripheral blood, which may morphologically mimic T-ALL. These
include T-cell prolymphocytic leukemia, adult T-cell leukemia/lymphoma, Sézary syndrome, and
rarely anaplastic large-cell lymphoma (ALCL).78 The differential diagnosis depends on a
combination of morphologic features, clinical presentation, and the finding of a mature T-cell
immunophenotype in the latter lymphomas. Certain immunophenotypic features, such as
expression of CD30 and the ALK protein, can also be used to exclude T-ALL. In this context, one
should also remember that some mature T-cell lymphomas, especially ALCL, may express
myeloid antigens, such as CD33, a feature typically associated with T-ALL.79 Last, the presence of
genetic lesions such as the t(2;5) for ALCL would further exclude ALL.

Non-hematopoietic neoplasms
A variety of blastic small blue-cell tumors of childhood may enter the differential diagnosis of
ALL/LBL in the pediatric age group. Ewing sarcoma is the childhood nonhematopoietic
neoplasm most likely to be considered in the differential diagnosis of LBL, especially in small
samples. It is typically composed of small blastic cells without prominent nucleoli, which are
CD45- and CD99+, similar to a subset of precursor B-cell ALL.80 On further evaluation, Ewing
sarcoma is always negative for B-lineage markers and Tdt, which should be included in
diagnostic panels for small blue-cell tumors of children. Merkel cell carcinoma, most often seen in

https://www.sciencedirect.com/science/article/pii/S0889858809000859 13/33
7/4/22, 05:44 Leucemia linfoblástica aguda - ScienceDirect

adults, may show immunophenotypic overlap with B-ALL/LBL, as both PAX-5 and Tdt expression
have been reported in this tumor.81, 82 Notably, PAX-5 expression, although largely specific for
B-lineage in hematopoietic neoplasms, may be seen in a variety of other nonhematopoietic
tumors, such as neuroendocrine carcinomas, and a variety of other subtypes of carcinomas.82, 83,
84, 85 In all of these entities, correlation with clinical presentation and morphology and applying
the appropriate panels of immunohistochemical stains allow for accurate distinction from
ALL/LBL.

Prognosis and Treatment

Prognosis
The prognosis of ALL has improved dramatically over the past several decades as a result of
adapting therapy to the level of risk for relapse, improvements in supportive care, and
optimization of the existing chemotherapy drugs. The outcome of pediatric ALL has evolved from
an overall survival of less than 10% in the 1960s to approximately 75% to 80% at present.9
However, adult patients have a less optimistic outlook. The remission rates have reached 85% to
90%, with overall survival rates of only 40% to 50%.86 About 75% of the patients present with poor
risk features and have a disease-free survival of 25%, and only 25% present with standard risk
features that confer disease-free survival greater than 50%.30 ALL patients are stratified and
treated according to algorithms that integrate the presenting features (patient age, leukocyte
counts, the presence or absence of central nervous system [CNS], or testicular involvement),
leukemia features (lineage, genetic subgroup), and of early therapy response (measuring the
dynamic of disease clearance in the first 1–2 weeks of therapy).9, 30 Patients at low risk for relapse
are treated primarily with antimetabolite therapy. Pediatric patients presenting with high-risk
features or showing induction failure or persistent minimal residual disease (MRD) after the first
2 weeks of induction receive more aggressive therapy and are considered for allogeneic
hematopoietic stem-cell transplantation. All the remaining cases are classified as standard risk
for relapse and are treated with intensive multiagent chemotherapy regimens.

Therapy of acute lymphoblastic leukemia


In most centers, the treatment of ALL involves short-term intensive chemotherapy (with high-
dose methotrexate, cytarabine, cyclophosphamide, dexamethasone or prednisone, vincristine, L-
asparaginase, and/or an anthracyclin).9, 30 This is followed by intensification or consolidation
therapy to eliminate residual leukemia, prevent or eradicate CNS leukemia, and ensure
continuation of remission. Radiation may be used for patients showing evidence of CNS or
testicular leukemia, although this approach is controversial at the current time, especially in
children.9 In adult patients, the use of growth factors such as granulocyte colony-stimulating
factor that accelerate hematopoietic recovery has greatly improved the success rate of ALL
therapy.87

https://www.sciencedirect.com/science/article/pii/S0889858809000859 14/33
7/4/22, 05:44 Leucemia linfoblástica aguda - ScienceDirect

Pharmacogenetics of acute lymphoblastic leukemia


Increasing numbers of pharmacogenetic studies have shown that germline polymorphisms and
mutations present in ALL patients may affect the levels of expression and functionality of drug-
metabolizing genes. They may lead to an increase in the likelihood of leukemia in their carriers,
may influence the response of leukemic blasts to specific chemotherapy agents, and may also
increase the probability of developing secondary (treatment-related) malignancies.88, 89 Such
genes are those encoding for thiopurine methyltransferase, glutathione S-transferase, cytochrome
P450 3A4, and methylene-tetrahydrofolate reductase. Furthermore, leukemic blasts with various
chromosomal abnormalities (such as hyperdiploid ALL) and therefore additional copies of the
wild-type genes for drug-metabolizing enzymes may differ from the somatic cells with respect to
their production of these enzymes, leading to altered resistance to the related drugs.90 Thus,
chemotherapy regimens need to be tailored not only to the specific features of each acute
leukemia but also to the patients' individual genetic background.

Minimal Residual Disease Studies in Acute Lymphoblastic Leukemia


The utility of MRD studies has been well documented in the management of children with ALL91
and in further stratification of adults with standard-risk ALL.92 They include several types of
techniques that can detect amounts of residual leukemia that cannot be identified reliably using
morphologic examination or conventional flow cytometry (“submicroscopic disease”). The
technical approaches currently used for the detection and quantification of MRD include flow
cytometry and molecular analysis (polymerase-chain reaction [PCR]) for leukemia-specific Ig and
TCR gene rearrangements or fusion transcripts. Although generally there is good correlation
between these methodologies in most cases, the use of both techniques is the ideal approach for
adequate MRD monitoring in all patients.

Flow cytometric detection of MRD (FC-MRD) is based on the presence of an aberrant leukemia-
associated immunophenotype, distinct from that of normal (benign) lymphoid precursors
present in the bone marrow and peripheral blood. For T-ALL, the detection of cells coexpressing
T-lineage antigens and Tdt or CD34 in peripheral blood or marrow is sufficient to support the
presence of MRD, since normal T-cell precursors are not normally encountered outside the
thymus. For B-ALL, the distinction is much more difficult, because, as previously discussed,
normal precursor B cells (hematogones) can be encountered at all anatomic sites, and more
refined analyses are required to differentiate these cells from leukemic B lymphoblasts.
Additionally, benign progenitor cells occurring in the postchemotherapy or post-transplantation
settings may have immunophenotypic features distinct from those encountered in reactive
conditions. In addition, leukemic cells may show immunophenotypic shifts during therapy and
between initial diagnosis and relapse.93 These factors have to be considered when constructing
panels of markers adequate for MRD monitoring and analyzing the data in this setting. Aberrant
leukemia-associated immunophenotypes can be identified in 95% of pediatric ALL.94, 95
Markers typically included in FC-MRD panels include myeloid antigens commonly expressed in
https://www.sciencedirect.com/science/article/pii/S0889858809000859 15/33
7/4/22, 05:44 Leucemia linfoblástica aguda - ScienceDirect

B-cell leukemia (e.g. CD13, CD15, CD33, CD65, CD66c) as well as markers often expressed
inappropriately for stage of maturation in ALL (CD21, normally only coexpressed on mature B
cells; CD38, lower than normal cells; CD58, higher than normal cells). The sensitivity of FC-MRD
in routine clinical samples is typically 1 in 104 cells.96 Under ideal experimental conditions and
when leukemic cells have a very distinct immunophenotype and at least 1 x 107 cells are available,
the sensitivity may be as high as 1 in 105 cells.96 Advantages of FC-MRD detection (as opposed to
PCR) include the possibility of direct quantitation and that of excluding dying cells and cellular
debris from analysis. Disadvantages include a lower sensitivity and the difficulties resulting from
immunophenotypic shifts in the leukemic cells, with possible disappearance of some of the
aberrancies used to identify MRD in a specific case.96 FC-MRD detection can be applied in a
different manner to peripheral blood and bone marrow samples, depending on the lineage of
ALL.97 In T-ALL, monitoring MRD levels using peripheral blood is an acceptable substitute for
bone marrow samples, as the MRD levels at the two locations always match. For B-ALL, bone
marrow residual disease may be found without peripheral blood involvement, and often the
levels of MRD found in the bone marrow significantly exceed those found in the blood. The
prognostic value of FC-MRD has been demonstrated in retrospective and prospective studies.95
At the current time, it is hoped that high-throughput methodologies for genetic analysis in ALL
will lead to the discovery of new leukemia-associated markers that can be used in FC-MRD
studies.95

PCR for Ig or TCR gene rearrangements. If using PCR with consensus primers (including
heteroduplex and GeneScan strategies), the detection limit of MRD is 1% to 5% depending on the
number of polyclonal B- or T-lymphocytes present in the sample.59 Since this sensitivity is
comparable to the use of morphologic and immunophenotypic evaluation, alternative approaches
have been employed. The sensitivity attained by using patient-specific, junctional region–specific
oligonucleotide probes is more suitable for MRD detection. These probes are generated through
sequencing of the junctional regions of the rearranged Ig and/or TCR genes found in the
leukemic cells of each patient at the time of initial diagnosis.98, 99 They are then used in real-
time quantitative PCR (RQ-PCR) assays in follow-up samples. Through this approach, the
sensitivity of MRD detection has increased to 10−4 to 10−6 (ie, 1–100 leukemic cells among 106
normal cells).

PCR for leukemia-specific fusion transcripts. This methodology detects and quantifies fusion
transcripts that correspond to the leukemia-associated translocations such as t(9;22), t(4;11),
t(1;19), or t(12;21) (See Table 2), typically via reverse transcriptase RQ-PCR assays.91 It can only be
applied to less than 50% of ALLs, which contain such fusion genes. The sensitivity of this
methodology is as high as 10−4, depending of the amount and quality of the RNA available in the
sample.91, 100 Due to its high sensitivity and lack of patient specificity, a major pitfall is related
to false-positive results due to cross-contamination.91 Although this modality appears to correlate
well with the other two methodologies in most patients,100 its use in the prediction of outcome in
ALL remains to be established.59

https://www.sciencedirect.com/science/article/pii/S0889858809000859 16/33
7/4/22, 05:44 Leucemia linfoblástica aguda - ScienceDirect

Relapsed Acute Lymphoblastic Leukemia


The overall frequency of relapse in ALL is approximately 25% in children and 50% in adults, with
a rate that is highly dependent on the immunophenotypic and genetic subtype or otherwise-
defined risk category of ALL.9, 30, 87 Recent insights have identified additional genetic predictors
of relapse, such as deletions of the IKZF1 (Ikaros) gene.101 The genetic subgroup also determines
the characteristics of the relapse and the prognosis of these patients. For instance, relapse in
patients with Philadelphia-positive ALL, representing approximately 10% of the relapsed ALL
(rALL) in some studies, typically occurs following a short complete remission (CR) and correlates
with an extremely poor prognosis, as a second CR cannot be induced in many of these patients.
ALLs with TEL/AML1 most often relapse following a long first CR, and a second CR is relatively
easy to induce and maintain, often for long periods of time. Most ALLs relapse in the first 3 to 5
years from diagnosis. Only a very small percentage relapse more than 5 years from diagnosis, and
relapses may occur 10 to 20 years later in a minority of patients.102 rALL may involve the bone
marrow or extramedullary tissues (most often at “sanctuary sites,” such as CNS, testis, ovary) or
both. The isolated bone marrow relapses appear to correlate with a less favorable prognosis than
the isolated extramedullary or combined relapses.9

The morphologic and immunophenotypic features of rALL are often largely similar to those seen
at the time of initial diagnosis. However, variable immunophenotypic shifts may also be
observed, whereby some antigens (most often Tdt, CD10, HLA-DR, myeloid antigens) may
increase or decrease in intensity or even be lost at relapse.93, 103 Such variations may be found in
34% to 73% of pediatric B-ALL and 15% of pediatric T-ALL.93, 103 The most extreme variations
consist of lineage switch, from B-ALL to T-ALL, or from ALL to AML or biphenotypic leukemia.
Conventional cytogenetics and molecular analysis typically identify the translocations and fusion
transcripts present at the time of initial diagnosis. In addition, cytogenetic analysis often (75%)
identifies newly acquired abnormalities.22 Very rarely, an entirely different karyotype may be
identified, suggesting the possibility of a second de novo ALL. Molecular studies document shifts
in the pattern of Ig and TCR gene rearrangements and even acquisition of new fusion genes.104 In
rare cases (0.5%–1.5%), detailed molecular studies may support a new (second) ALL, distinct from
the previous disease.104 It appears that a combination of immunophenotypic and molecular
studies may distinguish three categories of ALL relapse: rALL similar to the leukemia present at
initial diagnosis, rALL clonally derived from the initial leukemia, and, rarely, a second de novo
ALL.104 Recent high-throughput genomic studies using SNP arrays and comparing diagnostic
and relapse ALL samples correlate with these findings.105 In these latter studies, a minority (8%)
of rALL were similar to the initial diagnostic samples, a third (34%) were consistent with clonal
evolution of the cells predominant at diagnosis, and half (52%) were shown to have derived by
clonal evolution from minor clones present at the time of diagnosis that appeared to precede the
dominant clones in the sequence of genetic lesions (“ancestral clones”). About 6% of rALL were
completely genetically distinct from the diagnostic samples, suggesting de novo disease.

https://www.sciencedirect.com/science/article/pii/S0889858809000859 17/33
7/4/22, 05:44 Leucemia linfoblástica aguda - ScienceDirect

Summary
Although relatively homogeneous at the morphologic and immunophenotypic level, ALL/LBLs
encompass a family of extremely heterogeneous disorders when examined at the genetic level.
This heterogeneity is reflected in the outcome of pediatric and adult patients in the context of
contemporary therapies. High-throughput analysis methodologies have begun to characterize
this heterogeneity and, although only in their early stages, have begun to uncover new clinically
significant disease subsets, previously unidentified markers useful for MRD monitoring,
mechanisms and predictors of disease relapse, germline polymorphisms important in
individualized therapy, and new attractive therapeutic targets. These insights are likely to further
improve the treatment outcome of patients with ALL.

Special issue articles Recommended articles

References
1 S.H. Swerdlow, E. Campo, N.L. Harris, et al. (Eds.), WHO classification of tumours of
haematopoietic and lymphoid tissues, IARCPress, Lyon, France (2008), pp. 157-178
Google Scholar

2 National Cancer Institute. SEER Cancer Statistics Review, 1975–2006. Available at:
http://seer.cancer.gov/csr/1975_2006/. Accessed January 23, 2009.
Google Scholar

3 J.G. Gurney, R.K. Severson, S. Davis, et al.


Incidence of cancer in children in the United States. Sex-, race-, and 1-year age-specific
rates by histologic type
Cancer, 75 (8) (1995), pp. 2186-2195
View Record in Scopus Google Scholar

4 American Cancer Society


Cancer facts and figures 2008
Available at:
http://www.cancer.org
Accessed January 23, 2009
Google Scholar

5 R. Shivakumar, W. Tan, G.E. Wilding, et al.


Biologic features and treatment outcome of secondary acute lymphoblastic leukemia–a
review of 101 cases
Ann Oncol, 19 (9) (2008), pp. 1634-1638
Article Download PDF CrossRef View Record in Scopus Google Scholar

https://www.sciencedirect.com/science/article/pii/S0889858809000859 18/33
7/4/22, 05:44 Leucemia linfoblástica aguda - ScienceDirect

6 L.G. Spector, J.A. Ross, L.L. Robison, et al.


Epidemiology and etiology
C.H. Pui (Ed.), Childhood leukemias, Cambridge University Press, New York (2006), pp. 48-
66
CrossRef View Record in Scopus Google Scholar

7 K.B. Gale, A.M. Ford, R. Repp, et al.


Backtracking leukemia to birth: identification of clonotypic gene fusion sequences in
neonatal blood spots
Proc Natl Acad Sci U S A, 94 (25) (1997), pp. 13950-13954
View Record in Scopus Google Scholar

8 J.W. Taub, M.A. Konrad, Y. Ge, et al.


High frequency of leukemic clones in newborn screening blood samples of children with
B-precursor acute lymphoblastic leukemia
Blood, 99 (8) (2002), pp. 2992-2996
Article Download PDF View Record in Scopus Google Scholar

9 C.H. Pui
Acute lymphoblastic leukemia
C.H. Pui (Ed.), Childhood leukemias, Cambridge University Press, New York (2006), pp.
439-472
CrossRef View Record in Scopus Google Scholar

10 S.B. Kahwash, S.J. Qualman


Cutaneous lymphoblastic lymphoma in children: report of six cases with precursor B-cell
lineage
Pediatr Dev Pathol, 5 (1) (2002), pp. 45-53
View Record in Scopus Google Scholar

11 J.M. Bennett, D. Catovsky, M.T. Daniel, et al.


Proposals for the classification of the acute leukaemias. French- American-British (FAB)
co-operative group
Br J Haematol, 33 (4) (1976), pp. 451-458
CrossRef View Record in Scopus Google Scholar

12 J.M. Bennett, D. Catovsky, M.T. Daniel, et al.


The morphological classification of acute lymphoblastic leukaemia: concordance among
observers and clinical correlations
Br J Haematol, 47 (4) (1981), pp. 553-561
CrossRef View Record in Scopus Google Scholar

13 R.B. Lorsbach, M. Onciu, F.G. Behm


https://www.sciencedirect.com/science/article/pii/S0889858809000859 19/33
7/4/22, 05:44 Leucemia linfoblástica aguda - ScienceDirect

Bone marrow reticulin fiber deposition in pediatric patients with acute lymphoblastic
leukemia
Mod Pathol, 15 (1) (2002), p. 1048
Google Scholar

14 J.S. Lilleyman, I.M. Hann, R.F. Stevens, et al.


Blast cell vacuoles in childhood lymphoblastic leukaemia
Br J Haematol, 70 (2) (1988), pp. 183-186
CrossRef View Record in Scopus Google Scholar

15 P.J. Darbyshire, J.S. Lilleyman


Granular acute lymphoblastic leukaemia of childhood: a morphological phenomenon
J Clin Pathol, 40 (3) (1987), pp. 251-253
CrossRef View Record in Scopus Google Scholar

16 P.G. Dyment, R.A. Savage, J.T. McMahon


Anomalous azurophilic granules in acute lymphoblastic leukemia
Am J Pediatr Hematol Oncol, 4 (2) (1982), pp. 207-211
View Record in Scopus Google Scholar

17 P. Stein, S. Peiper, D. Butler, et al.


Granular acute lymphoblastic leukemia
Am J Clin Pathol, 79 (4) (1983), pp. 426-430
CrossRef View Record in Scopus Google Scholar

18 S. Sharma, S. Narayan, M. Kaur


Acute lymphoblastic leukaemia with giant intracytoplasmic inclusions–a case report
Indian J Pathol Microbiol, 43 (4) (2000), pp. 485-487
View Record in Scopus Google Scholar

19 E.T. Yanagihara, F. Naeim, R.P. Gale, et al.


Acute lymphoblastic leukemia with giant intracytoplasmic inclusions
Am J Clin Pathol, 74 (3) (1980), pp. 345-349
CrossRef View Record in Scopus Google Scholar

20 H.R. Schumacher, J.E. Champion, et al.


Acute lymphoblastic leukemia–hand mirror variant. An analysis of a large group of
patients
Am J Hematol, 7 (1) (1979), pp. 11-17
CrossRef View Record in Scopus Google Scholar

21 H. Horigome, R. Sumazaki, N. Iwasaki, et al.

https://www.sciencedirect.com/science/article/pii/S0889858809000859 20/33
7/4/22, 05:44 Leucemia linfoblástica aguda - ScienceDirect

Fatal eosinophilic heart disease in a child with neurofibromatosis-1 complicated by acute


lymphoblastic leukemia
Heart Vessels, 20 (3) (2005), pp. 120-122
CrossRef View Record in Scopus Google Scholar

22 R.D. Brunning, R.W. McKenna


Acute leukemias. Tumors of the bone marrow
Armed Forces Institute of Pathology, Washington, DC (1994)
p. 100–36
Google Scholar

23 A. Cantu-Rajnoldi, R. Invernizzi, A. Biondi, et al.


Biological and clinical features of acute lymphoblastic leukaemia with cytoplasmic
granules or inclusions: description of eight cases
Br J Haematol, 73 (3) (1989), pp. 309-314
CrossRef View Record in Scopus Google Scholar

24 S. Li, G. Lew
Is B-lineage acute lymphoblastic leukemia with a mature phenotype and l1 morphology a
precursor B-lymphoblastic leukemia/lymphoma or Burkitt leukemia/lymphoma?
Arch Pathol Lab Med, 127 (10) (2003), pp. 1340-1344
CrossRef View Record in Scopus Google Scholar

25 B.P. Nelson, D. Treaba, C. Goolsby, et al.


Surface immunoglobulin positive lymphoblastic leukemia in adults; a genetic spectrum
Leuk Lymphoma, 47 (7) (2006), pp. 1352-1359
CrossRef View Record in Scopus Google Scholar

26 M.C. Bene, G. Castoldi, W. Knapp, et al.


Proposals for the immunological classification of acute leukemias. European Group for
the Immunological Characterization of Leukemias (EGIL)
Leukemia, 9 (10) (1995), pp. 1783-1786
Google Scholar

27 E. Pilozzi, K. Pulford, M. Jones, et al.


Co-expression of CD79a (JCB117) and CD3 by lymphoblastic lymphoma
J Pathol, 186 (2) (1998), pp. 140-143
View Record in Scopus Google Scholar

28 J. Faber, H. Kantarjian, M.W. Roberts, et al.


Terminal deoxynucleotidyl transferase-negative acute lymphoblastic leukemia
Arch Pathol Lab Med, 124 (1) (2000), pp. 92-97
CrossRef View Record in Scopus Google Scholar
https://www.sciencedirect.com/science/article/pii/S0889858809000859 21/33
7/4/22, 05:44 Leucemia linfoblástica aguda - ScienceDirect

E. Coustan-Smith, C.G. Mullighan, M. Onciu, et al.


29
Early T-cell precursor leukaemia: a subtype of very high-risk acute lymphoblastic
leukaemia
Lancet Oncol, 10 (2009), pp. 147-156
Article Download PDF View Record in Scopus Google Scholar

30 S. Faderl, S. Jeha, H.M. Kantarjian


The biology and therapy of adult acute lymphoblastic leukemia
Cancer, 98 (7) (2003), pp. 1337-1354
View Record in Scopus Google Scholar

31 M. Mancini, D. Scappaticci, G. Cimino, et al.


A comprehensive genetic classification of adult acute lymphoblastic leukemia (ALL):
analysis of the GIMEMA 0496 protocol
Blood, 105 (9) (2005), pp. 3434-3441
Article Download PDF CrossRef View Record in Scopus Google Scholar

32 M.E. Ross, X. Zhou, G. Song, et al.


Classification of pediatric acute lymphoblastic leukemia by gene expression profiling
Blood, 102 (8) (2003), pp. 2951-2959
Article Download PDF View Record in Scopus Google Scholar

33 E.J. Yeoh, M.E. Ross, S.A. Shurtleff, et al.


Classification, subtype discovery, and prediction of outcome in pediatric acute
lymphoblastic leukemia by gene expression profiling
Cancer Cell, 1 (2) (2002), pp. 133-143
Article Download PDF View Record in Scopus Google Scholar

34 C.H. Pui, D. Campana, W.E. Evans


Childhood acute lymphoblastic leukaemia–current status and future perspectives
Lancet Oncol, 2 (10) (2001), pp. 597-607
Article Download PDF View Record in Scopus Google Scholar

35 N.L. Ramakers-van Woerden, R. Pieters, A.H. Loonen, et al.


TEL/AML1 gene fusion is related to in vitro drug sensitivity for L-asparaginase in
childhood acute lymphoblastic leukemia
Blood, 96 (3) (2000), pp. 1094-1099
View Record in Scopus Google Scholar

36 R.W. Stam, M.L. den Boer, J.P. Meijerink, et al.


Differential mRNA expression of Ara-C-metabolizing enzymes explains Ara-C sensitivity
in MLL gene-rearranged infant acute lymphoblastic leukemia
Blood, 101 (4) (2003), pp. 1270-1276
https://www.sciencedirect.com/science/article/pii/S0889858809000859 22/33
7/4/22, 05:44 Leucemia linfoblástica aguda - ScienceDirect

Article Download PDF View Record in Scopus Google Scholar

37 L. Kager, M. Cheok, W. Yang, et al.


Folate pathway gene expression differs in subtypes of acute lymphoblastic leukemia and
influences methotrexate pharmacodynamics
J Clin Invest, 115 (1) (2005), pp. 110-117
View Record in Scopus Google Scholar

38 C.G. Mullighan, S. Goorha, I. Radtke, et al.


Genome-wide analysis of genetic alterations in acute lymphoblastic leukaemia
Nature, 446 (7137) (2007), pp. 758-764
CrossRef View Record in Scopus Google Scholar

39 S.A. Armstrong, J.E. Staunton, L.B. Silverman, et al.


MLL translocations specify a distinct gene expression profile that distinguishes a unique
leukemia
Nat Genet, 30 (1) (2002), pp. 41-47
View Record in Scopus Google Scholar

40 S.A. Armstrong, M.E. Mabon, L.B. Silverman, et al.


FLT3 mutations in childhood acute lymphoblastic leukemia
Blood, 103 (9) (2004), pp. 3544-3546
Article Download PDF View Record in Scopus Google Scholar

41 C.G. Mullighan, C.B. Miller, I. Radtke, et al.


BCR-ABL1 lymphoblastic leukaemia is characterized by the deletion of Ikaros
Nature, 453 (7191) (2008), pp. 110-114
CrossRef View Record in Scopus Google Scholar

42 T. Taketani, T. Taki, K. Sugita, et al.


FLT3 mutations in the activation loop of tyrosine kinase domain are frequently found in
infant ALL with MLL rearrangements and pediatric ALL with hyperdiploidy
Blood, 103 (3) (2004), pp. 1085-1088
Article Download PDF View Record in Scopus Google Scholar

43 M. Hatano, C.W. Roberts, M. Minden, et al.


Deregulation of a homeobox gene, HOX11, by the t(10;14) in T cell leukemia
Science, 253 (5015) (1991), pp. 79-82
CrossRef View Record in Scopus Google Scholar

44 M.A. Kennedy, R. Gonzalez-Sarmiento, U.R. Kees, et al.


HOX11, a homeobox-containing T-cell oncogene on human chromosome 10q24
Proc Natl Acad Sci U S A, 88 (20) (1991), pp. 8900-8904

https://www.sciencedirect.com/science/article/pii/S0889858809000859 23/33
7/4/22, 05:44 Leucemia linfoblástica aguda - ScienceDirect

CrossRef View Record in Scopus Google Scholar

45 P. Ballerini, A. Blaise, M. Busson-Le Coniat, et al.


HOX11L2 expression defines a clinical subtype of pediatric T-ALL associated with poor
prognosis
Blood, 100 (3) (2002), pp. 991-997
Article Download PDF View Record in Scopus Google Scholar

46 O.A. Bernard, M. Busson-LeConiat, P. Ballerini, et al.


A new recurrent and specific cryptic translocation, t(5;14) (q35;q32), is associated with
expression of the Hox11L2 gene in T acute lymphoblastic leukemia
Leukemia, 15 (10) (2001), pp. 1495-1504
CrossRef View Record in Scopus Google Scholar

47 V. Valge-Archer, A. Forster, T.H. Rabbitts


The LMO1 and LDB1 proteins interact in human T cell acute leukaemia with the
chromosomal translocation t(11;14) (p15;q11)
Oncogene, 17 (24) (1998), pp. 3199-3202
CrossRef View Record in Scopus Google Scholar

48 K.M. Carlson, C. Vignon, S. Bohlander, et al.


Identification and molecular characterization of CALM/AF10 fusion products in T cell
acute lymphoblastic leukemia and acute myeloid leukemia
Leukemia, 14 (1) (2000), pp. 100-104
CrossRef View Record in Scopus Google Scholar

49 M. Narita, K. Shimizu, Y. Hayashi, et al.


Consistent detection of CALM-AF10 chimaeric transcripts in haematological
malignancies with t(10;11) (p13;q14) and identification of novel transcripts
Br J Haematol, 105 (4) (1999), pp. 928-937
View Record in Scopus Google Scholar

50 V. Asnafi, I. Radford-Weiss, N. Dastugue, et al.


CALM-AF10 is a common fusion transcript in T-ALL and is specific to the
TCRgammadelta lineage
Blood, 102 (3) (2003), pp. 1000-1006
Article Download PDF View Record in Scopus Google Scholar

51 D. Caudell, P.D. Aplan


The role of CALM-AF10 gene fusion in acute leukemia
Leukemia, 22 (4) (2008), pp. 678-685
CrossRef View Record in Scopus Google Scholar

https://www.sciencedirect.com/science/article/pii/S0889858809000859 24/33
7/4/22, 05:44 Leucemia linfoblástica aguda - ScienceDirect

52 A.A. Ferrando, D.S. Neuberg, J. Staunton, et al.


Gene expression signatures define novel oncogenic pathways in T cell acute
lymphoblastic leukemia
Cancer Cell, 1 (1) (2002), pp. 75-87
Article Download PDF View Record in Scopus Google Scholar

53 C. Graux, J. Cools, L. Michaux, et al.


Cytogenetics and molecular genetics of T-cell acute lymphoblastic leukemia: from
thymocyte to lymphoblast
Leukemia, 20 (9) (2006), pp. 1496-1510
CrossRef View Record in Scopus Google Scholar

54 U.R. Kees, N.A. Heerema, R. Kumar, et al.


Expression of HOX11 in childhood T-lineage acute lymphoblastic leukaemia can occur in
the absence of cytogenetic aberration at 10q24: a study from the Children's Cancer Group
(CCG)
Leukemia, 17 (5) (2003), pp. 887-893
CrossRef View Record in Scopus Google Scholar

55 A.P. Weng, A.A. Ferrando, W. Lee, et al.


Activating mutations of NOTCH1 in human T cell acute lymphoblastic leukemia
Science, 306 (5694) (2004), pp. 269-271
View Record in Scopus Google Scholar

56 V. Asnafi, K. Beldjord, M. Libura, et al.


Age-related phenotypic and oncogenic differences in T-cell acute lymphoblastic leukemias
may reflect thymic atrophy
Blood, 104 (13) (2004), pp. 4173-4180
Article Download PDF View Record in Scopus Google Scholar

57 T. Szczepanski, A. Beishuizen, M.J. Pongers-Willemse, et al.


Cross-lineage T cell receptor gene rearrangements occur in more than ninety percent of
childhood precursor-B acute lymphoblastic leukemias: alternative PCR targets for
detection of minimal residual disease
Leukemia, 13 (2) (1999), pp. 196-205
CrossRef View Record in Scopus Google Scholar

58 V. van der Velden, M. Bruggemann, P.G. Hoogeveen, et al.


TCRB gene rearrangements in childhood and adult precursor-B-ALL: frequency,
applicability as MRD-PCR target, and stability between diagnosis and relapse
Leukemia, 18 (12) (2004), pp. 1971-1980
CrossRef View Record in Scopus Google Scholar

https://www.sciencedirect.com/science/article/pii/S0889858809000859 25/33
7/4/22, 05:44 Leucemia linfoblástica aguda - ScienceDirect

59 J.J. van Dongen, A.W. Langerak


Immunoglobulin and T-cell receptor gene rearrangements
C.H. Pui (Ed.), Childhood leukemias, Cambridge University Press, New York (2006), pp.
210-234
CrossRef View Record in Scopus Google Scholar

60 T.M. Breit, M.C. Verschuren, I.L. Wolvers-Tettero, et al.


Human T cell leukemias with continuous V(D)J recombinase activity for TCR-delta gene
deletion
J Immunol, 159 (9) (1997), pp. 4341-4349
View Record in Scopus Google Scholar

61 A. Beishuizen, K. Hahlen, A. Hagemeijer, et al.


Multiple rearranged immunoglobulin genes in childhood acute lymphoblastic leukemia
of precursor B-cell origin
Leukemia, 5 (8) (1991), pp. 657-667
View Record in Scopus Google Scholar

62 T. Szczepanski, M.J. Willemse, B. Brinkhof, et al.


Comparative analysis of Ig and TCR gene rearrangements at diagnosis and at relapse of
childhood precursor-B-ALL provides improved strategies for selection of stable PCR
targets for monitoring of minimal residual disease
Blood, 99 (7) (2002), pp. 2315-2323
Article Download PDF Google Scholar

63 K.A. Brady, S.K. Atwater, C.A. Lowell


Flow cytometric detection of CD10 (cALLA) on peripheral blood B lymphocytes of
neonates
Br J Haematol, 107 (4) (1999), pp. 712-715
View Record in Scopus Google Scholar

64 T.W. Froehlich, G.R. Buchanan, J.A. Cornet, et al.


Terminal deoxynucleotidyl transferase-containing cells in peripheral blood: implications
for the surveillance of patients with lymphoblastic leukemia or lymphoma in remission
Blood, 58 (2) (1981), pp. 214-220
Article Download PDF View Record in Scopus Google Scholar

65 N. Meru, A. Jung, I. Baumann, et al.


Expression of the recombination-activating genes in extrafollicular lymphocytes but no
apparent reinduction in germinal center reactions in human tonsils
Blood, 99 (2) (2002), pp. 531-537
Article Download PDF View Record in Scopus Google Scholar

https://www.sciencedirect.com/science/article/pii/S0889858809000859 26/33
7/4/22, 05:44 Leucemia linfoblástica aguda - ScienceDirect

66 M. Onciu, R.B. Lorsbach, E.C. Henry, et al.


Terminal deoxynucleotidyl transferase-positive lymphoid cells in reactive lymph nodes
from children with malignant tumors: incidence, distribution pattern, and
immunophenotype in 26 patients
Am J Clin Pathol, 118 (2) (2002), pp. 248-254
View Record in Scopus Google Scholar

67 R.W. McKenna, L.T. Washington, D.B. Aquino, et al.


Immunophenotypic analysis of hematogones (B-lymphocyte precursors) in 662
consecutive bone marrow specimens by 4-color flow cytometry
Blood, 98 (8) (2001), pp. 2498-2507
Article Download PDF Google Scholar

68 S.H. Kroft
Role of flow cytometry in pediatric hematopathology
Am J Clin Pathol, 122 (Suppl) (2004), pp. S19-S32
CrossRef Google Scholar

69 E.G. van Lochem, V. van der Velden, H.K. Wind, et al.


Immunophenotypic differentiation patterns of normal hematopoiesis in human bone
marrow: reference patterns for age-related changes and disease-induced shifts
Cytometry B Clin Cytom, 60 (1) (2004), pp. 1-13
View Record in Scopus Google Scholar

70 M.N. Dworzak, G. Fritsch, C. Fleischer, et al.


Multiparameter phenotype mapping of normal and post-chemotherapy B lymphopoiesis
in pediatric bone marrow
Leukemia, 11 (8) (1997), pp. 1266-1273
CrossRef View Record in Scopus Google Scholar

71 E. Tiacci, S. Pileri, A. Orleth, et al.


PAX5 expression in acute leukemias: higher B-lineage specificity than CD79a and
selective association with t(8;21)-acute myelogenous leukemia
Cancer Res, 64 (20) (2004), pp. 7399-7404
View Record in Scopus Google Scholar

72 Y.O. Huh, T.L. Smith, P. Collins, et al.


Terminal deoxynucleotidyl transferase expression in acute myelogenous leukemia and
myelodysplasia as determined by flow cytometry
Leuk Lymphoma, 37 (3-4) (2000), pp. 319-331
CrossRef View Record in Scopus Google Scholar

73 J.G. Rossi, M.S. Felice, A.R. Bernasconi, et al.


https://www.sciencedirect.com/science/article/pii/S0889858809000859 27/33
7/4/22, 05:44 Leucemia linfoblástica aguda - ScienceDirect

Acute leukemia of dendritic cell lineage in childhood: incidence, biological characteristics


and outcome
Leuk Lymphoma, 47 (4) (2006), pp. 715-725
CrossRef View Record in Scopus Google Scholar

74 J. Feuillard, M.C. Jacob, F. Valensi, et al.


Clinical and biologic features of CD4(+)CD56(+) malignancies
Blood, 99 (5) (2002), pp. 1556-1563
Article Download PDF View Record in Scopus Google Scholar

75 N.P. Chan, E.S. Ma, T.S. Wan, et al.


The spectrum of acute lymphoblastic leukemia with mature B-cell phenotype
Leuk Res, 27 (3) (2003), pp. 231-234
Article Download PDF View Record in Scopus Google Scholar

76 R. Komrokji, J. Lancet, R. Felgar, et al.


Burkitt's leukemia with precursor B-cell immunophenotype and atypical morphology
(atypical Burkitt's leukemia/lymphoma): case report and review of literature
Leuk Res, 27 (6) (2003), pp. 561-566
Article Download PDF View Record in Scopus Google Scholar

77 F. Navid, A.D. Mosijczuk, D.R. Head, et al.


Acute lymphoblastic leukemia with the (8;14) (q24;q32) translocation and FAB L3
morphology associated with a B-precursor immunophenotype: the Pediatric Oncology
Group experience
Leukemia, 13 (1) (1999), pp. 135-141
CrossRef View Record in Scopus Google Scholar

78 M. Onciu, F.G. Behm, S.C. Raimondi, et al.


ALK-positive anaplastic large cell lymphoma with leukemic peripheral blood involvement
is a clinicopathologic entity with an unfavorable prognosis. Report of three cases and
review of the literature
Am J Clin Pathol, 120 (4) (2003), pp. 617-625
CrossRef View Record in Scopus Google Scholar

79 J. Juco, J.T. Holden, K.P. Mann, et al.


Immunophenotypic analysis of anaplastic large cell lymphoma by flow cytometry
Am J Clin Pathol, 119 (2) (2003), pp. 205-212
View Record in Scopus Google Scholar

80 M. Ozdemirli, J.C. Fanburg-Smith, D.P. Hartmann, et al.


Differentiating lymphoblastic lymphoma and Ewing's sarcoma: lymphocyte markers and
gene rearrangement
https://www.sciencedirect.com/science/article/pii/S0889858809000859 28/33
7/4/22, 05:44 Leucemia linfoblástica aguda - ScienceDirect

Mod Pathol, 14 (11) (2001), pp. 1175-1182


View Record in Scopus Google Scholar

81 C.J. Buresh, B.R. Oliai, R.T. Miller


Reactivity with TdT in Merkel cell carcinoma: a potential diagnostic pitfall
Am J Clin Pathol, 129 (6) (2008), pp. 894-898
CrossRef View Record in Scopus Google Scholar

82 H.Y. Dong, W. Liu, P. Cohen, et al.


B-cell specific activation protein encoded by the PAX-5 gene is commonly expressed in
merkel cell carcinoma and small cell carcinomas
Am J Surg Pathol, 29 (5) (2005), pp. 687-692
CrossRef View Record in Scopus Google Scholar

83 S. Denzinger, M. Burger, C.G. Hammerschmied, et al.


Pax-5 protein expression in bladder cancer: a preliminary study that shows no correlation
to grade, stage or clinical outcome
Pathology, 40 (5) (2008), pp. 465-469
Article Download PDF CrossRef View Record in Scopus Google Scholar

84 P. Mhawech-Fauceglia, R. Saxena, S. Zhang, et al.


Pax-5 immunoexpression in various types of benign and malignant tumours: a high-
throughput tissue microarray analysis
J Clin Pathol, 60 (6) (2007), pp. 709-714
CrossRef View Record in Scopus Google Scholar

85 E. Torlakovic, A. Slipicevic, C. Robinson, et al.


Pax-5 expression in nonhematopoietic tissues
Am J Clin Pathol, 126 (5) (2006), pp. 798-804
View Record in Scopus Google Scholar

86 N. Gokbuget, D. Hoelzer
Treatment of adult acute lymphoblastic leukemia
Semin Hematol, 46 (1) (2009), pp. 64-75
Article Download PDF View Record in Scopus Google Scholar

87 H. Kantarjian, D. Thomas, S. O'Brien, et al.


Long-term follow-up results of hyperfractionated cyclophosphamide, vincristine,
doxorubicin, and dexamethasone (Hyper-CVAD), a dose-intensive regimen, in adult acute
lymphocytic leukemia
Cancer, 101 (12) (2004), pp. 2788-2801
View Record in Scopus Google Scholar

https://www.sciencedirect.com/science/article/pii/S0889858809000859 29/33
7/4/22, 05:44 Leucemia linfoblástica aguda - ScienceDirect

88 C.H. Pui, M.V. Relling, W.E. Evans


Role of pharmacogenomics and pharmacodynamics in the treatment of acute
lymphoblastic leukaemia
Best Pract Res Clin Haematol, 15 (4) (2002), pp. 741-756
Article Download PDF View Record in Scopus Google Scholar

89 J.J. Yang, C. Cheng, W. Yang, et al.


Genome-wide interrogation of germline genetic variation associated with treatment
response in childhood acute lymphoblastic leukemia
JAMA, 301 (4) (2009), pp. 393-403
CrossRef View Record in Scopus Google Scholar

90 Q. Cheng, W. Yang, S.C. Raimondi, et al.


Karyotypic abnormalities create discordance of germline genotype and cancer cell
phenotypes
Nat Genet, 37 (8) (2005), pp. 878-882
CrossRef View Record in Scopus Google Scholar

91 T. Szczepanski
Why and how to quantify minimal residual disease in acute lymphoblastic leukemia?
Leukemia, 21 (4) (2007), pp. 622-626
CrossRef View Record in Scopus Google Scholar

92 M. Bruggemann, T. Raff, T. Flohr, et al.


Clinical significance of minimal residual disease quantification in adult patients with
standard-risk acute lymphoblastic leukemia
Blood, 107 (3) (2006), pp. 1116-1123
Article Download PDF CrossRef View Record in Scopus Google Scholar

93 E.R. van Wering, A. Beishuizen, E.T. Roeffen, et al.


Immunophenotypic changes between diagnosis and relapse in childhood acute
lymphoblastic leukemia
Leukemia, 9 (9) (1995), pp. 1523-1533
View Record in Scopus Google Scholar

94 D. Campana
Determination of minimal residual disease in leukaemia patients
Br J Haematol, 121 (6) (2003), pp. 823-838
View Record in Scopus Google Scholar

95 D. Campana
Minimal residual disease in acute lymphoblastic leukemia
Semin Hematol, 46 (1) (2009), pp. 100-106
https://www.sciencedirect.com/science/article/pii/S0889858809000859 30/33
7/4/22, 05:44 Leucemia linfoblástica aguda - ScienceDirect

Article Download PDF View Record in Scopus Google Scholar

96 D. Campana, E. Coustan-Smith
Minimal residual disease studies by flow cytometry in acute leukemia
Acta Haematol, 112 (1–2) (2004), pp. 8-15
View Record in Scopus Google Scholar

97 E. Coustan-Smith, J. Sancho, M.L. Hancock, et al.


Use of peripheral blood instead of bone marrow to monitor residual disease in children
with acute lymphoblastic leukemia
Blood, 100 (7) (2002), pp. 2399-2402
Article Download PDF View Record in Scopus Google Scholar

98 M. Bruggemann, V. van der Velden, T. Raff, et al.


Rearranged T-cell receptor beta genes represent powerful targets for quantification of
minimal residual disease in childhood and adult T-cell acute lymphoblastic leukemia
Leukemia, 18 (4) (2004), pp. 709-719
CrossRef View Record in Scopus Google Scholar

99 T. Szczepanski, V. van der Velden, T. Raff, et al.


Comparative analysis of T-cell receptor gene rearrangements at diagnosis and relapse of
T-cell acute lymphoblastic leukemia (T-ALL) shows high stability of clonal markers for
monitoring of minimal residual disease and reveals the occurrence of second T-ALL
Leukemia, 17 (11) (2003), pp. 2149-2156
CrossRef View Record in Scopus Google Scholar

100 I. Thorn, J. Botling, M. Hermansson, et al.


Monitoring minimal residual disease with flow cytometry, antigen-receptor gene
rearrangements and fusion transcript quantification in Philadelphia-positive childhood
acute lymphoblastic leukemia
Leuk Res (2009 Jan 19)
[Epub ahead of print]
Google Scholar

101 C.G. Mullighan, X. Su, J. Zhang, et al.


Deletion of IKZF1 and Prognosis in Acute Lymphoblastic Leukemia
N Engl J Med (2009)
Google Scholar

102 N.L. Lo, G. Cazzaniga, A. Di Cataldo, et al.


Clonal stability in children with acute lymphoblastic leukemia (ALL) who relapsed five or
more years after diagnosis
Leukemia, 13 (2) (1999), pp. 190-195
https://www.sciencedirect.com/science/article/pii/S0889858809000859 31/33
7/4/22, 05:44 Leucemia linfoblástica aguda - ScienceDirect

Google Scholar

103 W. Chen, N.J. Karandikar, R.W. McKenna, et al.


Stability of leukemia-associated immunophenotypes in precursor B-lymphoblastic
leukemia/lymphoma: a single institution experience
Am J Clin Pathol, 127 (1) (2007), pp. 39-46
View Record in Scopus Google Scholar

104 J. Zuna, H. Cave, C. Eckert, et al.


Childhood secondary ALL after ALL treatment
Leukemia, 21 (7) (2007), pp. 1431-1435
CrossRef View Record in Scopus Google Scholar

105 C.G. Mullighan, L.A. Phillips, X. Su, et al.


Genomic analysis of the clonal origins of relapsed acute lymphoblastic leukemia
Science, 322 (5906) (2008), pp. 1377-1380
CrossRef Google Scholar

Cited by (104)

CD99 in malignant hematopoiesis


2022, Experimental Hematology

Aqua-Plyometric Exercises: Potential Implications for Bone Mineral Density,


Functional Capacity, and Quality of Life in Survivors of Childhood Acute
Lymphoblastic Leukemia
2021, Seminars in Oncology Nursing

Show abstract

Tonsillectomy and hematologic malignancy: Should routine pediatric tonsillectomy


specimens be sent to pathology?
2021, International Journal of Pediatric Otorhinolaryngology

Show abstract

https://www.sciencedirect.com/science/article/pii/S0889858809000859 32/33
7/4/22, 05:44 Leucemia linfoblástica aguda - ScienceDirect

Differential expression of CD73, CD86 and CD304 in normal vs. leukemic B-cell
precursors and their utility as stable minimal residual disease markers in childhood
B-cell precursor acute lymphoblastic leukemia
2019, Journal of Immunological Methods

Show abstract

Vincristine-induced vocal cord paresis and paralysis in children


2019, International Journal of Pediatric Otorhinolaryngology

Citation Excerpt :
…Acute lymphoblastic leukemia (ALL) is the most common type of childhood cancer, with approximately
6,000 new cases diagnosed each year [1]. The disease comprises a set of lymphoid neoplasms that are
morphologically similar to B-lineage and T-lineage precursor cells [2]. Peak onset of ALL is between 2 and 5
years of age with survival rates of 90% in children [1].…

Show abstract

CircPVT1: a pivotal circular node intersecting Long Non-Coding-PVT1 and c-MYC


oncogenic signals
2022, Molecular Cancer

View all citing articles on Scopus

View Abstract

Copyright © 2009 Elsevier Inc. All rights reserved.

Copyright © 2022 Elsevier B.V. or its licensors or contributors.


ScienceDirect ® is a registered trademark of Elsevier B.V.

https://www.sciencedirect.com/science/article/pii/S0889858809000859 33/33

You might also like