throbber
Review
`
`Genetic Abnormalities and Challenges in
`the Treatment of Acute Myeloid Leukemia
`
`Genes & Cancer
`2(2) 95 –107
`© The Author(s) 2011
`Reprints and permission:
`sagepub.com/journalsPermissions.nav
`DOI: 10.1177/1947601911408076
`http://ganc.sagepub.com
`
`C. Chandra Kumar
`
`Submitted 01-Feb-2011; accepted 17-Mar-2011
`
`Abstract
`Acute myeloid leukemia (AML) is a hematopoietic disorder in which there are too many immature blood-forming cells accumulating in the bone marrow
`and interfering with the production of normal blood cells. It has long been recognized that AML is a clinically heterogeneous disease characterized by a
`multitude of chromosomal abnormalities and gene mutations, which translate to marked differences in responses and survival following chemotherapy.
`The cytogenetic and molecular genetic aberrations associated with AML are not mutually exclusive and often coexist in the leukemic cells. AML is a
`disease of the elderly, with a mean age of diagnosis of 70 years. Adverse cytogenetic abnormalities increase with age, and within each cytogenetic group,
`prognosis with standard treatment worsens with age. In the past 20 years, there has been little improvement in chemotherapeutic regimens and hence
`the overall survival for patients with AML. A huge unmet need exists for efficacious targeted therapies for elderly patients that are less toxic than available
`chemotherapy regimens. The multitude of chromosomal and genetic abnormalities makes the treatment of AML a challenging prospect. A detailed
`understanding of the molecular changes associated with the chromosomal and genetic abnormalities in AML is likely to provide a rationale for therapy
`design and biomarker development. This review summarizes the variety of cytogenetic and genetic changes observed in AML and gives an overview of
`the clinical status of new drugs in development.
`
`Keywords
`acute myeloid leukemia, genetic abnormalities, new drugs
`
`Introduction
`
`Acute myeloid leukemia (AML) is a clonal hematopoietic
`disorder resulting from genetic alterations in normal hema-
`topoietic stem cells. These alterations disrupt normal dif-
`ferentiation and/or cause excessive proliferation of
`abnormal immature leukemic cells known as blasts. As the
`disease progresses, blast cells accumulate in the bone mar-
`row, blood, and organs and interfere with the production of
`normal blood cells. This leads to fatal infection, bleeding,
`or organ infiltration in the absence of treatment within
`1 year of diagnosis.1-3 AML is characterized by more than
`20% blasts in bone marrow. AML can arise de novo or sec-
`ondarily either due to the progression of other diseases or
`due to treatment with cytotoxic agents (referred to as
`therapy-related AML). Up to 10% to 15% of patients with
`AML develop the disorder after treatment with cytotoxic
`chemotherapy (usually for a solid cancer). There are 2 main
`types of therapy-related AML. The “classic” alkylating-
`agent type has a latency period of 5 to 7 years and is often
`associated with abnormalities of chromosomes 5 and/or 7.4
`Exposure to agents, such as etoposide and teniposide, that
`inhibit the DNA repair enzyme topoisomerase II is associated
`with secondary AML with a shorter latency period, usually 1
`to 3 years, with rearrangements at chromosome 11q23.5
`Drugs, such as chloramphenicol, phenylbutazone, chloro-
`quine, and methoxypsoralen, can induce marrow damage
`that may later evolve into AML. Secondary AML may also
`
`occur because of progression of myelodysplastic syndrome
`(MDS) or chronic bone marrow stem cell disorders, such as
`polycythemia vera, chronic myeloid leukemia, primary
`thrombocytosis, or paroxysmal nocturnal hemoglobin-
`uria.6,7 Secondary AML has a particularly poor prognosis
`and is not considered to be curable, with the exception of
`secondary acute promyelocytic leukemia (APL).8 This is
`largely due to the high percentage of secondary AML asso-
`ciated with multidrug resistance (MDR) mechanisms: up to
`70% of secondary AML patients show overexpression of
`P-glycoprotein (Pgp) or other MDR mechanisms.9
`The genetic changes in leukemic blasts make them inef-
`fective at generating mature red blood cells, neutrophils,
`monocytes, and platelets. In addition, these AML blasts
`also inhibit normal blasts from differentiating into mature
`progeny. Inhibition does not result from “crowding out” of
`normal blasts; rather, inhibition may be mediated by vari-
`ous chemokines produced by AML blasts.10 AML pro-
`gresses rapidly and is typically fatal within weeks or months
`if left untreated. The most common cause of death in AML
`is bone marrow failure, and the principal sign of marrow
`failure is infection. Potential fatal organ infiltration, most
`
`Onconova Therapeutics Inc., Pennington, NJ, USA
`
`Corresponding Author:
`C. Chandra Kumar, Onconova Therapeutics Inc., 73 Route 31 North,
`Pennington, NJ 08534
`Email: ckumar@onconova.us
`
`Rigel Exhibit 1057
`Page 1 of 13
`
`

`

`96
`
`
`
`Genes & Cancer / vol 2 no 2 (2011)
`
`commonly involving the lung and the brain, becomes more
`likely as the disease progresses.
`AML is the most common acute leukemia affecting adults,
`and its incidence increases with age.1 Although the majority
`of patients under age 60 years achieve complete remission
`(CR) with traditional anthracycline- and cytarabine-based
`induction regimens, the long-term survival rates continue to
`be poor at approximately 30% to 40%.11-13 The prognosis is
`even poorer for those with high-risk AML, such as those
`who are older, those who had preceding MDS or myelopro-
`liferative disorders, or those with secondary AML from
`environmental exposures or prior chemotherapy. In such
`cases, CR is achieved in less than 40% of cases, with sur-
`vival rates of less than 10%.13 While 60% to 80% of younger
`patients achieve CR with standard therapy, only about 20%
`to 30% of the overall patient population has long-term dis-
`ease-free survival.3 Outcomes are worse for patients aged
`60 years or over, with CR rates in the range of 40% to 55%
`and poor long-term survival rates.3 Along with age, remis-
`sion rates and overall survival depend on a number of other
`factors, including cytogenetics, previous bone marrow dis-
`orders such as MDS, and comorbidities.3
`
`Epidemiology and Etiology of AML
`AML accounts for approximately 25% of all leukemias
`diagnosed in adults, and the median age at diagnosis is 67
`years.13,14 In the United States, 43,050 new cases of leuke-
`mia were reported in the year 2010, of which 12,330 were
`new cases of AML. There were 21,840 patients who died in
`the year 2010 because of leukemia, of which 8,950 were
`attributed to AML.15 The incidence of AML in the <65
`years’ age group is 1.8 cases per 100,000 patients, and the
`incidence in the >65 years’ age group is 17.9 cases per
`100,000 patients.15 The incidence of AML is expected to
`increase in the future in line with the aging population, and
`along with its precursor myelodysplasia, AML prevalence
`appears to be increasing, particularly in the population
`older than 60 years of age, and represents the most common
`type of acute leukemia in adults. Table 1 shows the inci-
`dence and prevalence of AML in the United States and
`other developed countries.
`Development of AML has been correlated with exposure
`to a variety of environmental agents, most likely due to
`links between exposure history and cytogenetic abnormali-
`ties. Radiation, benzene inhalation, alcohol use, smoking,
`dyes, and herbicide and pesticide exposure have all been
`implicated as potential risk factors for the development of
`AML.16,17 Survivors of the atomic bombs in Japan had an
`increased incidence of myeloid leukemias that peaked
`approximately 5 to 7 years following exposure.18 Therapeu-
`tic radiation also increases AML risk, particularly if given
`with alkylating agents such as cyclophosphamide, melpha-
`lan, and nitrogen mustard.
`
`Table 1. Number of Incidence and Prevalence Cases of Acute
`Myeloid Leukemia (AML) in the Major Pharmaceuticals Markets
`in 2010
`
`Markets
`
`US
`Europe
`Japan
`
`
`Incidence of
`new AML in 2010
`
`Prevalence of
`AML in 2010
`
`12,330
`12,923
`3,173
`28,426
`
`25,180
`22,790
`5,820
`53,790
`
`Note: Incident cases are the new cases diagnosed within a particular
`time frame; prevalent cases are all cases present at a particular time.
`Prevalence is thus a function of incident cases and duration of disease.
`
`Diagnosis and Classification of AML
`Demonstration of the accumulation of blasts resulting from
`the block in differentiation, characteristic of AML, is the
`essential requirement of diagnosis.19 The early signs of
`AML include fever, weakness and fatigue, loss of weight
`and appetite, and aches and pains in the bones or joints.
`Other signs of AML include tiny red spots in the skin, easy
`bruising and bleeding, frequent minor infections, and poor
`healing of minor cuts. The 2 systems commonly used in the
`classification of AML are the French-American-British
`(FAB) system and the World Health Organization (WHO)
`system. The FAB system is based on morphology and cyto-
`chemistry and recognizes 8 subtypes of AML, as shown in
`Table 2.20 In 1999, the WHO classification was introduced
`to include newer prognostic factors, such as molecular
`markers and chromosome translocations, and lowered the
`blast minimum criterion to 20%, thus including many cases
`classified as high-grade MDS in the FAB system.21 The
`WHO classification system identifies 4 AML subgroups: 1)
`AML with recurrent genetic abnormalities, 2) AML with
`multilineage dysplasia, 3) therapy-related AML and MDS,
`and 4) those that do not fall into any of these groups. This
`system created a minimum of 17 subclasses of AML, allow-
`ing physicians to identify subgroups of patients who might
`benefit from specific treatment strategies. Recently, a
`revised classification has been published as part of the
`fourth edition of the WHO monograph series.22 The aim of
`the revision was to incorporate new scientific and clinical
`information to refine diagnostic criteria for previously
`described neoplasms and to introduce newly recognized
`disease entities.
`
`Cytogenetic Abnormalities in AML
`AML is characterized by a high degree of heterogeneity
`with respect to chromosome abnormalities, gene muta-
`tions, and changes in expression of multiple genes and
`microRNAs. Cytogenetic abnormalities can be detected in
`approximately 50% to 60% of newly diagnosed AML
`
`Rigel Exhibit 1057
`Page 2 of 13
`
`

`

`Genetic changes and new drugs in the pipeline for AML / Kumar
`
`97
`
`Table 2. French-American-British (FAB) Classification of Acute Myeloid Leukemia (AML)
`
`FAB subtype
`
`AML-M0
`AML-M1
`AML-M2
`AML-M3
`AML-M4
`AML-M4 eos
`AML-M5
`AML-M6
`AML-M7
`
`Morphological classification
`
`% of all AML cases
`
`Undifferentiated acute myeloblastic leukemia
`Acute myeloblastic leukemia with minimal maturation
`Acute myeloblastic leukemia with maturation
`Acute promyelocytic leukemia
`Acute myelomonocytic leukemia
`Acute myelomonocytic leukemia with eosinophilia
`Acute monocytic leukemia
`Acute erythroid leukemia
`Acute megakaryoblastic leukemia
`
`5
`15
`25
`10
`20
`5
`10
`5
`5
`
`patients.23 The majority of AML cases are associated with
`nonrandom chromosomal translocations that often result in
`gene arrangements. Cytogenetics is the most important
`prognostic factor for predicting remission rate, relapse, and
`overall survival.23 Several chromosomal abnormalities
`such as monosomies or deletions of part or all of chromo-
`somes 5 or 7 (–5/–7 AML) and trisomy 8 are common in
`AML.24 The chromosomal abnormalities also include the
`long arm of chromosome 11 (11q); balanced translocations
`between chromosomes 15 and 17 (t(15;17)); chromosomes
`8 and 21 (t(8;21)); others such as (q22;q22), (q31;q22), and
`t(9;11); and inversion such as inv(16).25 Table 3 shows
`the most frequent chromosomal aberrations and their cor-
`responding fusion genes in AML. The translocation in
`t(15;17) is always associated with APL and leads to the
`expression of PML-RARα oncofusion gene in hematopoi-
`etic myeloid cells.26 Generally, patients with APL t(15;17)
`phenotype represent a unique group characterized by dis-
`tinct biological features and good prognosis, particularly
`when all-trans retinoic acid (ATRA) is used as part of remis-
`sion induction.
`Many of the gene rearrangements involve a locus encod-
`ing a transcriptional activator, leading to expression of a
`fusion protein that retains the DNA-binding motifs of the
`wild-type protein. Moreover, in many instances, the fusion
`partner is a transcriptional protein that is capable of inter-
`acting with a corepressor complex.27 A commonly accepted
`paradigm is that through aberrant recruitment of a corepres-
`sor to a locus of active transcription, the fusion protein
`alters expression of target genes necessary for myeloid
`development, thus laying the groundwork for leukemic
`transformation.28 Potential targeting of this interaction has
`become a major focus for the development of novel thera-
`peutics. ATRA serves as a prototype: by altering corepres-
`sor interaction with the APL fusion protein, ATRA
`effectively induces remission and has become a mainstay of
`treatment of this previously fatal disease.8 However, to
`date, APL represents both the most curable and the best-
`studied subtype of AML, while molecular data on other
`fusion proteins are limited or absent. Still, the work on
`
`Table 3. Acute Myeloid Leukemia (AML)–Associated Oncofusion
`Proteins
`
`Translocations
`
`Oncofusion protein
`
`Frequency of
`occurrence(% of AML)
`
`t(8;21)
`t(15;17)
`inv(16)
`der(11q23)
`t(9;22)
`t(6;9)
`t(1;22)
`t(8;16)
`t(7;11)
`t(12;22)
`inv(3)
`t(16;21)
`
`AML1-ETO
`PML-RARα
`CBF□-MYH11
`MLL-fusions
`BCR-ABL1
`DEK-CAN
`OTT-MAL
`MOZ-CBP
`NUP98-HOXA9
`MN1-TEL
`RPN1-EVI1
`FUS-ERG
`
`10%
`10%
`5%
`4%
`2%
`<1%
`<1%
`<1%
`<1%
`<1%
`<1%
`<1%
`
`PML-RARα has inspired the molecular analysis of many
`other AML-associated oncofusion proteins, especially
`AML1-ETO, CBFβ-MYH11, and MLL fusions.
`
`Oncofusion Proteins Associated with AML
`A total of 749 chromosomal aberrations have been cata-
`logued in AML.29 The frequencies of the 4 most common
`translocations are between 3% and 10%, while for others,
`the prevalence is significantly smaller. The most frequent
`oncofusion proteins, PML-RARα, AML1-ETO, CBFβ-
`MYH11, and MLL fusions, are described below.
`
`t(15;17), PML-RARα
`
`The t(15;17) translocation is found in approximately 95%
`of APLs, a specific subtype of AML. The translocation
`results in the expression of the PML-RARα oncofusion
`gene in hematopoietic myeloid cells.8 The PML-RARα
`oncofusion protein acts as a transcriptional repressor that
`interferes with gene expression programs involved in differ-
`entiation, apoptosis, and self-renewal.8
`
`Rigel Exhibit 1057
`Page 3 of 13
`
`

`

`98
`
`
`
`Genes & Cancer / vol 2 no 2 (2011)
`
`t(8;21), AML1-ETO
`
`Class I Mutations
`
`Approximately 10% of AML cases carry the t(8;21) trans-
`location, which involves the AML1 (RUNX1) and ETO
`genes, and express the resulting AML1-ETO fusion pro-
`tein. AML1 is a DNA-binding transcription factor crucial
`for hematopoietic differentiation,30,31 while ETO is a pro-
`tein harboring transcriptional repressor activities.32 The
`fusion protein AML1-ETO is suggested to function as a
`transcriptional repressor that blocks AML1-dependent
`transactivation in various promoter reporter assays, sug-
`gesting it may function as a dominant-negative regulator of
`wild-type AML1.33,34
`
`inv(16), CBFβ-MYH11
`
`inv(16) is found in approximately 8% of AML cases.
`inv(16) fuses the first 165 amino acids of core binding fac-
`tor β (CBFβ) to the C-terminal coiled-coil region of a
`smooth muscle myosin heavy chain (MYH11). CBFβ-
`MYH11 fusion protein is suggested to cooperate with
`AML1 to repress transcription.35,36
`
`11q23, MLL Rearrangements
`
`Mixed lineage leukemia (MLL) is implicated in at least
`10% of acute leukemias of various types. In general, the
`prognosis is poor for patients harboring MLL transloca-
`tions.37 In these patients, the MLL protein fuses to 1 of >50
`identified partner genes, resulting in an MLL fusion protein
`that acts as a potent oncogene.38 The amino-terminal por-
`tion of MLL serves as a targeting unit to direct MLL onco-
`protein complexes to their target loci through DNA binding,
`whereas the fusion partner portion serves as an effecter unit
`that causes sustained transactivation.
`
`Gene Mutations in AML
`Approximately 40% to 50% of patients with AML have a
`normal karyotype and represent the largest subset of AML.39
`All such cases of cytogenetically normal AML are currently
`categorized in the intermediate-risk group; yet, this group is
`quite heterogeneous, and not all patients in this subset have
`the same response to treatment. This is likely a result of the
`large variability in gene mutations and gene expression in
`this population. These alterations appear to fall into 2
`broadly defined complementation groups. One group (class
`I) comprises mutations that activate signal transduction
`pathways and thereby increase the proliferation or survival,
`or both, of hematopoietic progenitor cells. The other com-
`plementation group (class II) comprises mutations that
`affect transcription factors or components of the cell cycle
`machinery and cause impaired differentiation.
`
`Mutations in KIT, FLT3, and NRAS fall into the class I
`mutations.
`
`KIT mutations. Although patients with AML and inv(16)
`and t(8;21) in general have a more favorable prognosis,
`there remains a significant failure rate, and the long-term
`disease-free survival rate is approximately 60%. Studies
`have shown that activating KIT mutations in approximately
`30% to 40% of patients with inv(16) are associated with
`higher incidence of relapse and significantly lower survival.
`In those with t(8;21), the incidence of KIT mutations
`appears to be variable.40
`
`FLT3 mutations. Fms-like tyrosine kinase 3 (FLT3) is a
`receptor tyrosine kinase that plays a key role in cell sur-
`vival, proliferation, and differentiation of hematopoietic
`stem cells.41,42 It is frequently overexpressed in acute leuke-
`mias. FLT3 mutations occur in approximately 30% of AML
`patients and confer a poor prognosis. The 2 major types of
`mutations that occur are internal tandem duplication (ITD)
`mutations of the juxtamembrane region and point mutations
`in the tyrosine kinase domain (TKD), which frequently
`involve aspartic acid 835 of the kinase domain. Both muta-
`tions result in constitutive activation of the receptor’s tyro-
`sine kinase activity in the absence of ligand.41 The incidence
`of FLT3 mutations also increases with age, but the FLT3
`ITD mutations have less prognostic impact in patients >60
`years of age possibly because other adverse prognostic fac-
`tors are more prevalent.
`
`RAS mutations. Mutations in NRAS and KRAS occur in
`approximately 10% and 5% of AML patients, respectively.
`IRASS mutations occur only rarely in conjunction with
`FLT3 mutations and do not appear to have a significant
`impact on AML survival.43
`
`Class II Mutations
`
`In addition, mutations in MLL, brain and acute leukemia
`gene (BAAL), Wilms tumor gene (WT-1), CCAAT/
`enhancer-binding protein α (CEBPα), and nucleoplasmin 1
`(NPM1) have also been observed in AML patients.44-46
`Recently, mutations in DNA methyltransferase gene
`DNMT3A have been identified in one third of patients with
`de novo AML with intermediate-risk cytogenetics.47
`DNMT3A represents 1 of 3 human genes that encodes DNA
`methyltransferase that catalyzes the addition of methyl
`groups to cytosine within CpG dinucleotide, resulting in
`repression of nearby genes. Genomes with DNMT3A muta-
`tions commonly harbored additional mutations in FLT3,
`NPM1, and IDH1. The presence of any DNMT3A mutation,
`
`Rigel Exhibit 1057
`Page 4 of 13
`
`

`

`Genetic changes and new drugs in the pipeline for AML / Kumar
`
`99
`
`Table 4. Acute Myeloid Leukemia (AML) Cytogenetic Risk
`Groups
`
`Karyotype
`
`Frequency, %
`
`Complete
`remission, %
`
`Event-free
`survival, %
`
`Favorable
`t(8;21)
`inv(16)
`t(15;17)
`Intermediate
`Diploid, –Y
`Unfavorable
`−5/–7
`+8
`11q23, 20q-, other
`
`5-10
`5-10
`5-10
`
`40-50
`
`20-30
`10
`10-20
`
`90
`90
`80-90
`
`70-80
`
`50
`60
`60
`
`60-70
`60-70
`70
`
`30-40
`
`5-10
`10-20
`10
`
`either alone or in combination with FLT3 ITD mutation, is
`associated with significantly shorter overall survival (OS).47
`
`Prognostic Factors in AML
`Prognostic factors can be divided into those associated with
`treatment-related death occurring before response can be
`assessed and those associated with resistance to treatment.
`The predictor of treatment-related death is the patient’s per-
`formance status. Therapy-related AML or AML arising
`after MDS is usually more resistant to treatment than de
`novo AML.48 However, age and cytogenetics are the most
`important prognostic factors for predicting remission rate,
`relapse, and OS in AML. Risk stratification based on cyto-
`genetics divides patients into 3 main groups: patients with
`favorable, intermediate, and unfavorable cytogenetics
`depending on the presence or absence of specific chromo-
`somal abnormalities (Table 4). Studies have shown that the
`5-year survival rate was 55% for patients with favorable
`cytogenetics, 24% for patients with intermediate risk, and
`5% for patients with poor-risk cytogenetics.24 Adverse
`cytogenetic abnormalities increase with age, and within
`each cytogenetic group, prognosis with standard treatment
`worsens with age.3 A recent study demonstrated that the
`percentage of patients with unfavorable cytogenetics has
`been shown to increase from 35% in patients below 56
`years of age to 51% in patients over 75 years (Fig. 1).49
`
`Treatment of AML
`The primary objective of therapy for AML is to achieve and
`maintain CR. CR is defined as a marrow with less than 5%
`blasts, a neutrophil count greater than 1,000, and a platelet
`count greater than 100,000. CR is the only response that leads
`to a cure or at least an extension in survival. The probability of
`AML recurrence sharply declines to <10% after 3 years in
`CR.50 For the past 30 years, treatment of AML has consisted of
`the combination of an anthracycline, such as daunorubicin or
`
`Figure 1. Cytogenetic risk group by age group. Adapted with permission
`from Appelbaum FR, Gundacker H, Head DR, et al. Age and acute myeloid
`leukemia. Blood. 2006;107:3481-5.
`
`idarubicin, and cytarabine.51 Treatment of AML is divided into
`2 phases: 1) remission induction therapy (with possible postin-
`duction) and 2) postremission therapy.52 Generally, AML treat-
`ment includes at least one course of intensive induction
`chemotherapy followed by an additional course of intensive
`consolidation therapy and then maintenance therapy.
`
`Remission Induction Therapy
`In induction therapy, the goal is to achieve a marked reduc-
`tion in the number of malignant cells in order to establish
`normal hematopoiesis. A standard form of induction ther-
`apy consists of a standard dose of cytarabine (SDAraC,
`100-200 mg/m2), administered by continuous infusion for
`7 days and combined with an anthracycline administered
`intravenously for 3 days (referred to as 7 + 3 regimen).
`With standard induction regimens, remission is achieved in
`about 65% to 85% of younger patients but in less than 50%
`of patients over 60 years of age.2,53 This approach results in
`a long-term disease-free survival of approximately 30%,
`with treatment-related mortality of 5% to 10%. A number
`of studies have been conducted to improve the CR rate by
`use of alternative anthracyclines, incorporation of high-
`dose AraC (HDAraC), or addition of other agents such as
`etoposide, fludarabine, or cladribine. However, presently,
`there is no conclusive evidence to recommend one 7 + 3
`induction regimen over another. However, these studies
`clearly support the conclusion that further intensification
`of the induction regimen is not associated with an increased
`CR rate.
`In patients who fail to achieve CR following induction
`therapy, postinduction therapy is recommended. Postinduc-
`tion therapy with standard-dose cytarabine is recommended
`in patients who have received standard-dose cytarabine
`induction and have significant residual blasts.52 In other
`cases, postinduction therapy may consist of hematopoietic
`stem cell transplantation if a suitable donor can be found.
`
`Rigel Exhibit 1057
`Page 5 of 13
`
`

`

`100
`
`Postremission Therapy
`
`There are mainly 2 types of postremission therapy:
`
`1. Consolidation therapy is usually administered at
`doses approaching those used during induction.
`2. Maintenance therapy is usually defined as therapy
`less myelosuppressive than therapy used to pro-
`duce remission. Typically, patients receive the
`same regimen used during induction at approxi-
`mately monthly intervals for 4 to 12 months.
`
`Consolidation Therapy
`Although obtaining an initial remission is the first step in
`controlling the disease, it is important that patients continue
`with consolidation therapy to achieve a durable remission.
`Patients who do not receive consolidation therapy will
`relapse within 6 to 9 months.54,55 Consolidation therapy can
`consist of chemotherapy or hematopoietic stem cell trans-
`plantation (HSCT), and the choice of therapy is typically
`dependent on patient age, comorbidities, chance of recur-
`rence based on cytogenetics, and whether a patient has a
`suitable donor for HSCT.3 The use of HSCT is less common
`in patients aged over 60 years because of increased risks of
`transplant-related morbidity and mortality. Consolidation
`therapy comprises treatment with additional courses of
`intensive chemotherapy after the patient has achieved CR,
`usually with higher doses of the same drugs used during the
`induction period. High-dose AraC (2-3 g/m2) is now stan-
`dard consolidation therapy for patients aged <60 years of
`age. The median disease-free survival for patients who receive
`only the induction therapy is 4 to 8 months. However, 35%
`to 50% of adults aged <60 years who receive consolidation
`treatment survive 2 to 3 years.55 HSCT has a central role in
`the treatment of AML. However, because of the morbidity
`and mortality of the procedure, it tends to be used in patients
`who have a substantial risk of relapse.56 APL, a subtype of
`AML, is treated differently from other subtypes of AML;
`the vitamin A derivative ATRA (Vesanoid, Roche, Basel,
`Switzerland) can induce differentiation of leukemic promy-
`elocytes, resulting in high remission rates.8 Older patients
`are generally treated with lower intensity therapies such as
`subcutaneous cytarabine or hydroxyl urea in an attempt to
`minimize treatment-related mortality.
`
`Maintenance Therapy
`Maintenance therapy, which is considered less myelosuppres-
`sive than the induction and consolidation forms of treatment, is
`used in patients who have previously obtained CR. It is a strat-
`egy to further reduce the number of residual leukemic cells and
`prevent a relapse. Its role in the routine management of AML
`patients is controversial and depends mainly on the intensity of
`induction and consolidation therapies.52
`
`Genes & Cancer / vol 2 no 2 (2011)
`
`Table 5. Outcomes in Acute Myeloid Leukemia (AML) by Age
`Criteria
`
`Complete response, %
`Disease-free survival, %
`Early death, %
`Overall survival, %
`Median survival, mo
`
`Age <60 y
`
`Age >60 y
`
`70
`45
`10
`30
`24
`
`45
`20
`25
`10
`10
`
`Treatment of Relapsed and Refractory Disease
`
`Despite the substantial progress in the treatment of newly
`diagnosed AML, 20% to 40% of patients still do not achieve
`remission with standard induction chemotherapy, and 50% to
`70% of first CR patients are expected to relapse over 3 years.57
`The prognosis for patients with AML refractory to first-line
`treatment or in first or subsequent relapse is generally poor.
`The duration of first remission in relapsed patients is the most
`important prognostic factor correlating with the probability of
`second CR and survival.58 Patients who relapsed in less than 6
`months have a significantly poor prognosis compared to
`patients who relapsed after a first CR lasting >6 months.
`Treatment strategies for relapse are dependent on patient
`age.52 For patients less than 60 years old who have experi-
`enced an early (<6 months) relapse after induction chemo-
`therapy, the US National Comprehensive Cancer Network
`(NCCN) guidelines recommend participation in a clinical trial
`or HSCT.52 However, if patients have relapsed after a long
`(6 months or greater) remission, they can be retreated with a
`chemotherapy regimen or a development drug in the context
`of a clinical trial.52 The recommended option for patients aged
`60 years or older is participation in a clinical trial.52
`HSCT is the most commonly used treatment modality at
`relapse in patients aged below 60 years. In older patients,
`use of HSCT at relapse is rare, and single agents including
`azacitidine (Vidaza, Celgene, Summit, NJ), gemtuzumab
`ozogamicin (Mylotarg, Pfizer, New York City, NY), and
`hydroxyurea are most commonly used, although there is a
`lack of clear consensus over the optimum regimen.
`
`Age Is a Major Determinant of Survival
`Treatment recommendations for AML patients differ
`depending on whether patients are above or below 60
`years old.52 Table 5 shows the treatment outcomes based
`on age criteria. Survival in AML depends on age, with sig-
`nificantly lower survival rates reported for older adults.3
`Statistics from the Surveillance, Epidemiology and End
`Results (SEER) Program from 1996 to 2002 show 5-year
`survival rates of 34.4% for adults aged below 65 years and
`4.3% for those aged 65 years or older.54 While selected
`older patients can benefit from standard therapies, this
`group of patients experiences greater treatment-related
`
`Rigel Exhibit 1057
`Page 6 of 13
`
`

`

`Genetic changes and new drugs in the pipeline for AML / Kumar
`
`101
`
`Table 6. Therapeutic Strategies Being Investigated in the Treatment of Acute Myeloid Leukemia (AML)
`
`Therapeutic approach
`
`Epigenetic regulation
`
`Differentiation-inducing therapeutics
`
`Angiogenesis inhibition
`
`Inhibition of signaling pathways
`
`Modulation of drug resistance
`Modified traditional chemotherapeutics
`
`Immune therapy
`
`Examples
`
`Histone deacetylase inhibitors: vorinostat, panobinostat, belinostat
`DNA methyl transferase inhibitors: Vidaza, Dacogen
`Retinoid X receptor agonists
`Arsenic trioxide
`Inhibition of angiogenesis: Velcade
`Thalomid, Revlimid
`Tyrosine kinase inhibitors: midostaurin, lestaurtinib, sorafenib, KW-2449, AC220
`Cell cycle inhibitors: ON 01910.Na
`Farnesyl transferase inhibitors: Zarnestra, Sarasar
`mTOR inhibitors: Afinitor, PI-103, temsirolimus, GSK21110183
`PARP inhibitors: ABT-888
`MEK1/2 inhibitors: AZD6244, AS703026, PD98059, GSK1120212
`Bcl-2 inhibitors: oblimersen, obatoclax, ABT-263
`XIAP inhibitor: AEG-35156
`Aminopeptidase inhibitors (tosedostat)
`Valspodar, zosuquidar
`Nucleoside analogs: clofarabine, sapacitabine, elacytarabine
`Alkylating drugs: irofulven, Temodar, Onrigin
`Topoisomerase inhibitors: Hycamtin
`Antibodies: Mylotarg, lintuzumab, Avastin, T-cell targeted therapy
`
`toxicity, lower remission rates, shorter disease-free sur-
`vival, and shorter OS times.3 Older adults are less likely to
`achieve CR and to remain relapse free if they have
`achieved CR.3 In addition, these patients are more likely
`to experience treatment-related death, which is in the
`range of 15% to 30% in reported clinical trials.3 This is
`because patients over the age of 60 years are characterized
`by a higher prevalence of unfavorable cytogenetics and
`myelodysplasia, a greater incidence of MDR, and more
`frequent comorbidities that often make them unsuitable
`for intensive treatment.3
`
`Novel Agents in the Pipeline for AML
`Identification of specific gene mutations, chromosomal
`translocations, and alterations in signaling pathways and
`gene transcription in AML has led to the development of a
`number of targeted agents. A number of therapeutic
`approaches are being investigated in the treatment of AML
`(Table 6). These include histone deacetylase inhibitors,
`DNA methyl transferase inhibitors, retinoid X receptor
`agonists, proteosome inhibitors, antiangiogenesis inhibi-
`tors, FLT3 inhibitors, farnesyl transferase inhibitors,
`mTOR inhibitors, poly ADP-ribose polymerase (PARP)
`inhibitors, MEK1/2 inhibitors, modulators of drug resis-
`tance, and immune-modulating agents.59 In addition, a
`number of traditional chemotherapeutics in new formula-
`tions are also being investigated. Table 7 lists the mole-
`cules that are being investigated in late-stage clinical trials
`for AML. Clinical trial results of key drugs in AML are
`summarized below.
`
`Flt-3 Inhibitors
`Despite an exciting rationale for the use of FLT3 tyrosine
`kinase inhibitors (TKIs) in AML, the clinical results have
`so far been modest. Several FLT3 inhibitors are currently
`being developed suc

This document is available on Docket Alarm but you must sign up to view it.


Or .

Accessing this document will incur an additional charge of $.

After purchase, you can access this document again without charge.

Accept $ Charge
throbber

Still Working On It

This document is taking longer than usual to download. This can happen if we need to contact the court directly to obtain the document and their servers are running slowly.

Give it another minute or two to complete, and then try the refresh button.

throbber

A few More Minutes ... Still Working

It can take up to 5 minutes for us to download a document if the court servers are running slowly.

Thank you for your continued patience.

This document could not be displayed.

We could not find this document within its docket. Please go back to the docket page and check the link. If that does not work, go back to the docket and refresh it to pull the newest information.

Your account does not support viewing this document.

You need a Paid Account to view this document. Click here to change your account type.

Your account does not support viewing this document.

Set your membership status to view this document.

With a Docket Alarm membership, you'll get a whole lot more, including:

  • Up-to-date information for this case.
  • Email alerts whenever there is an update.
  • Full text search for other cases.
  • Get email alerts whenever a new case matches your search.

Become a Member

One Moment Please

The filing “” is large (MB) and is being downloaded.

Please refresh this page in a few minutes to see if the filing has been downloaded. The filing will also be emailed to you when the download completes.

Your document is on its way!

If you do not receive the document in five minutes, contact support at support@docketalarm.com.

Sealed Document

We are unable to display this document, it may be under a court ordered seal.

If you have proper credentials to access the file, you may proceed directly to the court's system using your government issued username and password.


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket