You are on page 1of 11

See discussions, stats, and author profiles for this publication at: https://www.researchgate.

net/publication/260487636

Acute Lymphoblastic Leukemia: Characterization and its Prognostic Values

Article · October 2013

CITATION READS

1 691

2 authors:

Radha Rathee Neelkamal Sharma


Maharshi Dayanand University Maharshi Dayanand University
16 PUBLICATIONS   34 CITATIONS    24 PUBLICATIONS   12 CITATIONS   

SEE PROFILE SEE PROFILE

Some of the authors of this publication are also working on these related projects:

Wild life forensics View project

Wildlife forensic indentification View project

All content following this page was uploaded by Radha Rathee on 05 March 2014.

The user has requested enhancement of the downloaded file.


International Journal of Pharma Research & Review, Nov 2013; 2(11):27-36

Review Article

Acute Lymphoblastic Leukemia: Characterization and its Prognostic Values


Neelkamal, *Radha Rathee

Department of Genetics, M. D. University, Rohtak, Haryana, India.


________________________________________________________________________________________________________________________________________________________

ABSTRACT
Acute lymphoblastic leukemia is a type of leukemia which is characterized by 20% or more lymphoblasts
in the bone marrow and/or the blood. It is a rapidly developing, abnormal growth of the cells that are
precursors of lymphoblasts. The peak incidence occurs between age 2 and 5 years. The most frequent
signs are lymphadenopathies, hepatosplenomegaly, fever, anemia, signs of hemorrhage, and bone
tenderness. Biological findings include hyperleukocytosis due to circulating lymphoblasts. Most of the
cases of acute lymphoblastic leukemia show chromosomal and genetic abnormalities. These anomalies
occur spontaneously in important regulatory genes in a lymphoid cell population. The causative factors
may be like smoking, high birth weight, diet, and high socioeconomic status, electromagnetic field, being
exposed to radiation, pesticides, past treatment with chemotherapy or other drugs that weaken
the immune system. It is a biologically heterogeneous disorder, so that morphologic, immunologic,
cytogenetic, and molecular genetic characterizations of leukemia lymphoblasts are needed to establish
the diagnosis or to exclude other possible causes of bone marrow failure and, finally, to classify its
subtypes. The survival rate for children younger than 15 years of age reaches about 75%, but, despite the
significant improvement of outcome during the last decades, still roughly 25% of patients suffer from a
relapse of the disease. With the need to stratify patients in risk groups and to provide risk-adapted
therapy, treatment requires high levels of organization, expertise and knowledge.

Keywords: Acute lymphoblastic leukemia, associated risk factor, etiology, prognosis.

Received 06 Sept 2013 Received in revised form 28 Sept 2013 Accepted 05 Oct 2013

*Address for correspondence:


Radha Rathee
Research Scholar, Lab No. 22, Dept. of Genetics, M. D. University, Rohtak-124001, Haryana, India.
E-mail: radharathee@gmail.com
________________________________________________________________________________________________________________________________________________________

INTRODUCTION
Leukemia is a cancer of the blood or bone cytoplasm and prominent cytoplasmic
marrow characterized by an abnormal vacuolization. Approximately 85% of
increase of blood cells, usually white blood children with ALL have predominant L1
cells or leukocytes. Acute lymphoblastic morphology, 14% have L2, and 1% has L3,
leukemia is a rapidly developing, abnormal while the L2 subtype is more common in
growth (neoplasm) of the cells that are adults [3]. Individual chromosomal
precursors of lymphoblasts. The French- abnormalities are strong independent
American-British Cooperative Working indicators of outcome, especially risk of
Group defines three categories of relapse. Diagnostic cytogenetics identifies
lymphoblasts i.e. L1, L2 & L3. L1 patients with a higher rate of relapse and
lymphoblasts are small cells characterized those who are likely to have a high-risk
by a high nucleus-to-cytoplasm ratio [1]. relapse [4].
The pale blue cytoplasm is scanty and is Epidemiology:
limited to a small portion of the perimeter Reported annual incidence of ALL is
of the cell. The L2 lymphoblasts have approximately 9-10 cases per 100,000
indistinct nucleoli and nuclear membranes populations in childhood [5]. The overall
that vary from round to clefted [2]. L3 survival according to age, with follow-up of
lymphoblasts are a heterogeneous group of all patients in December 2008 through the
cells identical to Burkitt-like leukemia and population registry supports the strong
characterized by deeply basophilic correlation between age and outcome [6, 7].

Radha Rathee et.al, IJPRR 2013; 2(11) 27


International Journal of Pharma Research & Review, Nov 2013; 2(11):27-36

In 2013, leukemia is expected to strike female ratio was 2.03:1 (Fig.1) that is much
approximately 12 times as many adults higher than what is seen in the developed
(43,749) as children and adolescents world [15]. In developed countries, the age
younger than 15 years (3,605) [1, 8]. In distribution of ALL shows a major peak at
India, 60-85% of all leukemias reported are pre-school age (between 1 and 5 years of
acute lymphoblastic leukemia [3]. There age) with a slow decline toward
were geographic variations in frequency of adolescence [16]. The age distribution of
leukemia [9]. ALL is reported to be the most children of ALL in developed countries
frequent in the south [10] and intermediate shows a very marked early peak between 2-
in the East, West [11] and central India [12]. 5 years, followed by a small peak between
Interestingly, the incidence of ALL is lesser 11-15 years and the median age of 4 years
in east India [13] as well as Northern areas [17, 18, 19]. There has been a gradual
[14]. In a study from Haryana by Kumar et increase in the incidence of ALL in the past
al, there were 70.2% children and 29.8% 25 years [20].
adult patients of ALL in which male to

Figure 1: Percentage Frequency of Sex Ratio in Patients of Acute Lymphocytic Leukemia.

Etiology: cell death called apoptosis. A translocation


In ALL lymphoblast are produced by a part can activate a proto-oncogene by two
of DNA which is called proto-oncogene. different mechanisms. A more frequent
Most of the proto-oncogenes involved in event is a merger of two genes to form a
leukemia encode transcription factors, fusion gene that produces abnormal
many of which have revealed to be chimaeric protein inducing leukemia. As an
important regulators of the proliferation, example, translocation t (1; 19) in ALL
differentiation and survival of blood cell creates the fusion of E2A (immunoglobulin
precursors [21, 22]. In leukemia’s, including enhancer binding factors E12/E47) and
ALL, chromosomal translocations occur PBX1 (pre-B-cell leukemia transcription
regularly. It is thought that most factor 1) genes. In the E2A-PBX1 fusion
translocations occur before birth during protein transactivating domains of E2A are
fetal development. These translocations joined to the DNA-binding domain of PBX1,
create a rearrangement of genes, which which alters the transcriptional properties
leads a so-called protooncogene to of the PBX1 transcription factor [23, 24].
transform into an oncogene. The oncogene Another event that may initiate leukemia is
causes leukemia either by stimulating cell inactivation of a tumor suppressor gene.
division or by inhibiting the programmed Tumor suppressor genes are essential for

Radha Rathee et.al, IJPRR 2013; 2(11) 28


International Journal of Pharma Research & Review, Nov 2013; 2(11):27-36

normal cell development and they prevent Ionizing radiation is considered a ‘known’
carcinogenesis. Very few tumor suppressor cause of childhood leukemia. The risk is also
genes have been reported in acute higher for those exposed at an earlier age
leukemias. Screening for chromosomal [32] and secondary leukemias in the
regions with loss of heterozygosity (LOH) is individuals treated by radiotherapy
one way to track novel tumor suppressor [33]. Radiation from nuclear power plants is
genes. In childhood ALL, the regions that also a known cause for both kinds of
most frequently show LOH are the short leukemia [34]. X-ray examinations of
arms of chromosomes 9 and 12 (in about pregnant women may be associated with
30-40% and 25-30% of the patients, increased risk of subsequent childhood ALL
respectively) [25, 26]. [35]. A study found that exposure to X rays
Scientific research has shown that all after birth increased the risk of leukemia.
malignancies are due to subtle or less subtle Infants receiving diagnostic X-rays had 60%
changes in DNA that lead to unimpaired cell more leukemia than other children [36].
division and breakdown of inhibitory The exposure to post-natal diagnostic X-
processes. Many of the described molecular rays is associated with increased risk of
mutations bear evidence of childhood ALL, specifically B-cell ALL, but
immunoglobulin joining region (VDJ) and T- not AML or T-cell ALL [37].
cell receptor (TcR) recombinase activity. A Several studies have linked leukemia to
few other studies have also evaluated the pesticides. One large recent study of 491
possible role of infections during infancy in children with ALL found that risk was
the etiology of ALL, with some showing a increased by home use of some kinds of
protective effect [27] and others suggesting pesticides and by use of multiple different
the opposite [28]. pesticides. Herbicide use during pregnancy
Another mechanism by which a was associated with 50% increase in risk. A
translocation causes leukemia is transfer of study of nearly 2,000 children found that
a normally quiet transcription factor gene the risk of acute lymphoblastic leukemia
to the neighborhood of active promoter or was increased if the children's mothers
enhancer elements, which accelerate the were exposed to solvents, paints, or
function of the gene. For example, in thinners before conception or during
translocations t(8;14), t(2;8) and t(8;22) in pregnancy or to plastics after birth. The
Burkitt leukemia, the gene encoding the father's exposure to plastics before
MYC transcription factor is exposed to the conception was associated with greater
enhancer elements of an immunoglobulin risk. This study reported that the timing of
gene. These enhancer elements cause over exposure was an important factor [38].
expression of the MYC gene, which is Several studies have found that exposure to
important in the regulation of cell division electro magnetic field (EMFs) increases risk
and cell death [29]. Further of leukemia for children [37]. It was found
characterization of these genes revealed that children living near high voltage power
that they are often involved directly or installations were more likely to be found to
indirectly in the development and have leukemia than other children [39]. One
homeostasis of normal blood cells, and that recent study found that risk of leukemia
abnormal protein products of fusion genes was elevated when exposure to EMFs was
created by specific translocations and consistent over the term of the pregnancy
inversions can deregulate proliferation, and in cases where the design of the water
differentiation or programmed cell death system in the home led to ‘ground currents’
(apoptosis) of blood cell precursors [30, from connections between plumbing pipes
31]. and the grounding for the electricity [40].
Risk Factors: Only one environmental risk factor
The causative factor of ALL is unknown but (ionizing radiation) has been significantly
there are some risk factors which are linked with either ALL or AML; most
known to be associated with ALL. These environmental risk factors [e.g.,
factors are like ionizing radiation, electromagnetic fields (EMFs), cigarette
pesticides, smoking, chemicals, and EMFs. smoking] have been weakly or

Radha Rathee et.al, IJPRR 2013; 2(11) 29


International Journal of Pharma Research & Review, Nov 2013; 2(11):27-36

inconsistently associated with either form events that occur postnatally [33]. There is
of childhood leukemia [41]. little indication that propensity for ALL is
Some genetic diseases have also association passed on from parents to children.
with acute lymphocytic leukemia. Children Immunophenotyping:
with trisomy 21 (i.e., Down syndrome) are Numerous immunophenotypic features
up to 15 times more likely to develop have been examined for their potential
leukemia than normal children. Other less prognostic value. Flow cytometry is the
common pre-existing chromosomal preferred method of diagnosis and
abnormalities have been linked to leukemia, immunophenotyping acute lymphoblastic
included are Klinefelter's syndrome, Bloom leukemia. ALL presumably arises from
syndrome, and Fanconi's anemia. Lymphoid malignant transformation of B- or T-cell
malignancies, with a predominance of T- progenitor cells. B-cell leukemia occurs
ALL, have been reported in patients with more frequently than T-cell leukemia. The
ataxia-telangiectasia (AT), an autosomal ALL arise from B-cell in 85% patients and
recessive disorder characterized by from T-cell in 15% cases. B-cell is more
increased chromosomal fragility [42, 43, 44, commonly seen in children, but can occur at
45]. Literature supports the hypothesis that any age. It is the most common type of
an infection is involved in the etiogy of leukemia found in children (nearly 75% of
acute lymphocytic leukemia in children, cases occur in children under six years of
particulrily those cases occuring in children age), although it affects both children and
between 2 and 5 years of age [9, 46, 47]. adults [53]. In children, B-cell ALL accounts
Viruses have also been linked to some for 60-80% cases whereas T-cell comprises
forms of leukemia. Assosiation of human T- only 11-20% (Fig. 2) [15]. T-cell ALL
cell lymphotrophic virus type 1 with adult represents approximately 15% to 20% of
T-cell leukemia, of epstein-Barr virus with all cases of ALL in Western countries [54,
mature B-cell ALL and of human 55]. The stages of ALL include Early pre-B
imunodeficiency virus (HIV) with ALL, Common ALL, Pre-B-cell ALL, Mature
lymphoproliferative disorders have been B-cell ALL (Burkitt leukemia), Pre-T-cell
described [48, 49]. ALL and Mature T-cell ALL [56]. B- and T-
Many cases of ALL that develop in children cell lymphoblastic leukemia cells express
have a prenatal origin. Evidence in support surface antigens that parallel their
of this comes from the observation that the respective lineage developments. Precursor
immunoglobulin or T-cell receptor antigen B-cell ALL cells typically express CD10,
rearrangements that are unique to each CD19, and CD34 on their surface along, with
patient’s leukemia cells can be detected in nuclear terminal deoxynucleotide
blood samples obtained at birth. Similarly, transferase (TdT), while precursor T-cell
there are data that patients with ALL ALL cells commonly express CD2, CD3, CD7,
characterized by specific chromosomal CD34, and TdT [57]. In a study by Bayram et
abnormalities had blood cells carrying the al the most frequently detected five
abnormalities at the time of birth [50, 51, antigens were I2, CD10, CD41, CD2 and
52]. Genetic studies of identical twins with CD7/CD19 at the time of diagnosis and
concordant leukemia further support the CD41, I2, CD10, CD19 and CD2 at the time of
prenatal origin of some leukemia [52]. The relapse. Flow cytometric investigations
emerging association between certain revealed that antigen levels determined at
combinations of dermatoglyphic traits and the time of diagnosis increased or
specific chromosome aberrations quickly decreased by 10% at the time of relapse
established a useful diagnostic and an [58]. CD19 is also expressed on the earliest
integral part of the medical diagnostic [50]. B-precursor lymphocytes that are
In pediatric leukemia, data have indicated malignantly transformed in ALL. Therefore,
that most chromosome translocations and B-lineage ALL seems to be most suitable for
preleukemic clones arise in utero during a bispecific approach aimed at CD19 [59].
fetal hematopoiesis with secondary genetic

Radha Rathee et.al, IJPRR 2013; 2(11) 30


International Journal of Pharma Research & Review, Nov 2013; 2(11):27-36

Figure 2: Percentage Frequency of Cell Type in Children of Acute Lymphocytic Leukemia.

Cytogenetics: Chromosomes in Leukemia (TIWCL) found


There have been many technical difficulties the majority of cytogenetic changes in cases
which make it difficult to gain information of B precursor ALL, with only 39%
for chromosomal findings in ALL. occurring in T-cell ALL [64, 66].
Chromosome studies in ALL exhibit poor Most studies on karyotypic abnormalities
morphology; chromosomes tend to spread and their clinical significance have been
poorly, and appear blurred and fuzzy with performed in childhood ALL. Adult ALL
indistinct margins, making banding studies showed nonrandom chromosomal
challenging or even impossible [60, 61]. abnormalities similar to those found in
Williams et al used a direct technique of childhood ALL, but their distribution and
bone marrow (BM) chromosomal analysis their biological significance were different.
by using specific flaming techniques and However, in adult ALL the role of
modified G-banding has been developed by cytogenetics in patient management has
Williams et al [61]. They identified clonal largely been centered on the presence of the
karyotypic abnormalities in 94% to 98% of Philadelphia (Ph) chromosome which
cases of ALL. Such improved techniques usually arises from t(9;22)(q34;q11.2) and
also detected randomly occurring results in BCR-ABL fusion [68]. Among the
cytogenetic abnormalities in cases with several changes, ploidy distribution and
hyperdiploid chromosome numbers (>50) recurrent translocation associated with
that had previously been classified as specific morphology and immunphenotype
normal in karyotype [61]. These results are well recognized in ALL. Numerical
showed a high prevalence of clonal chromosomal abnormalities alone are less
chromosomal abnormalities in ALL, as was common in adult ALL, possibly reflecting a
shown for acute non lymphoblastic fundamental difference in the pathogenesis
leukemias by Yunis, who used high- between childhood and adult ALL [69].
resolution banding techniques [62]. The Among adults, patients with normal
majority of cases of ALL demonstrate an karyotype and those with isolated
abnormal karyotype, either in chromosome 9p/CDKN2A-CDKN2B deletions had a
number (ploidy) or as structural changes relatively favorable (standard) prognosis,
such as translocations, inversions, or whereas those with 6q deletions,
deletions. These changes were detected in miscellaneous, and hyperdiploid karyotype
only half of ALL patients in the first banding had an intermediate prognosis, and patients
studies [63]. Improvements in spreading with t(9;22)/BCR/ ABL1,t(4;11)/MLL/AF4,
and banding techniques have resulted in t(1;19)/TCF3/PBX1 constituted the
higher rates of detection, and most studies unfavorable prognosis group [70]. In
now report chromosomal changes in 60% childhood ALL numerous good and high-
to 85% of ALL cases [64, 65, 66, 67]. The risk cytogenetic subgroups have been
third International Workshop on

Radha Rathee et.al, IJPRR 2013; 2(11) 31


International Journal of Pharma Research & Review, Nov 2013; 2(11):27-36

identified which are regularly used to chemotherapy in order to minimize the side
stratify patients to particular therapies [71]. effects of the treatment, whereas patients
Diagnosis: with high risk may receive intensive
ALL is diagnosis with a medical treatment including stem cell
history, physical examination, complete transplantation. Therefore the differences
blood count, blood smears, cytogenetics and in the overall outcomes between the
immunophenotyping. The higher the white different risk groups have reduced in recent
blood cell counts the worse the prognosis years. The first aim of the treatment is to
[72]. Blast cells are seen on blood smear in reach remission, a condition in which the
majority of cases. clinical symptoms have disappeared and no
Pathological examination, cytogenetics (in leukemic cells can be detected by
particular the presence of Philadelphia conventional methods. The treatment of
chromosome), and immunophenotyping childhood ALL takes 2-2.5 years. The
establish whether myeloblastic treatment results have significantly
(neutrophils, eosinophils, or basophils) or improved during the past two decades, and
lymphoblastic (B lymphocytes or T at present up to 80% of the childhood
lymphocytes) cells are the problem. RNA patients’ recovered [75]. The rate is much
testing can establish how aggressive the higher than adults with ALL, of whom only
disease is; different mutations have been 30-40% patients were cured [71].
associated with shorter or longer survival. ALL is the most common childhood
Medical imaging can find invasion of malignancy; dramatic advances in its
other organs commonly the lung, liver, treatment over the past three decades have
spleen, lymph nodes, brain, kidneys, and changed it from a universally fatal to an
reproductive organs [73, 74]. ALL almost curable disease in 85% of cases. As
lymphoblasts were classified using the pediatric oncologists have become more
French-American-British (FAB) criteria as successful at treating ALL, much of the
having L1, L2 and L3 morphology. Most clinical research efforts have focused on
cases of ALL that show L3 morphology stratifying patients into various risk groups
express surface immunoglobulin (Ig) and based on known prognostic features, so that
have a C-MYC gene translocation identical to patients with lower-risk disease could be
that seen in Burkitt lymphoma i.e. t(8;14). treated less intensively with much less side
On the bases of Immunophenotype the effects and toxicities, while patients with a
World Health Organization (WHO) classifies higher risk of treatment failure could be
ALL as either B or T lymphoblastic targeted for more aggressive therapies [76].
leukemia. B lymphoblastic leukemia is In the risk classification of ALL, not only
subdivided by the presence or absence of cytogenetic alterations, but also many other
specific recurrent genetic abnormalities i.e. factors are taken into account. These
t(9;22), MLL rearrangement, t(12;21), include, for example, white blood cell count
hyperdiploidy, hypodiploidy, t(5;14), and (WBC) at diagnosis, age, response to
t(1;19). primary therapy and the phenotype of the
Prognosis: blasts (precursor-B cell / immature B cell /
Four main treatment elements can be T cell) [77]. The groups of patients formed
generally recognized in chemotherapy according to the existing criteria however,
protocols adopted by international quite heterogeneous as regards the
cooperative groups: induction with the aim outcome of the patients, leading to
of complete remission, CNS preventive excessive treatment of some patients and
therapy, consolidation and maintenance failure of treatment in others.
therapy. ALL treated with chemotherapy Remission and Survival:
and the cases with poor prognosis are also The leukemia karyotype has emerged as
with stem cell transplantation. The form one of the most important factors in both
and intensity of the treatment are childhood and adult ALL. The cytogenetic
determined based on the risk group. abnormalities confer important prognostic
Patients with good or standard risk may be information in ALL was first reported by
given less intensive conventional Secker-Walker et al in 1982 in a series of

Radha Rathee et.al, IJPRR 2013; 2(11) 32


International Journal of Pharma Research & Review, Nov 2013; 2(11):27-36

childhood ALL [78]. Complete remission CONCLUSION


(CR) rates, remission durations, as well as The present study has identified several
disease-free-survivals (DFS) were risk factors of developing ALL so general
significantly affected by the karyotypic awareness can be made in society to reduce
abnormalities [64]. Cytogenetic studies in the load of disease. Cytogenetic
childhood ALL have associated a better abnormalities were found associated with
prognosis with hyperdiploid karyotype and clinical and prognostic factors so further
a worse prognosis with balanced investigation may help to know mechanism
translocation [78, 79]. Among adult patients behind this. With identification of risk
the highest likelihood of cure (21% to 30%) categories on the basis of cytogenetic
was projected in patients with chromosome groups, patients with low risk may be given
numbers of >50, or 47 to 50, with 6q2, or less intensive conventional chemotherapy
with a normal karyotype [80]. avoiding the toxic effect of treatment
The importance of cytogenetics, as the whereas patients with high risk may receive
single most important prognostic factor in intensive treatment including stem cell
adult ALL, has been reported previously by transplantation. Improvements in
the CALGB and by the GIMEMA and MRC cytogenetic techniques have yielded
UKALLXII/ECOG study groups [67, 70, 81, significant insight as to the importance of
82, 83]. The correlation of the karyotype in cytogenetic abnormalities in the
ALL with other recognized prognostic factor pathophysiology and prognosis of
is an independent prognostic not only in hematological malignancies.
childhood [81] but also in adult patients ACKNOWLEDGMENT
[82]. Based on clinical risk criteria as well We would like to express gratitude to Dr.
as modern laboratory investigation Minakshi Vashist, Prof & Head,Deptt. Of
including cytogenetics, patients can be Genetics, M.D.U. Rohtak, Dr. Sunita Singh
divided into prognostic and assigned to Prof. Deptt. of Pathology, Dr. Pankaj Abrol,
risk-adjusted treatment protocols. The Head, Deptt. of Paedeatics and Dr. Gahlot,
clinical outcome of patients with Head, Deptt of Medicine Pt. B. D. Sharma
hyperdiploidy varies in different series, University of Health Sciences, Rohtak for
being more favorable in children than in the cooperation and help in collecting data.
adults, where a poor outcome has been I express my sincere thank to UGC for
repeatedly reported [65, 82]. ALL is a funding this project.
biologically heterogeneous disorder, so that REFERENCES
morphologic, immunologic, cytogenetic, and 1. Conter V, Bartram CR, Valsecchi MG, Schrauder
molecular genetic characterizations of A, Panzer-Grümayer R, Möricke A, et al.
Molecular response to treatment redefines all
leukemia lymphoblasts are needed to
prognostic factors in children and adolescents
establish the diagnosis or to exclude other with B-cell precursor acute lymphoblastic
possible causes of bone marrow failure and, leukemia: results in 3184 patients of the
finally, to classify ALL subtypes. The AIEOP-BFM ALL 2000 study. Blood
survival rate for children younger than 15 2010;115(16):3206-14.
years of age reaches about 75%, but, 2. Robison LL. Late Effects of Acute Lymphoblastic
despite the significant improvement of Leukemia Therapy in Patients Diagnosed at 0-
20 Years of Age. ASH Education Program Book
outcome during the last decades, still 2011;2011(1):238-42.
roughly 25% of patients suffer from a 3. Arora RS, Eden TOB, Kapoor G. Epidemiology of
relapse of the disease [83]. Even if the childhood cancer in India. Indian Journal of
management of relapse remains largely Cancer 2009;46(4):264-73.
controversial, an increasing use of high dose 4. Moorman AV, Richards SM, Robinson
chemotherapy blocks and stem cell HM, Strefford JC, Gibson BE, Kinsey SE, et
al. Prognosis of children with acute
transplantation is adopted in most cases.
lymphoblastic leukemia (ALL) and
With the need to stratify patients in risk intrachromosomal amplification of
groups and to provide risk-adapted therapy, chromosome 21
treatment requires high levels of (iAMP21). Blood 2007;109, 2327–30.
organization, expertise and knowledge. 5. Ribera JM, Oriol A. Acute lymphoblastic
leukemia in adolescents and young adults.

Radha Rathee et.al, IJPRR 2013; 2(11) 33


International Journal of Pharma Research & Review, Nov 2013; 2(11):27-36

Hematol Oncol Clin North 21. Biondi A, Cimino G, Pieters R, Pui CH. Biological
Am 2009;23(5):1033-42. and therapeutic aspects of infant leukemia.
6. Kent EE, Sender LS, Largent JA, Anton-Culver H. Blood 2000;96:24–33.
Leukemia survival in children, adolescents, and 22. Smith MA, Seibel NL, Altekruse SF, Ries LAG,
young adults: influence of socioeconomic status Melbert DL, O'Leary M, et al. Outcomes for
and other demographic factors. International Children and Adolescents with Cancer:
Journal of Studies of Cancer in Human Challenges for the Twenty-First Century.
Populations 2009;20(8):1409-20. Journal of Clinical Oncology 2010;28(15):2625-
7. Hunger SP, Lu X, Devidas M, Camitta BM, 34.
Gaynon PS, Winick NJ. Improved survival for 23. Lu Q, Wright DD, Kamps MP. Fusion with E2A
children and adolescents with acute converts the Pbx1 homeodomain protein into a
lymphoblastic leukemia between 1990 and constitutive transcriptional activator in human
2005: a report from the children's oncology leukemias carrying the t(1;19). Mol Cell
group. J Clin Oncol 2012;30(14):1663-9. Biol 1994; 14:3938-42.
8. Cancer Facts & Figures 2013. Atlanta, GA: 24. Hunger SP, Lu X, Devidas M, Camitta BM,
American Cancer Society; 2013. Gaynon PS, Winick NJ, et al. Improved survival
9. Greaves MF, Colman SM, Beard ME, Bradstock for children and adolescents with acute
K, Cabrera ME, Chen PM, et al. Geographical lymphoblastic leukemia between 1990 and
distribution of acute lymphoblastic leukemia 2005: a report from the children's oncology
subtypes: second report of the collaborative group. J Clin Oncol 2012;30(14):1663-9.
group study. Leukemia 1993;7:27–34. 25. Baccichet A, Benachenhov N, Couture F, Leclere
10. Varghese PR, Elayidom NB, Joseph CD, Kumar S. JM, and Sinnnett D. Microsatellite instability in
Epidemiological observations on leukemia in childhood T cell ALL. Leukemia 1997; 11:797-
Kerala (A study of 1016 cases over three years). 802.
Ind J Haematol 1984;2:15-17. 26. Moorman AV, Richards SM, Martineau M,
11. Advani SH, Jussawalla DJ, Nagraj RD, Cheung KL, Robinson HM, Jalali GR, et al.
Gangadharan P, Shetty P. A study of 1126 Outcome heterogeneity in childhood high-
leukemia cases--epidemiologic and end-result hyperdiploid acute lymphoblastic leukemia.
analysis. Ind J Cancer 1979;16:8-17. Blood 2003;102(8):2756-62.
12. Pradhan PK, Tiwari SK, Dabke AT, Agarwal S. 27. Jourdan-Da Silva N, Perel Y, Mechinaud F,
Pattern of 1eukemias in Raipur (Madhya Plouvier E, Gandemer V, Lutz P, et al. Infectious
Pradesh-an analysis of 162 cases). Ind J Cancer diseases in the first year of life, perinatal
1982;19:20-3. characteristics and childhood acute
13. Chatterjea JB, Ghose S, Ray RN. Incidence of leukemia. Br J Cancer 2004;90:139–45.
leukemia. (An analysis of 544 cases studied in 28. Roman E, Simpson J, Ansell P. Childhood acute
Calcutta). J Assoc Phys Ind 1962;10:673-6. lymphoblastic leukemia and infections in the
14. Rani S, Beohar PC, Mohanty TK, Mathur MD. first year of life: a report from the United
Leukemia pattern in Delhi-a 10 year study of Kingdom Childhood Cancer Study. Am J
490 cases. Ind J Cancer 1962; 19:81-6. Epidemiol 2007; 165: 496-04.
15. Kumar A, Rathee R, Vashist M, Neelkamal, Singh 29. Knudson AG. Chasing the cancer demon. Ann
S, Gupta S. Acute Lymphocytic Leukemia: An Rev Genet 2000;34:1–19.
epidemiological and hematological study from 30. Besson C, Gonin C, Brebion A, Delaunay C,
Haryana. Biosciences Biotechnology Research Panelatti G, Plumelle Y. Incidence of
Asia 2012;9(2):813-17. hematologic malignancies in Martinique,
16. Gurney JG, Severson RK, Davis S, Robison LL. French West Indies, over representation of
Incidence of cancer in children in the United multiple myeloma and adult T cell leukemia /
States. Sex-, race-, and 1-year age-specific rates lymphoma. Leukemia 2001;15:828–31.
by histologic type.Cancer1995;75:2186-95. 31. Yanada M, Jinnai I, Takeuchi J, Ueda T,
17. Hanson MR, Mulvihill JJ. Epidemiology of child- Miyawaki S, Tsuzuki M, et al. Clinical features
blood cancer In Levine AS, ed Cancer in the and outcome of T-lineage acute lymphoblastic
Young. NewYork: Masson; 1980.p.3-12. leukemia in adults: A low initial white blood
18. Draper GJ, Kroll ME, Stiller CA. Childhood cell count, as well as a high count predict
Cancer. Cancer Surv 1994;307:493-517. decreased survival rates. Leukemia Research
19. Gurney JG, Daris S, Severson RF, Fang JY, Ross 2007;31(7):907-14.
JA, Robinson LL. Trends in Cancer incidence 32. Miller RW. Special susceptibility of the child to
among children in US. Cancer 1996;78:532-41. certain radiation- induced cancer.
20. Holmes L, Hossain J, Vignes-Kendrick M, Opara Environmental Health prospective
F. Sex variability in pediatric leukemia survival: 1995;103(6):41-4.
Large cohort evidence. ISRN Oncology 2012; 1- 33. Greaves M. In utero origins of childhood
9. leukemia. Early Hum Dev 2005;81:123-29.
34. Schmitz-Feuerhake I, Dannheim B, Heimers A,
Oberheitmann B, Schroder H, Ziggel H.

Radha Rathee et.al, IJPRR 2013; 2(11) 34


International Journal of Pharma Research & Review, Nov 2013; 2(11):27-36

Leukemia in the proximity of a German boiling- International Journal of Biotechnology and


water nuclear reactor: evidence of population Biosciences 2012;2(4):272-75.
exposure by chromosome studies and 51. Taub JW, Konrad MA, Ge Y, Naber JM, Scott JS,
environmental radioactivity. Environmental Matherly LH, et al. High frequency of leukemic
Health Perspectives 1997;105(6):1499-504. clones in newborn screening blood samples of
35. Doll R, Wakeford R. Risk of childhood cancer children with B-precursor acute lymphoblastic
from fetal irradiation. Br J Radiol 1997;70:130– leukemia. Blood 2002;99:2992–6.
39. 52. Greaves MF, Maia AT, Wiemels JL, Ford AM.
36. Infante-Rivard C, Fortier I, Olson E. Markers of Leukemia in twins: lessons in natural history.
infection, breast-feeding and childhood acute Blood 2003;102:2321–33.
lymphoblastic leukemia. British Journal of 53. Jamieson CH, Ailles LE, Dylla SJ, Muijtjens M,
Cancer 2000;83:1559–64. Jones C, et al. Granulocyte-macrophage
37. Bartley K, Metayer C, Selvin S, Ducore J, Buffler progenitors as candidate leukemic stem cells in
P. Diagnostic X-rays and risk of childhood blast-crisis CML. N Engl J Med 2004;351: 657–
leukemia. International Journal of 67.
Epidemiology 2010;39(6):1628-37. 54. Greaves MF, Janossy G, Peto J. Immunological
38. Shu XO, Linet MS, Steinbuch M, Wen WQ, defined subclasses of acute lymphoblastic
Buckley JD, Neglia JP. Breast feeding and risk of leukemia in children. Relationship to
acute childhood leukemia. J Nat Cancer Inst presenting features and prognosis. Br J Haemat
1999; 91(20):1765–72. 1981;48:179-83.
39. Feychting M, Schulgen G, Olsen JH, and Ahlbom 55. Rajalekshmy KR, Abitha AR, Pramila R,
A. Magnetic field and childhood cancer- a Gnanasagar T, Maitreyan V, Shanta V.
analysis of two Scandinavian studies. European Immunophenotyping of acute lymphoblastic
Journal of Cancer 1995;31:2035-39. leukemia in Madras, India. Leuk Res
40. Wertheimer N, Savitz DA, and Leeper E. 1994;18:183-90.
Childhood cancer in relation to indicator of 56. Bene MC, Castoldi G, Knapp W. Proposals for
magnetic field from ground current sources. the immunological classification of acute
Bioelectrimagnetic 1995; 16: 86-96. leukemias. European Group for the
41. Belson M, Kingsley B, Holmes A. Risk factors for Immunological Characterization of Leukemias
acute leukemia in children: A review. (EGIL). Leukemia 1995;9(10): 1783-6.
Environmental Health Perspectives 57. Pui CH, Jeha S. New therapeutic strategies for
2007;115(1):138-45. the treatment of acute lymphoblastic leukemia.
42. Toledano SR, Lange BJ. Ataxia-telangiectasia Nat Rev Drug Discov 2007;6(2):149-65.
and acute lymphoblastic leukemia. Cancer 58. Bayram I, Erbey F, Kömür M, Kibar F, Tanyeli A.
1980;45:1675-8. Flow cytometry results at diagnosis and relapse
43. Shaw MP, Eden OB, Grace E, Ellis PM. Acute in childhood acute lymphoblastic leukemia.
lymphoblastic leukemia and Klinefelter’s Asian Pac J Cancer Prev 2010;11(5):1321-4.
syndrome. Pediatr Hematol Oncol 1992;9:81-5. 59. Topp MS, Kufer P, Gökbuget N, Goebeler M,
44. Mertens AC, Wen W, Davies SM. Congenital Klinge M, Neumann S, et al. Targeted Therapy
abnormalities in children with acute leukemia: With the T-Cell–Engaging Antibody
a report from the Children’s Cancer Group. J Blinatumomab of Chemotherapy-Refractory
Pediatr 1998;133:617-23. Minimal Residual Disease in B-Lineage Acute
45. Chessells JM, Harrison G, Richards SM. Down’s Lymphoblastic Leukemia Patients Results in
syndrome and acute lymphoblastic leukemia: High Response Rate and Prolonged Leukemia-
clinical features and response to treatment. Free Survival. Journal of Clinical Oncology
Arch Dis Child 2001;85:321-5. 2011;29:18-21.
46. Greaves MF. Etiology of acute leukemia. Lancet 60. Mitelman F. The Third International Workshop
1997;349: 344-9. on Chromosomes in Leukemia. Cancer Genet
47. Kinlen LJ. Infection and childhood leukemia. Cytogenet 1981; 4:96-8.
Cancer Causes and Control 1998;9:237–9. 61. Williams CK. Some biological and
48. Lombardi L, Newcomb EW, Dalla-Favera R. epidemiological characteristics of human
Pathogenesis of Burkitt lymphoma: expression leukemia in Africans. IARC Sci Publ 1984;4:687-
of an activated c-myc oncogene causes the 712.
tumorigenic conversion of EBV-infected human 62. Yunis JJ. Recurrent chromosomal defects are
B lymphoblasts. Cell 1987;49:161-70. found in most patients with acute
49. Gessain A, Renaud M, Guy T. Genetic variability nonlymphocytic leukemia. Cancer Genet
and molecular epidemiology of human and Cytogenet 1984;11:125-27.
simian T-cell leukemia / lymphoma virus type I. 63. Heim S, Bekassy AN, Garwicz S, Heldrup J,
Meladies Infectieuses 1996;13:132-45. Kristoffersson U, Mandahl N, et al. Bone
50. Kumar A, Rathee R, Vashist M, Neelkamal. marrow karyotypes in 94 children with acute
Dermatoglyphics in children of acute leukemia. Eur J Haematol 1990;44:227-29.
lymphocytic leukemia from Haryana.

Radha Rathee et.al, IJPRR 2013; 2(11) 35


International Journal of Pharma Research & Review, Nov 2013; 2(11):27-36

64. Third International Workshop on 77. Gustafsson G, Schmiegelow K, Forestier E,


Chromosomes in Leukemia (TIWCL). Clausen N, Glomstein A, Jonmundsson G, et al.
Chromosomal abnormalities and their clinical Improving outcome through two decades in
significance in acute lymphoblastic leukemia. childhood ALL in the Nordic countries: the
Cancer Res 1983; 43:868-70. impact of high-dose methotrexate in the
65. Fenaux P, Laï JL, Morel P, Nelken B, Taboureau reduction of CNS irradiation. Nordic Society of
O, Deminatti M, et al. Cytogenetics and their Pediatric Haematology and Oncology (NOPHO).
prognostic value in childhood and adult acute Leukemia 2000;14: 2267–75.
lymphoblastic leukemia (ALL) excluding L3. 78. Secker-Walker LM, Swansbury GJ, Hardisty RM,
Hematol Oncol 1989;7:307-10. Sallan SE, Garson OM, Sakurai M, et al.
66. Rieder H, Ludwig WD, Gassmann W, Thiel E, Cytogenetics of acute lymphoblastic leukemia
Löffler H, Hoelzer D, et al. Chromosomal in children as a factor in the prediction of long-
abnormalities in adult acute lymphoblastic term survival. Br J Haemato 1982;52:389-99.
leukemia: Results of the German ALL/AUL 79. Williams DL, Tsiatis A, Brodeur GM, Look AT,
study group. Recent Results Cancer Res Melvin SL, Bowman WP, et al. Prognostic
1993;131:3-7. importance of chromosome number in 136
67. Secker-Walker LM, Prentice HG, Durrant J. untreated children with acute lymphoblastic
Cytogenetics adds independent prognostic leukemia. Blood 1982;60:864.
information in adults with acute lymphoblastic 80. Bloomfield CD, Secker-Walker LM, Goldman A.
leukemia on MRC trial UKALL XA. Adult Six year follow-up of the clinical significance of
Leukemia Working Party. Br J Haematol karyotype in acute lymphoblastic leukemia.
1997;96:601–10. Cancer Genet Cytogenet 1989;40:171–85.
68. Faderl S, Garcia G, Thomas DA, Kantarjian 81. Groupe Français de Cytogénétique
HM. Philadelphia chromosome-positive acute Hématologique (GFCH). Cytogenetic
lymphoblastic leukemia—current concepts and abnormalities in adult acute lymphoblastic
future perspectives. Rev Clin Exp leukemia: Correlations with hematologic
Hematol 2002;6:142–60. findings and outcome. A collaborative study of
69. Charrin C, Thomas X, French M. A report from the Groupe Français de Cytogénétique
the LALA-94 and LALA-SA groups of Hématologique. Blood 1996;87:3135-8.
hypodiploidy with 30 to 39 chromosomes and 82. Wetzler M, Dodge RK, Mrózek K. Prospective
near-triploidy: 2 possible expressions of a sole karyotype analysis in adult acute lymphoblastic
entity conferring poor prognosis in adult acute leukemia: the cancer and leukemia Group B
lymphoblastic leukemia (ALL). Blood experience. Blood 1999; 93:3983-6.
2004;104:2444-51. 83. Moorman AV, Richards SM, Robinson
70. Mancini M, Scappaticci D, Cimino G, Nanni M, HM, Strefford JC, Gibson BE, Kinsey SE, et
Derme V, Elia L. A comprehensive genetic al. Prognosis of children with acute
classification of adult acute lymphoblastic lymphoblastic leukemia (ALL) and
leukemia (ALL): analysis of the GIMEMA 0496 intrachromosomal amplification of
protocol. Blood 2005;105:3434-41. chromosome 21 (iAMP21). Blood 2007;
71. Pui CH, Evans WE. Treatment of acute 109:2327–30.
lymphoblastic leukemia. N Engl J Med
2006;354:166–78.
72. Collier JAB. Oxford Handbook of Clinical
Specialties, Third Edition. Oxford;1991.p.810.
73. Margolin JF, Steuber CP, Poplack DG. Acute
lymphocytic leukemia. In: Principles and
Practice of Pediatric Oncology (Pizzo PA,
Poplack DG, eds). Philadelphia: JB Lippincott,
2001.p. 489–44.
74. Gökbuget N, Hoelzer D. Recent approaches in
acute lymphoblastic leukemia in adults. Rev
Clin Exp Hematol 2002;6(2):200-2.
75. Jaffe E, Harris N, Stein H, Vardiman J. Pathology
and Genetics of Tumours of Haematopoietic
and Lymphoid Tissues. IARC, Lyon. Oncologist
2001; 5(4):321-8.
76. Friedmann AM, Weinstein HJ. The role of
prognostic features in the treatment of
childhood acute lymphoblastic leukemia.
Hematol Oncol Clin North Am
2009;23(5):1033-42.

Radha Rathee et.al, IJPRR 2013; 2(11) 36

View publication stats

You might also like