throbber
Signal Transduction and Targeted Therapy
`
`www.nature.com/sigtrans
`
`OPEN
`REVIEW ARTICLE
`Targeting multiple signaling pathways: the new approach
`to acute myeloid leukemia therapy
`Jenna L. Carter1,2, Katie Hege1, Jay Yang3,4, Hasini A. Kalpage 5, Yongwei Su3,4,6, Holly Edwards3,4, Maik Hüttemann5,
`Jeffrey W. Taub1,4,7,8 and Yubin Ge 1,3,4
`
`Acute myeloid leukemia (AML) is the most common form of acute leukemia in adults and the second most common form of acute
`leukemia in children. Despite this, very little improvement in survival rates has been achieved over the past few decades. This is
`partially due to the heterogeneity of AML and the need for more targeted therapeutics than the traditional cytotoxic
`chemotherapies that have been a mainstay in therapy for the past 50 years. In the past 20 years, research has been diversifying the
`approach to treating AML by investigating molecular pathways uniquely relevant to AML cell proliferation and survival. Here we
`review the development of novel therapeutics in targeting apoptosis, receptor tyrosine kinase (RTK) signaling, hedgehog (HH)
`pathway, mitochondrial function, DNA repair, and c-Myc signaling. There has been an impressive effort into better understanding
`the diversity of AML cell characteristics and here we highlight important preclinical studies that have supported therapeutic
`development and continue to promote new ways to target AML cells. In addition, we describe clinical investigations that have led
`to FDA approval of new targeted AML therapies and ongoing clinical trials of novel therapies targeting AML survival pathways. We
`also describe the complexity of targeting leukemia stem cells (LSCs) as an approach to addressing relapse and remission in AML
`and targetable pathways that are unique to LSC survival. This comprehensive review details what we currently understand about
`the signaling pathways that support AML cell survival and the exceptional ways in which we disrupt them.
`
`Signal Transduction and Targeted Therapy (2020)5:288
`
`; https://doi.org/10.1038/s41392-020-00361-x
`
`1234567890();,:
`
`INTRODUCTION
`Epidemiology of acute myeloid leukemia (AML)
`There are about 20,000 new cases of AML diagnosed each year in
`the United States.1,2 AML can affect people of all ages, but it is
`much more common in older adults with the age-adjusted
`incidence for those aged ≥65 years being 20.1 per 100,000 person-
`years compared with 2.0 per 100,000 person-years for those aged
`<65 years. The median age at diagnosis is 68 years and is most
`frequently diagnosed among people aged 65–74 years. Addition-
`ally,
`incidence is modestly increased in males compared with
`females and in Caucasians compared with other ethnic groups.3
`In most patients, the precise inciting event(s) leading to AML is
`unknown, but a genetic origin is strongly implicated. Environ-
`mental factors including exposure to chemicals such as benzene
`have also been associated with AML. Patients with a prior history
`of myelodysplastic syndromes (MDS) or myeloproliferative neo-
`plasms (MPN) and those who have previously received radiation
`and/or chemotherapy are also at increased risk of developing
`AML. Collectively, patients with AML with an antecedent myeloid
`disorder and those with therapy-related AML are considered to
`have secondary AML. In large population-based studies, 25% of
`AML cases are considered secondary and these are associated
`
`with lower rates of response to therapy and an inferior prognosis
`compared with de novo AML.4 AML can also be seen in patients
`with inherited genetic syndromes such as Fanconi’s anemia,
`Bloom syndrome, Down syndrome, and others.5–7 There has also
`been an increased interest in the study of inherited predisposi-
`tions to myeloid malignancies such as those seen with familial
`mutations of CEBPA, DDX4, and RUNX1.8
`Premalignant evidence of clonal hematopoiesis can be found in
`healthy individuals, as evidenced by acquired mutations in genes
`such as DNMT3A, TET2, and ASXL1.9 The prevalence of such clonal
`hematopoiesis of indeterminate potential (CHIP) increases with
`age and is associated with an increased risk of developing a
`subsequent hematological malignancy, particularly MDS or AML.
`However, the large majority of people who have CHIP never
`develop a hematologic cancer.10
`
`AML pathobiology
`transformation of myeloid
`AML results from the malignant
`precursor cells that are driven by a number of acquired genetic
`abnormalities. The biology is complex and involves a number of
`interdependent genetic mechanisms and pathways. The two-hit
`model of leukemogenesis hypothesizes that many cases of AML
`
`1Cancer Biology Graduate Program, Wayne State University School of Medicine, Detroit, MI, USA; 2MD/PhD Program, Wayne State University School of Medicine, Detroit, MI, USA;
`3Department of Oncology, Wayne State University School of Medicine, Detroit, MI, USA; 4Molecular Therapeutics Program, Barbara Ann Karmanos Cancer Institute, Wayne State
`University School of Medicine, Detroit, MI, USA; 5Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI, USA; 6National Engineering
`Laboratory for AIDS Vaccine, Key Laboratory for Molecular Enzymology and Engineering, The Ministry of Education, School of Life Sciences, Jilin University, Changchun, China;
`7Division of Pediatric Hematology/Oncology, Children’s Hospital of Michigan, Detroit, MI, USA and 8Department of Pediatrics, Wayne State University School of Medicine, Detroit,
`MI, USA
`Correspondence: Jeffrey W. Taub (jtaub@med.wayne.edu) or Yubin Ge (gey@karmanos.org)
`These authors contributed equally: Jenna L. Carter, Katie Hege
`
`Received: 24 July 2020 Revised: 21 September 2020 Accepted: 23 September 2020
`
`© The Author(s) 2020
`
`CELGENE 2071
`APOTEX v. CELGENE
`IPR2023-00512
`
`

`

`Targeting multiple signaling pathways: the new approach to acute myeloid. . .
`Carter et al.
`
`2
`
`are the result of the cooperation of two types of mutations:
`mutations that result in unhindered cell proliferation (class I
`mutations such as FMS-like tyrosine kinase 3 (FLT3), NRAS, c-KIT)
`and mutations that
`result
`in the arrest of normal myeloid
`differentiation (class II mutations such as RUNX1-RUNX1T1, CEBPA,
`TP53).11 Although this two-hit model
`is a simplification of the
`biology of AML, it serves as a useful conceptual framework.
`Cytogenetic abnormalities can be found in 50–60% of AML
`cases and strongly correlate with prognosis.12 For example, the
`core-binding factor (CBF) leukemias with the balanced transloca-
`tions t(8;21)(q22;q22), inv(16)(p13;q22), and t(16;16)(p13;q22) are
`associated with a favorable prognosis while AML with deletion 7,
`a monosomal karyotype, or a complex karyotype are associated
`with a dismal prognosis.13 The overwhelming majority of AML
`cases are also typified by genetic mutations such as NPM1, FLT3,
`IDH2 and TP53.14 Together
`isocitrate dehydrogenase 1 (IDH1),
`these acquired cytogenetic and molecular abnormalities encode
`transcription factors, tumor suppressors, DNA repair proteins,
`signaling molecules, regulators of apoptosis, and other diverse
`mechanisms, which promote leukemia development.
`AML appears to be maintained by a pool of self-renewing
`leukemia stem cells (LSCs) that are typically characterized by a
`CD34+CD38−CD123+ immunophenotype.15 AML stem cells are
`highly resistant to chemotherapy since they are primarily in the G0
`phase of the cell cycle and preferentially express multiple drug-
`resistant proteins such as P-glycoprotein and Bcl-2.16,17 The
`persistence of these LSCs predisposes to relapse even if bulk
`AML cells are effectively eliminated by treatment. As discussed
`below, there is great interest in therapies targeting LSCs in order
`to prevent relapse.
`Patients with AML usually present with manifestations of
`cytopenias due to bone marrow (BM) failure such as fatigue, fevers,
`infection, or bleeding. AML can sometimes present with signs and
`symptoms of hyperleukocytosis, which usually affects the pulmon-
`ary and central nervous systems. Extramedullary collections of
`leukemic blasts (e.g., myeloid sarcoma) is much less common but
`can occur in almost any organ, with skin (aka. leukemia cutis) and
`lymph node involvement being the most common.18 The diagnosis
`of AML is typically made by documenting ≥20% myeloblasts in a
`BM biopsy or aspirate specimen using adjunctive tests such as flow
`cytometry, cytogenetics, and fluorescence in situ hybridization to
`confirm and categorize the leukemia.18 Molecular profiling of AML
`is now considered mandatory in the era of precision medicine.
`There are a number of platforms available but next-generation
`sequencing panels focused on the most critical and common gene
`mutations are now routinely being employed. At a minimum, the
`European LeukemiaNet recommends testing for FLT3-ITD, FLT3-TKD,
`TP53, NPM1, RUNX1, ASXL1, and CEBPA mutations.19 One could also
`argue that screening for IDH1 and IDH2 mutations should be
`considered essential particularly at the time of relapse due to the
`availability of IDH1 and IDH2 inhibitors.
`
`AML classification
`The original FAB (French–American–British) classification of AML
`was the first attempt to systematically categorize this disease and
`(FAB M0–M7)
`divided AML into groups
`largely based on
`morphology and a few histochemical stains. The modern World
`Health Organization (WHO) classification is based on a combina-
`tion of morphology,
`immunophenotype, clinical characteristics,
`and genetics with the goal of identifying distinct biologic entities
`of AML with defined molecular pathways.20 The WHO classifica-
`tion recognizes six major categories of AML:
`(a) AML with
`recurrent genetic abnormalities; (b) AML with myelodysplasia-
`related features; (c) therapy-related AML and MDS; (d) AML, not
`otherwise specified;
`(e) myeloid sarcoma; and (f) myeloid
`proliferations related to Down syndrome.
`There are currently 11 genetic subtypes of AML recognized in
`the WHO classification including t(8;21)(q22;q22), inv(16)(p13;q22),
`
`t(16;16)(p13;q22), and several others. AML with the following gene
`mutations have also been included: NPM1, CEBPA (biallelic), BCR-
`ABL1, and RUNX1. AML with NPM1 or biallelic CEBPA mutations are
`considered favorable while AML with RUNX1 mutations are
`unfavorable.21,22
`Although AML with FLT3 mutation is not included in the WHO
`classification as a distinct entity, it is the most commonly (~30% of
`AML) mutated gene in AML and its presence predicts an
`unfavorable prognosis.23 FLT3 internal tandem duplication (FLT3-
`ITD) mutations result in a constitutively active FLT3, a transmem-
`brane tyrosine kinase, which in turns results in the growth and
`proliferation of leukemia cells.24 Because of its association with
`high rates of
`relapse, allogeneic hematopoietic stem cell
`transplant (SCT)
`is generally recommended in first remission.
`FLT3-ITD mutations are also an example of the complex interplay
`of genetic abnormalities seen in AML and their diverse effects on
`outcomes. Many of these mutations are often found in the same
`patient. NPM1 mutations can often co-exist with FLT3-ITD
`mutations resulting in a genotype with an intermediate-risk
`prognosis, depending on the FLT3 allelic ratio.25
`About 5–10% of AML patients have acute promyelocytic
`leukemia (APL) with PML-RARA fusion gene. This is characterized
`by a reciprocal translocation between chromosomes 15 and 17 (t
`(15;17)(q24;q21)) resulting in the production of a PML-RARA fusion
`gene. APL remains the paradigm of the genetic classification and
`treatment of AML given its disease-defining molecular signature
`and excellent outcomes with targeted therapies. APL is clinically
`characterized by disseminated intravascular coagulation and
`hyperfibrinolysis, which can result in a potentially fatal hemor-
`rhagic diathesis. However,
`if managed promptly and appropri-
`ately, the majority of patients are cured with treatment regimens
`that
`include a combination of
`targeted biologic therapies
`including all-trans retinoic acid and arsenic trioxide.26 Due to the
`unique characteristics of APL with PML-RARA fusion gene, this
`entity is not specifically covered in the remainder of this review.
`
`Treatment of AML
`The standard treatment for newly diagnosed AML remained static
`for many decades and was divided into induction therapy and
`consolidation therapy (Fig. 1). The goals of induction therapy are
`achievement of a complete morphologic remission, which results
`in the restoration of normal hematopoiesis and allows for
`subsequent therapy that maximizes the probability of long-term
`remission and potentially a cure.
`A combination of a daunorubicin and cytarabine was introduced
`approximately half a century ago and remained the standard
`therapy for most patients until very recently (Fig. 1). The most
`common iteration of
`this combination consists of 7 days of
`infusional cytarabine and 3 days of daunorubicin, the so-called
`“7+3” regimen. Remission rates are reported between 30 and 80%
`depending on patient and disease-related factors but long-term
`survivals and cure rates are appreciably lower due to relapses. This
`intensive chemotherapy approach is accompanied by a number of
`potential complications,
`including prolonged marrow aplasia,
`profound cytopenias, need for transfusional support, and risks of
`neutropenic infection and sepsis. Mortality rates during induction
`
`Fig. 1 History of AML therapies. Timeline of approved clinical
`therapies in the United States for the treatment of AML
`
`Signal Transduction and Targeted Therapy (2020)5:288
`
`

`

`range from 5% reported in clinical trials of younger patients to >20%
`reported in analyses of real-world data from the SEER database.27,28
`In patients who achieve a complete remission (CR), the cure rate
`approaches zero in the absence of some form of post-remission
`therapy. The antimetabolite cytarabine has been the mainstay of
`consolidation therapy for decades. A higher dose of cytarabine
`such as HIDAC improves survival in younger patients and in those
`who carry favorable cytogenetics.29 SCT is often used as post-
`remission therapy since its application is associated with the
`lowest rates of leukemia relapse. This benefit is balanced by risks
`of transplant-related mortality, including age, pre-existing comor-
`bidities, and other molecular lesions, and by risks of long-term
`complications in the form of graft-vs-disease. Retrospective
`analyses have clearly shown a survival benefit in favor of SCT in
`patients with AML who have adverse-risk genetic abnormalities
`(intermediate- or poor-risk AML).30 The absence of randomized
`data makes definitive conclusions in the intermediate-risk patient
`population less than clear-cut; however, most experts would favor
`SCT in eligible patients in the absence of contraindications.
`Older patients (>65 years) with AML have a much poorer
`prognosis compared with their younger counterparts primarily due
`to an increased prevalence of adverse-risk genetic features that
`promote resistance to chemotherapy.31 Intensive chemotherapy in
`this population is associated with lower rates of remission, increased
`toxicities, higher rates of early mortality, and dismal
`long-term
`survival rates in the absence of transplantation.31,32 For this reason,
`less intensive therapies such as single-agent hypomethylating
`agents (HMAs) and low-dose cytarabine (LDAC) have been used
`prior to the development and recent approval of novel agents.
`HMAs (azacitidine and decitabine) are commonly used in the
`United States for the first-line treatment of AML. However, their use
`as a single-agent therapy for AML has always been met with a lack
`of enthusiasm given the modest activity of these drugs. HMAs are
`associated with a CR rate of approximately 17.8 and 27.8% for
`decitabine and azacitidine, respectively, a duration of remission of
`10.4 months (azacitidine), and an overall survival (OS) of 7.7 and
`10.4 months for decitabine and azacitidine, respectively, compared
`to no responses in supportive care patients.33,34 Overall results with
`LDAC are likely to be worse, though it has improved OS and higher
`rates of supportive care in elderly adults who cannot receive
`conventional therapy.35,36 However patients with adverse cytoge-
`netics have no benefit in remission or survival on LDAC.36 Single-
`agent HMA use has not brought clear improvements in survival in
`AML patients and limited Food and Drug Administration (FDA)
`this use.33,34 Fortunately, novel drugs such as
`approval
`for
`venetoclax, an inhibitor of the antiapoptotic protein Bcl-2, appear
`to markedly potentiate the activity of HMA or LDAC making
`venetoclax-based combinations a more attractive option for older
`patients.37,38 HMAs are continued to be used in combination
`therapies and will be highlighted extensively in this review.
`Unlike acute lymphoblastic leukemia (ALL), there has been no
`role for maintenance therapy in AML. However, maintenance
`with an oral form of azacitidine, CC-486, was recently reported to
`improve survival compared with placebo in patients who
`completed standard 7+3 induction and cytarabine consolida-
`tion.39 This therapy was just recently (September 2020) approved
`for maintenance therapy in adults with AML in first remission
`and is likely to be considered standard of care for these patients
`(Fig. 1). In the subset of patients with FLT3 mutations, tyrosine
`kinase inhibitors (TKIs) are often used as posttransplantation
`maintenance despite the lack of regulatory approval. Addition-
`ally, there are several clinical trials investigating new potential
`AML maintenance therapies
`(NCT01546038, NCT03564821,
`NCT03881735, NCT03515512, NCT03728335, NCT03932643).
`
`AML clinical outcomes
`The OS of all AML patients is around 30% but it is quite dependent
`on age: long-term survivals for patients less than and older than
`
`Signal Transduction and Targeted Therapy (2020)5:288
`
`Targeting multiple signaling pathways: the new approach to acute myeloid. . .
`Carter et al.
`
`3
`
`65 years is about 50% and 10%, respectively.40 Survival rates in
`children are about 60–65%. Age acts as a surrogate for a panoply
`of patient and disease-related factors.41 Patient variables include
`performance status, comorbidities, and impaired organ function.
`Disease-related factors include clinical characteristics and biologi-
`cal features, such as cytogenetics and genetic mutations. Patients
`can be divided into adverse-,
`intermediate-, and favorable-risk
`groups based on cytogenetics and molecular features; however,
`much heterogeneity exists even within these subgroups.19 There
`has been a consistent and substantial improvement in the survival
`of younger patients over the past several decades despite the lack
`of drug approvals during this time period.40 The improvement is
`likely due to better supportive care measures and implementation
`and improvement of the risk-adapted use of transplantation.
`Significant improvements in the elderly AML population have
`been much harder to come by. The adverse-risk biology of AML in
`older adults is associated with increased drug resistance leading
`to early mortality, shorter remissions, and inferior survivals. This
`has led to a therapeutic nihilism in some treating physicians when
`faced with the prospect of treating older patients with either
`intensive chemotherapy, which is deemed too toxic, or less
`intensive therapies with less than desired effectiveness. Fortu-
`nately,
`recent drug approvals have improved the treatment
`landscape of AML, particularly in older patients, so that almost
`all newly diagnosed patients should be offered some form of
`therapy.
`
`Newly approved AML therapies
`For many years, treatment options for AML remained stagnant,
`with the standard 7+3 regimen of cytarabine and daunorubicin
`and SCT being the only option for patients (Fig. 1). The difficulty in
`developing new therapies for AML attributes mainly to myelo-
`suppression, which probably is the biggest hurdle overall
`in
`developing new drugs for this deadly disease. This is due to a
`significant amount of overlap in processes and signaling between
`AML cells and normal hematopoietic cells.42 Fortunately, the past
`few years have seen the development of more targeted therapies
`to better address the pathobiology and heterogeneity of AML and
`includes the FDA approval of midostaurin, gemtuzumab ozoga-
`micin, CPX-351, enasidenib, ivosidenib, gilteritinib, venetoclax, and
`glasdegib (Fig. 1). This surge in approved AML therapies has
`diversified the treatment options for patients and marks a turning
`point in how AML is approached.
`
`FLT3 inhibitors: midostaurin and gilteritinib. As mentioned, FLT3
`mutations are common in AML, occurring in approximately 30% of
`patients, and can be due to ITD mutations or point mutations in
`the tyrosine kinase domain (TKD).43 Constitutive activation of the
`mutant FLT3 supports tumorigenesis in hematopoietic precursor
`cells, so inhibition of FLT3 has been heavily pursued as a targeted
`therapeutic option for these patients.43 Midostaurin and gilter-
`itinib are type I FLT3 inhibitors and are effective against FLT3-ITD
`and FLT3-TKD.44 Midostaurin was approved by the FDA for
`therapy in adult patients with newly diagnosed FLT3-mutated AML
`in April 2017 and was followed by the approval of gilteritinib for
`the treatment of adult patients with relapsed/refractory (R/R) AML
`with FLT3 mutations in November 2018 (Fig. 1).45,46
`
`IDH1 and IDH2 inhibitors: enasidenib and ivosidenib. Mutations in
`IDH1 and IDH2 occur in about 20% of AML patients and are
`common in other malignancies, such as glioblastoma.47 These
`mutant enzymes have become promising targets for new
`therapies, as they promote tumorigenesis in cells through
`production of the oncometabolite, 2-hydroxyglutarate (2-HG).48
`Inhibition of the mutant IDH1 and IDH2 enzymes is able to reduce
`the production of 2-HG to normal physiologic levels, and this
`promotes the differentiation of leukemia cells.48 Enasidenib (AG-
`221), an IDH2 mutant inhibitor, and ivosidenib (AG-120), an IDH1
`
`

`

`Targeting multiple signaling pathways: the new approach to acute myeloid. . .
`Carter et al.
`
`4
`
`mutant inhibitor, were approved for treatment of R/R AML in
`August 2017 and July 2018, respectively (Fig. 1).49,50
`
`Bcl-2 inhibitor: venetoclax. AML has been shown to be dependent
`on dysregulations of the apoptotic pathway,
`including over-
`expression of Bcl-2, which is an important antiapoptotic protein.51
`This has supported the development of Bcl-2 inhibitors to
`promote the induction of apoptosis in AML cells and has led to
`the discovery of venetoclax, a potent and selective Bcl-2
`inhibitor.52,53 After being approved for use in chronic lymphocytic
`leukemia (CLL) (2016), demonstrating promising results in early
`clinical trials, and being well tolerated in older patients, venetoclax
`was FDA approved in November 2018 for the treatment of AML
`(Fig. 1).37,54,55 It was approved for use in combination with
`azacitidine or decitabine or LDAC for the treatment of newly
`diagnosed AML patients aged ≥75 years or who have comorbid-
`ities that preclude the use of intensive induction chemotherapy.
`Currently, venetoclax is being studied in numerous clinical trials in
`combination and single-agent therapies.
`
`Hedgehog (HH) pathway inhibition: glasdegib. Aberrant activation
`of the HH signaling pathway has been shown to be increased in
`AML cells and has also been correlated with worse prognosis
`and drug resistance in AML.56,57 Numerous studies have demon-
`strated that targeting this pathway showed antitumor activity and
`combination with current therapies improves efficacy.56 This led
`to the development of glasdegib, a HH pathway inhibitor that
`works by binding to and inhibiting the transmembrane protein
`Smoothened (SMO).58 Glasdegib was FDA approved in November
`2018 for use in combination with LDAC for the treatment of newly
`diagnosed AML patients aged ≥75 years or who have comorbid-
`ities that preclude the use of intensive induction chemotherapy
`(Fig. 1).59
`
`Antibody–drug conjugate (ADC): gemtuzumab ozogamicin. CD33
`has been found to be highly expressed on the leukemia cells of
`most AML patients and has developed as a targetable antigen.60
`Gemtuzumab ozogamicin is an ADC of a CD33-directed huma-
`nized monoclonal antibody (mAb) that is covalently linked to N-
`acetyl gamma calicheamicin (cytotoxic drug). The antibody
`portion localizes to CD33 antigens found on myeloid leukemia
`blasts and calicheamicin is internalized and induces double-strand
`breaks in DNA and cell death. Gemtuzumab ozogamicin was
`originally approved for monotherapy in CD33+ AML patients
`aged ≥60 years in May 2000 but was withdrawn from the market
`in 2010 due to concerns about toxicity. Additional studies using
`gemtuzumab ozogamicin at lower doses in combination with
`currently approved therapies confirmed its safety. This led to the
`approval of gemtuzumab ozogamicin in September 2017 for the
`treatment of CD33-positive AML in adults and pediatric patients
`aged ≥2 years (Fig. 1).61
`
`Cytotoxic therapy: CPX-351. CPX-351 is a liposomal formulation of
`cytarabine and daunorubicin, two standard of care chemotherapy
`drugs used in the treatment of AML. CPX-351 utilizes liposomal-
`encapsulated delivery system to avoid the first-pass metabolism,
`enhancing the pharmacodynamics and pharmacokinetics (PK) of the
`drugs and potentially leading to greater efficacy.62 Given its success
`in clinical trials in improving patient response rate and survival, CPX-
`351 was FDA approved in August 2017 for the treatment of adults
`with newly diagnosed AML with myelodysplasia-related changes or
`therapy-related AML (Fig. 1).62,63
`
`TARGETING SIGNALING PATHWAYS IN AML
`Apoptotic pathways
`Apoptosis is essential for ensuring the homeostasis of healthy
`tissue via two highly regulated pathways—the mitochondrial
`
`(intrinsic) pathway and death receptor (DR; extrinsic) pathway. The
`extrinsic pathway of apoptosis is initiated by DRs or tumor
`necrosis factor (TNF)
`family receptors, which are cell surface
`receptors that are activated by ligand interactions. Activation of
`these receptors by external stimuli results in the recruitment and
`activation of caspase 8 and ultimately leads to cell death. The
`intrinsic pathway is usually initiated in a cell-autonomous way
`when a cell undergoes stress and is unable to repair damages. The
`intrinsic apoptosis pathway is governed by the Bcl-2 family
`proteins, which consists of highly regulated proapoptotic and
`antiapoptotic proteins. Proapoptotic effectors Bak and Bax are
`necessary to initiate apoptosis in the cell and, when activated, will
`form pores on the outer membrane of mitochondria. This process
`is referred to as mitochondrial outer membrane permeabilization
`and results in release of cytochrome c, a proapoptotic factor, from
`the mitochondrial
`intermembrane space into the cytosol. Cyto-
`chrome c promotes assembly of the apoptosome and activation of
`caspase 9—ultimately leading to cell death. Antiapoptotic
`proteins (e.g., Bcl-2, Bcl-xL, and myeloid cell leukemia 1 (Mcl-1))
`bind, sequester, and inhibit proapoptotic proteins (Bak/Bax) to
`prevent apoptosis. BH3-only proteins (e.g., Bid, Bim, Bad, and
`Noxa) enhance apoptotic activity through activation of proapop-
`totic effectors or neutralization of antiapoptotic proteins. The
`imbalance of
`interactions between these proapoptotic and
`antiapoptotic proteins control the caspase cascade and cell death
`(Fig. 2a).64
`Apoptosis is frequently dysregulated in cancer. Cancer cells
`evade apoptosis through different mechanisms, but many involve
`overexpression of antiapoptotic proteins (Bcl-2, Mcl-1, and Bcl-xL)
`or loss of expression of proapoptotic proteins (Bak/Bax).65 These
`dysregulations promote cancer cell survival during treatment as
`many cancer therapies are dependent on intrinsic apoptosis
`induction to promote cancer cell death.51,65,66 It has been well
`established in preclinical studies that both Bcl-2 and Mcl-1 are
`frequently overexpressed in AML cells.67 Overexpression of Bcl-2
`and/or Mcl-1 is related to poor prognoses and are also associated
`with chemotherapy resistance. This understanding has led to the
`development of many therapeutics to target the dysregulated
`intrinsic apoptotic pathway for the treatment of AML, and here we
`summarize the targeting of Bcl-2 and Mcl-1.
`
`The antiapoptotic protein Bcl-2 was found to be
`Bcl-2 inhibitors.
`overexpressed and critical for leukemogenesis of AML.67 It is
`frequently overexpressed in leukemia progenitor and stem cells
`and in myeloid leukemia blasts in comparison to normal
`hematopoietic cells.68 True to its nature of preventing apoptosis,
`Bcl-2 has also been shown to play a role in resistance to
`chemotherapy of AML.69 Based on the importance of Bcl-2, many
`pharmaceutical companies have developed methods to target Bcl-
`2 for the treatment of AML.
`Oblimersen is a single-stranded anti-sense oligonucleotide
`(ASO) that is complementary to the first six codons of the open
`reading frame of the Bcl-2 mRNA sequence. The binding of
`oblimersen to the Bcl-2 mRNA targets the duplex for cleavage by
`RNase H and prevents translation into the Bcl-2 protein (Fig. 2c).70
`In preclinical studies, oblimersen was shown to decrease Bcl-2
`mRNA to nearly undetectable ranges and showed promising
`efficacy in in vivo leukemia xenograft models.70,71 Following the
`promising preclinical data, oblimersen was the first drug targeting
`Bcl-2 to be used for clinical trials in AML. A phase I trial combined
`oblimersen with standard of care chemotherapy (cytarabine and
`daunorubicin) and showed that 48% of patients achieved CR, with
`a median survival of 12.6 months. BM samples from patients who
`achieved CR had higher expression levels of Bcl-2 prior
`to
`induction therapy and showed significant decreases in Bcl-2
`mRNA following treatment. Oblimersen had no additional
`toxicities compared to the known standard chemotherapy
`toxicities and no cardiac toxicities were noted.72 A phase III
`
`Signal Transduction and Targeted Therapy (2020)5:288
`
`

`

`Targeting multiple signaling pathways: the new approach to acute myeloid. . .
`Carter et al.
`
`5
`
`Fig. 2 Targeting antiapoptotic proteins induces apoptosis in AML. a Antiapoptotic proteins Mcl-1 and Bcl-2 bind and sequester apoptotic
`effector proteins Bak/Bax to prevent Bak/Bax oligomerization and subsequent induction of apoptosis. b BH3 mimetics bind to the BH3-
`binding site of antiapoptotic proteins, Bcl-2 and Mcl-1, and release Bax/Bak to promote oligomerization and MOMP, which leads to
`subsequent induction of apoptosis. c Oblimersen is an anti-sense oligonucleotide that binds specifically to Bcl-2 mRNA to prevent Bcl-2
`translation and promote AML cell apoptosis. Selinexor (KPT330) inhibits XPO1 expression and subsequently decreases Mcl-1 stability to
`promote AML cell apoptosis. CDK9 inhibitors prevent the transcription of Mcl-1 gene to promote apoptosis of AML cells
`
`intensive induction and consolidation
`randomized trial of
`chemotherapy ± oblimersen was performed in 503 untreated
`older AML patients (>60 years).73 The study found that the
`addition of oblimersen to induction and consolidation therapy
`resulted in no significant improvement in CR rates, OS, or disease-
`free survival. These results limited further study of oblimersen in
`AML, but investigation into Bcl-2 inhibition continued.
`With a greater understanding of the intrinsic apoptotic pathway
`came a new method of targeting Bcl-2 through the development
`of “BH3 mimetic” small-molecule inhibitors. As mentioned above,
`BH3 only proteins (BIM, PUMA, BIC, NOXA, BID, etc.) activate
`apoptosis by binding to the BH3-binding site of antiapoptotic
`proteins and activating BAX/BAK to oligomerize in mitochondrial
`outer membrane. The identification of this function led to the
`development of small molecules that could mimic BH3 only
`proteins and have been thus termed BH3 mimetics (Fig. 2b).66 The
`effort to develop these small molecules have been immense and
`has now resulted in seven BH3 mimetics entering clinical trials and
`the FDA approval of the Bcl-2 inhibitor, venetoclax (Fig. 1).66
`The first BH3 mimetic tested in clinical trial of AML was
`obatoclax, which was
`thought
`to inhibit Bcl-2 and other
`antiapoptotic proteins (Bcl-xL, Bcl-w, Bcl-b, A1, and Mcl-1)
`(Fig. 2b).74 In preclinical studies, obatoclax was shown to inhibit
`cell growth and induce apoptosis in both AML cell
`lines and
`primary patient samples75 and was able to potentiate the
`cytotoxic effect of cytarabine in AML cells.76 A phase I study of
`obatoclax included patients with myelodysplasia or refractory
`leukemia, including AML, ALL, and CLL. This study looked at the
`efficacy of obatoclax via continuous intravenous (IV) infusion at
`increasing doses and frequencies.77 Obatoclax was well tolerated,
`but only one AML patient achieved CR and there was no
`improvement in disease progression. Another phase I/II trial of
`single agent obatoclax in untreated older patients (≥70 years) with
`AML was performed t

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