You are on page 1of 20

Cambridge University Press

978-0-521-19661-1 - Childhood Leukemias: Third Edition


Edited by Ching-Hon Pui
Excerpt
More information

Section 1
History and general issues
History of leukemia: historical perspectives
Chapter

1
Paul S. Gaynon, Toska J. Zomorodian, and Donald Pinkel
Introduction Recent clinical advances in Philadelphia chromosome-positive
11 12
The best way to study variable human disease in variable human beings is to
ALL and chronic myelogenous leukemia (CML) hint at
study variable human disease in variable human beings. the thrilling promise of coming decades.
1 This chapter focuses on the history of those leukemias that
Philip Schein and Barbara Scheffler (paraphrase)
affect young people, namely ALL, a predominately pediatric dis-
ease; AML, a disease that affects more adults than children; and
The last half century has witnessed striking progress in the treat- CML, which is found in young people only rarely.
ment of childhood leukemia, largely by empiric manipulation of a
rather limited chemotherapeutic armamentarium through well-
2 Emerging appreciation of leukemia biology
designed clinical studies. Approximately 50 years ago, the first
attempts to cure this dread disease were dismissed as futile and Current understanding of the molecular biology of leukemia is
even cruel in providing unrealistic hopes to desperate fam-ilies. provided in Chs. 7 and 8. T he object here is to trace its
However, improved supportive care has allowed patients to develop-ment over time.
survive through periods of perilous pancytopenia, induced by the While the first written description of cancer may be traced
disease and its treatment, and cure has become routine, albeit not back to Egypt in about 1600 BC, the first report of a patient with
universal, in economically privileged countries. Out-comes leukemia appeared only in 1827, about three and a half mil-lennia
13
remain poor for relapsed patients and for those in eco-nomically later (Table 1.1). Examination of blood cells was not possible
underprivileged countries where treatment may not be until the advent of the compound microscope (1665– 1673) by
3 13
accessible. Robert Hooke and especially by Anton van Leeuwen-hoeck.
Appreciation of the biology of leukemia has advanced from Jan Swammerdam and Joseph Lieutaud were the first to describe
13
clinical descriptions, through cytomorphology and histochem- red cells and white cells, in 1668 and 1749, respec-tively. About
istry, to molecular genetics. The pace has quickened percep-tively 20 years later, William Hewson described the lymphocyte and the
since the start of the twenty-first century. Genetic studies, at first 13
lymphatic system. In 1845, the category of illnesses now called
focused on the single most visually obvious chromo-somal leukemia was linked to white blood cells in three independent
abnormality by karyotype, are now expanded to genome-wide reports by Rudolf Virchow (Fig. 1.1), John Hughes Bennett, and
4,5 13
analyses and beyond genomics to epigenomics. We are moving David Craigie. Virchow coined the term “leukemia” in
from a descriptive catalogue of genetic abnormalities to an 13,28
1847. A French physician, Alfred Donne,´ had already
appreciation of cooperating functional aberrations affecting described patients with exceedingly high white blood counts with
differentiation, apoptosis, and proliferation. We not only divide maturation arrest and differentiated them from purulence in his
leukemia into major clinical/cytomorphologic subsets such as 1844 textbook, Cours de Microscopie compli-mentaire des
acute and chronic, lymphoid and myeloid but also recognize the Etudes medicales´. His textbook presented observa-tions he had
tremendous heterogeneity within each subset. Patients sharing a 29
made in extant correspondence 6 years earlier. However, credit
genetic abnormality, such as t(1;19), may have differing asso- for the first report of leukemia may best belong to a second
ciated abnormalities. Emerging evidence demonstrates oligo- 14
6,7 Frenchman, Alfred Velpeau, in 1827.
clonality and the consequences of clonal evolution. Some Henry Fuller was the f irst to diagnose leukemia by micro-
investigators feel that the limits of indiscriminate intensifica-tion 13
scopic examination of the blood in a living patient. In 1850, he
of therapy have been reached. However, improved event-free
described the first reported case of childhood leukemia, in a 9-
survival with intensified anthracycline treatment in acute myeloid
8,9
year-old girl. In a series of investigations, published in 1856,
leukemia (AML) and with “augmented” postinduc-tion 13
Virchow distinguished leukemia from leukocytosis and pro-
10
intensification in acute lymphoblastic leukemia (ALL) suggests posed his theory of its cellular origins – still fundamental to our
that our bag of tricks is not yet completely empty. understanding of the disease today. In 1868, Ernst Neumann
Childhood Leukemias, 3rd edn, ed. Ching-Hon Pui. Published by Cambridge University
Press. C Cambridge University Press 2012.

1
© in this web service Cambridge University Press www.cambridge.org
Cambridge University Press
978-0-521-19661-1 - Childhood Leukemias: Third Edition
Edited by Ching-Hon Pui
Excerpt
More information

Section 1: History and general issues


Table 1.1. Evolving insights and technology Fig. 1.1. Rudolf
Virchow, the father of
1600 BC First written description of cancer leukemia research,
1670 Examination of the blood with the compound microscope established leukemia as
a medical entity in the
1827 14
First clinical description of leukemia by Velpeau years 1845 and 1856.
1847 15
Term “leukemia” coined by Virchow
1872 Neumann concluded that leukemia is a disease of the bone
marrow
1877 16
Ehrlich introduced histochemical staining
1913 Acute and chronic, lymphoid and myeloid leukemias
1914 Boveri proposed that leukemia arises from a single cell through
17
chromosomal changes
1934 18
Flow cytometry
Nowell and Hungerford describe the
1960 Metaphase cytogenetics; 19
Philadelphia chromosome
1975 Monoclonal antibodies
20

1978 21
Host polymorphisms (thiopurine methyltransferase)
1980 Fluorescent in situ hybridization
22

1985 23
Polymerase chain reaction
1996 Gene expression arrays
24

1997 25
Comparative genomic hybridization
1998 Minimal residual disease by the polymerase chain reaction
26,27
fluorescence in situ hybridization and the polymerase chain
reaction (PCR) later supplemented karyotyping and identified
first reported the bone marrow changes in leukemia and by abnormalities in the leukemia cells of an increasing majority of
1872 concluded that leukemia was a disease of the bone patients. Conventionally, patients are classified by the most
marrow, but 20 years passed before his observations found visually obvious chromosomal abnormality. However, we now
widespread acceptance.
13,28
At the time, many believed that know that most patients with leukemia harbor multiple genetic
bone was a solid, impenetrable substance and could not accept abnormalities with losses outnumbering gains of genetic mate-
4
that blood cells might move back and forth between the rial. Epigenetic changes complement genetic changes. Event-
marrow and the peripheral blood. In 1876, Friedrich Mosler free survival varies not only among cytogenetic subsets but also
13,28 within each subset. Patients sharing a single abnormality such as
introduced the antemortem bone marrow puncture.
Introduction of histochemical staining methods by the t(1;19) may have differing associated abnormalities. The word
medical student and Nobel Laureate Paul Ehrlich in 1877 allowed “associated” is chosen rather than “secondary” because which
discrimination among leukocyte subsets. Ehrlich first identified a abnormalities are more important than others remains to be
primitive cell, which he described as the ancestor of the various elucidated. A single patient may harbor a variety of subclones
hematopoietic lineages. Leukemic marrow involvement was with overlapping but distinct, evolutionarily linked constella-
32
noted in the absence of peripheral blood involvement – so-called tions of cooperating abnormalities. The predominating clone at
16
aleukemic leukemia. The observa-tion that specific dyes relapse may differ from the predominant clone at diagnosis, being
affected specific cell types differently but reproducibly gave rise a direct descendant or sharing a common ancestor.
to the notion that chemicals might also have differential effects Flow cytometric studies examined DNA content and cell
on different types of cell and serve as treatment. In 1900, Otto membrane expression of lineage-associated membrane and
Nageli¨ identified the myeloblast and the lymphoblast. By 1913, cytoplasmic proteins, starting in the mid 1970s. Typical child-
leukemia was classified as ALL, AML, chronic lymphocytic hood early pre-B-ALL may be distinguished from pro-B-ALL,
13,28 typically seen in infants, and T-cell ALL, typically seen in
leukemia (CLL), and CML.
In 1914, Theodor Boveri suggested that cancer might arise older boys. Leukemias harboring translocation t(8;14)
from a single cell with genetic instability leading to chromo- (q11;q32) and associated translocations display a mature B-
somal changes, some too small to be seen by microscopy and cell immunophenotype.
17 Initial response to remission-induction treatment emerged as a
unresponsiveness to external growth regulation. Two World
Wars and a Great Depression intervened. consistent prognostic factor. Leukemia at levels too low for
Metaphase cytogenetics appeared in the early 1960s. Bet- microscopic recognition is termed minimal residual disease
30 26
ter banding techniques led to increased chromosome detail. (MRD). Quantitative PCR and flow cytometric examination
However, leukemic blasts are labile and metaphase spreads have supplemented and may replace morphologic evaluation of
31
may show only residual normal metaphases. Interphase peripheral blood and bone marrow aspirates for response
2
© in this web service Cambridge University Press www.cambridge.org
Cambridge University Press
978-0-521-19661-1 - Childhood Leukemias: Third Edition
Edited by Ching-Hon Pui
Excerpt
More information

Chapter 1: History of leukemia


33 Table 1.2. Emergence of conventional anti-leukemic drugs
evaluation. Measurement of MRD is a robust prognostic
34,35 1865 Lissauer administered potassium arsenite to a woman with
indi-cator. However, some patients with undetectable
14
MRD at remission may still relapse and some patients with chronic myelogenous leukemia
persistent MRD are cured, emphasizing the paramount 1895 Radiation therapy provides only transient benefit
importance of treatment after remission induction. 1930 Gloor reported a “cure” following arsenic trioxide, irradiation,
Host factors contribute to outcome. An intriguing report and transfusion, perhaps the first successful hematopoietic
36 13,28
links vincristine pharmacology and outcome. Polymor- stem cell transplant
phisms of thiopurine methyltransferase, first identified by R. 1943 52
Isolation of folic acid
21
Weinshilboum, nucleoside transporters MRP4 and 1948 Nitrogen mustard for Hodgkin disease
53

SLC29A1, and inosine triphosphate pyrophosphatase affect Antifols: aminopterin then methotrexate (amethopterin) for
37 54
mercaptop-urine metabolism. Genome-wide association acute lymphoblastic leukemia
studies are link-ing specific polymorphisms to variations in 1951 Adrenocorticotropic hormone then prednisone for acute
38 39 55
drug effects and the development of leukemia itself. lymphoblastic leukemia
17 1953 Mercaptopurine, methotrexate licensed by the FDA
Currently, we still follow Boveri and understand leukemia
1955 Prednisone licensed by FDA
as a phenotype resulting from heterogeneous constellations of
40 1958 Dexamethasone licensed by FDA
genetic – including microRNAs – and epigenetic changes. Dif-
ferences among leukemia gene expression profiles are said to 1959 Cyclophosphamide licensed by FDA
exceed differences between lung adenocarcinoma or melanoma 1963 Vincristine licensed by FDA
41,42 1969 Cytarabine licensed by FDA
and bladder cancer. In some cases (e.g., t(12;21)), but not in
others, (e.g., t(1;19)), an initial genetic change may arise prena- 1978 Native L-asparaginase licensed by FDA
43
tally. The heterogeneity in treatment outcomes within specific 1979 Daunorubicin licensed by FDA
genetic subtypes of leukemia may be related to differences in the 1983 Etoposide licensed by FDA
cooperative mutations or in host factors, currently poorly defined. 1987 Mitoxantrone licensed by FDA
In no genetic subset is cure impossible or assured.
1994 Pegylated L-asparaginase licensed by FDA
Leukemia is oligoclonal rather than clonal. Clonal evolution
ultimately results in overt leukemia in affected patients and evo- 1995 All-trans-retinoic acid approved for acute promyelocytic
lution continues after clinical presentation, resulting in relapse leukemia
43 2000 Arsenic trioxide licensed for acute promyelocytic leukemia by
and refractory disease in some cases. Relapse or refractory FDA
6
disease may arise from small, covert subclones. Treatment fail- 2001 Imatinib licensed for chronic myelogenous leukemia by FDA
ure may arise from increased blast proliferation or decreased cell 2004 Clofarabine licensed by FDA
44
death. Reaccumulation of blasts at relapse provides fur-ther 2005 Nelarabine licensed by FDA
opportunity for clonal evolution. However, the molecular
mechanisms of treatment failure – likely varied and possibly FDA, US Food and Drug Administration.
redundant – remain to be elucidated.
Interestingly, we are able to cure a large majority of 13
patients with rather undifferentiated therapies.
45–48
A variety blood groups, not corrected until 1900 by Karl Landsteiner. X-
of seem-ingly different regimens yield similar results with rays were discovered by William Rontgen¨ in 1895 and quickly
13
similar prog-nostic factors. Exceptions may be Burkitt applied to leukemia, resulting again in only transient benefit.
leukemia, often with a t(8;14) for which a brief fractionated Remissions, disappearance of leukemia with recovery of normal
alkylator therapy rather than a prolonged maintenance therapy hematopoiesis, remained rare anecdotes. The first likely cure was
49,50 reported by Gloor in an adult in 1930, following arsenic tri-oxide,
seems crucial, and now Philadelphia chromosome-
irradiation, and blood transfusion from two siblings, per-haps
positive ALL, where addition of a tyrosine kinase inhibitor to 13
11 presaging later bone marrow transplantation. In 1948, Bruce
cytotoxic chemotherapy is essential.
Wiseman commented that “a fresh point of view with respect to
13,28
this disease would not be undesirable.”
First treatments for leukemia Military research on mustard gas during World War I and II
Attempts to treat leukemia began soon after its recognition. In revealed its ef fects on the hematopoietic system, namely ane-mia,
1865, Heinrich Lissauer administered Fowler’s solution, a leukopenia, and thrombocytopenia. Lifting of wartime censorship
potassium bicarbonate-based solution of arsenic trioxide led to clinical trials in the USA and the UK that showed the
51
(potassium arsenite) to a woman with CML, achieving tem- benefit of methyl-bis-(β-chloroethyl)amine, a nitrogen mustard, in
porary benefit (Table 1.2). Use of arsenicals continued into the lymphoma, particularly Hodgkin disease, similar to that obtained
56 13
1930s, only to experience a recent rebirth as arsenic trioxide with radiotherapy. Some patients who had become resistant to
57
(As2O3). Blood transfusion was first applied to leukemia by irradiation responded to nitrogen mus-tard. Durable responses
Callender in London in 1873, also with only temporary bene- were more common in lymphoma than
13
fit. Transfusions were impeded by clotting and ignorance of
3
© in this web service Cambridge University Press www.cambridge.org
Cambridge University Press
978-0-521-19661-1 - Childhood Leukemias: Third Edition
Edited by Ching-Hon Pui
Excerpt
More information

Section 1: History and general issues


Fig. 1.2. Yellapragada Fig. 1.3. Sidney Farber
Subbarao developed a and his colleagues
method to synthesize discovered that a
folic acid and, with synthetic anti-folate, 4-
considerable input from amino-pteroylglutamic
Dr. Sidney Farber, he acid, produced
subsequently developed remissions of childhood
methotrexate to treat leukemia, leading to
leukemia. antimetabolite
chemotherapy and a
cure for many children
with leukemia.

Fig. 1.4 Gertrude Elion,


recipient of the Nobel rapidly dividing leukemia cells might require more folic acid
Prize in 1988, working 59,60
than normal cells.
with George Hitchings,
used innovative methods
Yellapragada Subbarao (Fig. 1.2) and colleagues provided
to develop a number of Sidney Farber (Fig. 1.3) and Louis Diamond with a num-ber of
drugs: mercaptopurine, anti-fols for clinical trials. A “def inite benef it” was obtained for
allopurinol, 59
10 of 16 patients with aminopterin. Five were reported in
pyrimethamine, 54
trimethoprim and detail. The response of the medical community and
acyclovir. particularly the pediatric housestaff, who interacted most closely
with these ultimately dying children and their fam-ilies, was
61
uniformly negative, despite the cautiously worded conclusions.
However, the results were confirmed by others. Amethopterin
(subsequently known as methotrexate), with an affinity for
dihydrofolate reductase 100 000 times greater than folic acid,
62
replaced aminopterin. The lympholytic effect of adrenal
63 64,65
corticosteroids was noted, first in mice and later in people.
Prednisone rapidly replaced adrenocorticotropic hormone. Then
6-mercaptopurine was synthesized, following from the
laboratory investigations of purines by Gertrude Elion (Fig. 1.4)
in leukemia. In the absence of better alternatives, investigators 66
and George Hitchings.
found reason for optimism and work continued to seek more
effective, less-toxic alkylating agents. Cyclophosphamide was
synthesized in 1959 and still remains in use.
13,28 Clinical trials methodology
Folic acid, a B vitamin, was recognized as crucial to Several novel, perhaps revolutionary, concepts contribute
13,28 greatly to progress:
hematopoiesis in 1943 and was chemically isolated in 1946.
Folate overcomes the maturation arrest in megaloblastic ane-mia prospective tests of candidate treatment interventions
and people speculated that folic acid might overcome the adherence to a written document, the experimental
maturation arrest in leukemia. In some experiments, folic acid
protocol
retarded cancer growth while in others it enhanced it. Heinle and
58 a prospective analysis plan
Welch observed that a folate-deficient diet caused a decrease in multicenter collaboration for adequate accrual
peripheral blast count. Sidney Farber at Harvard Medical School
concurrent controls through random treatment allocation
treated 90 patients with various malignancies with folic acid
54,59,60 complete remission rates and event-free survival as
analogues. An acceleration of leukemic growth was noted
surrogates for survival
in the bone marrows of treated children that had not been seen in
inclusion of all eligible patients in primary analyses:
untreated children, leading to a hypothesis that anti-folates
(antifols) might suppress leukemic growth as intent to treat.
4
© in this web service Cambridge University Press www.cambridge.org
Cambridge University Press
978-0-521-19661-1 - Childhood Leukemias: Third Edition
Edited by Ching-Hon Pui
Excerpt
More information

Chapter 1: History of leukemia


Leukemia treatment was dictated by rigorous adherence to a All eligible patients are included in primary analyses. Naive
written document, an experimental protocol. The idea seems intuition suggests that analyses might best be restricted to the
obvious today but in the 1950s protocols were often dismissed patients who actually receive the treatment under study. Intent to
as “cookbook” medicine. A researcher complained, “I can’t treat forbids exclusion of any eligible patients, including those
tell you what I will do this afternoon until I look at my mice. who receive therapy differing than that prescribed by protocol for
How can I tell you what I will do tomorrow?” By the 1970s, any reason. Some may understandably wonder how a patient who
Glidewell and Holland wrote, “A protocol was designed does not receive prescribed therapy can contribute to its
which specified in detail the application and evaluation of the assessment. However, rules for exclusion are rarely prospective
67
therapeutic pro-grams which were to be compared.” and provide an opportunity for bias. Prospectively, one cannot
The importance of statistics and collaboration were recog- predict whether a patient or physician will or will not toler-ate or
nized early. Analyses were guided by a prospective plan. Cancer comply with planned therapy and, therefore, the “intent to treat”
is fortunately rare in young people. Useful clinical trials require strategy provides the best, least biased estimate of efficacy.
substantial accrual for statistical power. Institutions and inves-
tigators banded together in a number of cooperative groups, such
as the Children’s Cancer Study Group, Cancer and Acute Supportive care
Leukemia Group B, and the Southwest Oncology Group. The In describing 40 years of therapy, Freireich commented: “In the
Pediatric Oncology Group emerged from the Southwest Oncol- early days when children were given methotrexate in a typi-cal
ogy Group and, in 2000, joined with the Children’s Cancer Group leukemia ward, there was not only blood on patients and on their
to form the Children’s Oncology Group. More tradi-tional 71
researchers, working alone or in small teams, scoffed at the beds but on the walls and on the windows.” During the 100
notion of innovation by committee and despaired at the challenge years between Virchow’s establishment of leukemia as an entity
of distributing intellectual or academic credit among the almost and the advent of alkylating agents, comforting the patient with
anonymous collaborating investigators. narcotics and human empathy was the first con-sideration. Today,
Identification of a historical or concurrent comparison or many patients are cured, but empathy and attention to symptoms
68 remain crucial to maximizing a child’s chance for recovery, by
control group is a critical step in clinical research. Gehan, aiding a distressed family to cope with a terrifying diagnosis, to
however, pointed out the critical contribution of non-randomized form a team with the medical person-nel, and to participate
trials in establishing new therapies. However, ran-domization, knowledgeably in the child’s sometimes challenging treatment
first employed in the negative studies of patulin for the common (Table 1.3).
69 70
cold and positive studies of streptomycin in tuberculosis,
provides concurrent controls for rigorous evaluation of candidate
therapies. Some hold randomization unethical, arguing that
Transfusion therapy
Credit for discovery of the circulation of the blood is gener-ally
physicians owe the patient their best treatment opinion, but most 89,90
accept that mistaken opinion is fre-quent enough to make given to William Harvey in 1616. Andrea Cesalpino, an
randomization an honest option. While an individual physician Italian, may in fact have preceded Harvey in this discov-ery. The
may hold a belief dearly, the uncer-tainty of knowledgeable concept of transfusion soon followed and several early and
colleagues provides adequate justifi-cation. Random allocation of usually unsuccessful attempts were documented in the mid
therapies with adequate patient numbers provides the strongest seventeenth century. Deaths led to bans in Paris, Rome, and
test of efficacy. However, pre-sentation of an option for London, which remained in place for 150 years. In 1828, Blun-
randomization may leave the sub-ject with exaggerated feelings dell, motivated by the plight of women with postpartum hem-
of uncertainty and of personal responsibility for the therapy orrhage, advocated human–human transfusion in place of the
more convenient animal–human transfusion and reported a
assigned. Historically, most ran-domized outcome trials show no
successful direct blood transfusion in a woman with postpar-tum
difference among treatment regimens. Gains, when realized, are 89,90
marginal –not all or none – and of ten at the expense of increased hemorrhage. However, severe unpredictable reactions
morbidity. discouraged further use. Landsteiner’s identification of human
73,91
Remission, disappearance of all microscopically detectable blood groups in 1901 enabled safer blood transfusion. Dur-
leukemia and recovery of normal hematopoiesis, was found to ing World War I, Rous and Turner discovered that a citrate dex-
add predictably to survival. James F. Holland, Emil Frei III, trose solution and cold would preserve red blood cells. Robert-
and colleagues in Acute Leukemia Group B noted that time in son, an American Army surgeon who had worked with Rous,
remission added to survival and proposed that duration of used this solution and packing boxes containing ice to preserve
remission might serve as a measure of treatment efficacy.
67 human red blood cells for prompt transfusion of wounded sol-
89
Today, remission-induction rate, event-free survival (i.e., the diers near the battle lines.
time from diagnosis to induction failure, first relapse, second For children with acute leukemia, the introduction of the
malignancy, or death), and disease-free survival (i.e., the time hospital blood bank in 1937 was the first step in prolonging their
74
from first remission to first relapse, second malignancy, or lives. By the late 1940s, blood transfusions together with the
death) remain useful determinants of treatment outcome. newly available antibacterial agents became generally accepted
5
© in this web service Cambridge University Press www.cambridge.org

You might also like