throbber
review
`
`How we treat older patients with acute myeloid leukaemia
`
`Avraham Frisch,1 Jacob M. Rowe1,2,3
`
`and Yishai Ofran1,3
`
`1Department of Hematology and Bone Marrow Transplantation, Rambam Health Care Campus, Haifa, 2Department of Hematology,
`Shaare Zedek Medical Center, Jerusalem, and 3The Ruth and Bruce Rappaport Faculty of Medicine, Technion, Israel Institute of
`Technology, Haifa, Israel
`
`Summary
`
`After decades when intensive chemotherapy remained the
`only effective anti-acute myeloid leukaemia (AML) treat-
`ment, a torrent of novel, less toxic agents are about to revo-
`lutionise AML therapy. Prolonged remissions with good
`quality of life become achievable for many patients previ-
`ously considered only for palliative care because they could
`not tolerate intensive therapy. As treatment options multiply,
`the importance of genetic profile is recognised, even for
`advanced-age patients for whom cure is unlikely. With lack
`of randomised comparative trials for most treatment regi-
`mens, one can only extrapolate data from existing studies to
`make evidence-based decisions. We herein present seven
`common clinical
`scenarios
`illustrating the complexity of
`treating older AML patients and describe our approach to
`their management. In each case, up-to-date data on relevant
`agents to be offered to a particular patient are discussed. The
`current review is limited to the drugs, available and approved
`in the Western world and many promising agents, still under
`investigation, are not discussed.
`
`Keywords: acute myeloid leukaemia, older adults, molecular
`profile, intensive chemotherapy, novel agents.
`
`Acute myeloid leukaemia (AML) is the most prevalent acute
`leukaemia in adults, with a median age at diagnosis of
`68 years. For decades, the main treatment option for newly-
`diagnosed AML patients has been intensive chemotherapy,
`which optimally offers a complete remission (CR) rate of
`70% and a long-term survival of about 40%.1 Specifically for
`patients with advanced age, not only is the remission rate
`significantly inferior, but also toxicity and treatment-related
`mortality are excessive compared to younger patients. Fortu-
`nately, in recent years, novel, effective and less toxic drugs
`
`Correspondence: Yishai Ofran, Department of Hematology and Bone
`
`Marrow Transplantation, Rambam Health Care Campus, 8 Ha’Aliya
`
`st., Haifa 3109601, Israel.
`
`E-mail: y_ofran@rambam.health.gov.il
`
`First published online 30 April 2020
`doi: 10.1111/bjh.16701
`
`CELGENE 2082
`APOTEX v. CELGENE
`IPR2023-00512
`ª 2020 British Society for Haematology and John Wiley & Sons Ltd
`British Journal of Haematology, 2020, 191, 682–691
`
`have become available. Moreover, improvements in support-
`ive care, and better tools for risk stratification and patient
`selection for allogeneic stem cell transplantation (allo-SCT),
`have all contributed to an improved outcome. Choosing
`wisely from the many treatment options available for AML
`patients with advanced age is currently a common clinical
`dilemma with no simple textbook answer. It is challenging to
`translate the accumulating but limited data regarding the
`effectiveness of each drug into a reliable, effective and evi-
`dence-based approach. We herein, with the use of six vign-
`ettes, present common treatment options and discuss a few
`case scenarios, aiming to apply principles that can be used in
`common clinical practice.
`
`Case 1: Is there still a role for intensive
`chemotherapy for elderly AML patients?
`
`A previously healthy 72-year-old woman is referred because
`of increased fatigue. Her complete blood count (CBC) shows
`a white blood cell (WBC) count of 1,300/ll with 6% blasts,
`32,000/ll platelets and haemoglobin of 6 5 g/dl. Her marrow
`is infiltrated with myeloid blasts, and molecular mutation
`analysis is negative for NPM1, FLT3-ITD mutations and for
`core binding factor (CBF) aberrations. Her echocardiogram
`demonstrates normal
`left systolic and diastolic functions
`without pulmonary hypertension or wall motion abnormali-
`ties. She has normal renal and liver functions. Cytogenetic
`results will be available within a week. What is the optimal
`approach for this patient? Should cytogenetics impact the
`chosen strategy?
`This is a case of an apparently fit patient who, had she
`been 10 years younger, would have been considered for
`intensive chemotherapy by most physicians. The question
`therefore
`is whether
`intermediate-
`and/or
`adverse-risk
`patients should be treated differently in an advanced age. To
`answer this question,
`it is prudent to consider both the
`immediate treatment plan and post-remission therapy. It has
`been well-known for decades that achieving CR in the end of
`induction is a prerequisite but
`insufficient condition for
`long-term remission. A substantial number of patients will
`relapse early after achieving CR, if no consolidation therapy
`
`

`

`is prescribed.2,3 Thus, an immediate/induction treatment
`plan should be such as to allow optimal post-remission ther-
`apy, particularly in fit patients, preserving the option for
`allo-SCT.
`Following intensive chemotherapy, fit patients aged 70–
`80 years with intermediate-risk AML have about 60–65%
`probability to achieve CR, with a 10–15% early death risk.1,4
`It should be noted that in the large prospective study by the
`Eastern Cooperative Oncology Group-American College of
`Radiology Imaging Network Cancer Research Group (ECOG-
`ACRIN, E2906) exploring intensive chemotherapy in patients
`with advanced age, minimal residual disease (MRD) negativ-
`ity was achieved in 58/147 (39 5%) of patients who achieved
`remission. These MRD-negative patients have an excellent
`long-term survival rate.5 Further improvement of intensive
`regimes in older patients is challenging. The addition of gem-
`tuzumab ozogamicin (GO) in induction, known to improve
`CR and survival in younger patients, is probably too toxic
`for patients over 70 years.6,7 As GO increases induction toxi-
`city by prolonging the time to bone marrow recovery8 and
`increasing early mortality,9
`its use in combination with
`intensive chemotherapy for older patients cannot be recom-
`mended.
`For long-term remission in fit older patients, allo-SCT
`remains the most effective post-remission strategy, with 38%
`2-year overall survival (OS) in CR1 for patients older than
`69 years and a 3-year OS of 49% for patients older than
`60 years.10,11
`A novel alternative for this patient, considered by some, is
`using venetoclax with hypomethylating agents (HMA) or
`low-dose cytarabine (LDAC; discussed later
`in case 2).
`Although data from phase Ib/II studies reported CR + CR
`with incomplete blood count recovery (CRi) rates as high as
`73% and low early mortality rates, early results from the
`phase III trial studying the LDAC and venetoclax combina-
`tion found a much lower response rate of 47% and a median
`OS of 7 2 months only. Furthermore, updated results of
`using an HMA + venetoclax combination in both NPM1 and
`IDH2 wildtype AML patients demonstrated a 2-year OS of
`26 6%.12 Of note, given SCT ineligibility of most patients
`included in the phase Ib/II trials, data about the outcome of
`patients fit to be transplanted are insufficient. Interestingly,
`in 21 patients (out of 145) initially treated with HMAs and
`venetoclax combination and then transplanted,13,14 the med-
`ian OS is reported to be as high as 24 4 months. However,
`the required depth of response and optimal timing for SCT
`after induction with a venetoclax combination still need to
`be assessed.
`Therefore, as long-term data are lacking, and in the
`absence of prospective
`comparisons between intensive
`chemotherapy and venetoclax combinations, it seems that for
`fit intermediate-risk AML patients, outcomes are better when
`intensive chemotherapy rather than venetoclax combinations
`is used.
`
`Treatment of Older Patients with AML
`
`Assuming that our patient has AML with adverse-risk
`cytogenetics would lead to complicated decision-making.
`Available data in patients with advanced age suggest that, for
`patients presenting with a monosomal/complex karyotype,
`the CR rates with intensive chemotherapy are as low as 30–
`35% and become even lower if TP53 is mutated/deleted.4,15,16
`Moreover, early death rates are higher, presumably because
`of prolonged cytopenia
`and the need for
`additional
`chemotherapy.
`Novel agents (e.g., venetoclax + azacytidine) offer at least
`the same CR with reduced early death rates. However, as dis-
`cussed earlier, to achieve a long-term outcome, the feasibility
`and efficacy of allo-SCT after these novel therapies need to
`be demonstrated in prospective studies.
`Older patients who are not candidates for allo-SCT but
`are in remission after chemotherapy are commonly offered at
`least one cycle of intermediate-dose cytarabine. The overall
`results, however, remain disappointing, with a relapse-free
`survival (RFS) of only 16% after 4 years.2
`Historically, maintenance therapy with various agents con-
`sistently failed to demonstrate OS advantage. Exceptions are
`three early studies showing an event-free survival (EFS) bene-
`fit with LDAC17,18 and with IL-2 and histamine combina-
`tion,19 but this has not been adopted in clinical practice,
`particularly in the U.S. Recently,
`the Haemato-Oncology
`Foundation for Adults in the Netherlands (HOVON) pub-
`lished a phase III trial where patients >60 years who received
`intensive chemotherapy and achieved remission were ran-
`domised to azacytidine maintenance or observation alone.
`This study reported an EFS benefit without OS prolonga-
`tion.20 Other studies established an OS benefit for azacytidine
`maintenance, but with some limitations. Oliva et al. pre-
`sented at the 23rd European Hematology Association Con-
`gress a phase III
`trial where azacytidine maintenance,
`preceded by intermediate-dose cytarabine consolidation, led
`to improved OS compared to non-maintenance.21 Notably,
`the number of patients participating in that study was rela-
`tively small. A phase II ECOG-ACRIN trial as an ancillary to
`the large prospective E2906 study included a randomisation
`for patients who achieved CR1 with intensive chemotherapy.
`One year of decitabine maintenance was shown to improve
`OS in the FLT3-ITD-negative subgroup.22 Finally, the most
`exciting breakthrough confirming the value of HMA mainte-
`nance therapy is in the results of the phase III randomised
`placebo-control QUAZAR AML-001 study. In this trial, CC-
`486, an oral formulation of azacytidine, was tested as mainte-
`nance therapy for both de novo and secondary AML patients
`aged 55 and older. The drug was given until relapse or allo-
`SCT, and was reported to double EFS and OS.23,24
`Our suggestion is to treat this patient with a curative
`intent, comprising intensive chemotherapy and, if possible,
`an allo-SCT. If a decision to consolidate her with chemother-
`apy is taken, at least 1 year of post-remission maintenance
`with HMAs should also be considered.
`
`ª 2020 British Society for Haematology and John Wiley & Sons Ltd
`British Journal of Haematology, 2020, 191, 682–691
`
`683
`
`

`

`Frisch et al.
`
`Case 2: Initial therapy in an unfit older patient
`
`An active 74-year-old male presents with AML. His ECOG
`performance status (PS) is 1. He is known to have type II
`diabetes and hypertension, a prior cerebrovascular accident
`(CVA) from which he recovered with no sequelae and
`chronic obstructive pulmonary disease (COPD) with moder-
`ate pulmonary hypertension. His WBC count is 60 000/ll,
`he has no disseminated intravascular coagulation (DIC) and
`no evidence of end-organ injury on admission. Two months
`previously, his blood counts were within normal limits. He
`has a normal karyotype and molecularly is FLT3-ITD-nega-
`tive/NPM1-positive. Is there a preferred targeted approach
`for this patient?
`Older patients with comorbidities have lower CR rates
`and higher mortality with intensive chemotherapy than
`younger patients. Assessing fitness for intensive induction is
`problematic and biased by considerable subjectivity; thus,
`published data need to be carefully scrutinised. In general,
`truly unfit patients have an early mortality rate of approxi-
`mately 30% with standard chemotherapy, as reported by
`real-world data from the Swedish registry.1
`Despite the good PS, his comorbidities, which can be eval-
`uated using
`either Hematopoietic Cell Transplantation
`Comorbidities Index or Charlson Comorbidities Index, are
`predictive of early mortality.25,26 Thus, treatment with inten-
`sive chemotherapy should to be discouraged.
`Few low-intensity options are currently approved for unfit
`newly-diagnosed AML patients. These regimens have not
`been directly compared;
`thus, recommending a particular
`regimen for this patient is based on a careful review of the
`available data.
`
`Venetoclax
`
`BCL2 superfamily proteins regulate mitochondrial apoptotic
`signalling, with BCL2, MCL1 and BCLXL proteins being the
`key anti-apoptotic members. Venetoclax is a potent oral
`specific BCL2-inhibitor which has only a modest activity
`against AML blasts as a single agent, possibly due to MCL1-
`dependent apoptosis evasion.27 However, based on phase I-II
`trials, the combination of venetoclax with either HMA or
`LDAC was approved by the FDA (though not the European
`Medicines Agency) for newly-diagnosed older or unfit AML
`patients.
`DiNardo et al.13,14 demonstrated that a combination of
`venetoclax with HMA in newly-diagnosed patients (median
`age 74 years) who were HMA-na€ıve induced CR/CRi rates of
`67%, compared with the historic rates of 20–25% CR/CRi
`for azacytidine or decitabine when used as monotherapy.28,29
`Moreover, high response rates were reported even in patients
`with poor cytogenetics or P53 mutations, with 60% and 47%
`CR/CRi rates, respectively. With a median follow-up of
`15 months,
`the median OS was not reached in NPM1-
`
`mutated AML patients. With the same duration of follow-
`up, the median OS was not reached if isocitrate dehydroge-
`nase (IDH) 1/2 was also mutated. Early mortality rates were
`as low as 3% in the first 30 days and 8% in 60 days. Inter-
`estingly, MRD-negativity was defined using a low-sensitivity
`3 and was reached in 30% of responsive
`threshold of 10
`patients. An optimal venetoclax dose, when prescribed in
`combination with HMA, was determined to be 400 mg/day,
`with no additive value for 800 mg/day and excessive toxicity
`in the 1200 mg/day dose.
`Another phase II trial examined the combination of LDAC
`(20 mg/day for 10 days) with 600 mg venetoclax. This study
`also included patients who failed previous treatment with
`HMA (about a third of the patients). The CR/CRi rates were
`26 and 28% respectively, but were dramatically influenced by
`presence of secondary AML or prior HMA treatment. For
`patients without prior HMA treatment
`the CR/CRi rates
`were 62%, and the median OS was 13 5 months. Of note,
`patients with NPM1 mutation had favourable response to
`this combination, with 89% CR/CRi rates.24
`Retrospective observations report encouraging results of
`venetoclax combinations even in the relapse setting. Studies
`demonstrate a response spectrum of 21–64%, emphasising
`the heterogeneity of
`these
`select
`reports, with higher
`responses in IDH1/2-mutated AML, intermediate-risk cytoge-
`netics and RUX1-mutated AML.30–32
`The kinetics of response in the newly-diagnosed AML and
`relapse settings are similar. The vast majority of the patients
`who respond to venetoclax-containing regimens do so in the
`two cycles.13,14,24,30 It
`is therefore rational
`to assess
`first
`response after two cycles.
`Venetoclax-containing regimens have a few important lim-
`itations. The major haematologic toxicity is neutropenia,
`which may often be profound. If severe neutropenia is evolv-
`ing while the patient
`is in remission, granulocyte-colony
`stimulating factor (GCSF) support provides a short and min-
`imal benefit;
`therefore, reducing or withholding doses is
`advised. Detailed dose modification varies and relies on min-
`imal data. Published recommendations include withholding
`venetoclax administration until neutrophil recovery and then
`resuming treatment in lower dose (50–75% full dose), reduc-
`ing the HMA/LDAC doses or extending intervals between
`treatment cycles.33 The two latter might be advised when
`marrow hypocellularity or pancytopenia is evident. Such rec-
`ommendations are based on clinical experience/observation
`of dose modifications that lead to peripheral count recovery
`in some patients. Experience in the practical use of veneto-
`clax is crucial, and precise data regarding the long-term effect
`of any modifications on survival and relapse are lacking.
`Venetoclax is metabolised via CYP3A4, and thus strong
`CYP3A4 inhibitors, commonly used for other indications in
`AML patients, may increase venetoclax blood levels. Com-
`monly used azoles, particularly voriconazole and posacona-
`zole, and quinolone antibiotics may have a strong interaction
`
`684
`
`ª 2020 British Society for Haematology and John Wiley & Sons Ltd
`British Journal of Haematology, 2020, 191, 682–691
`
`

`

`with venetoclax. It is recommended to reduce the venetoclax
`dose by 75% when co-administering strong CYP3A4 inhibi-
`tors, like the aforementioned azoles, and by 50% for moder-
`ate CYP3A4 inhibitors.24,34 However, as previously noted,
`these recommendations are based on limited pharmacoki-
`netic data with no clinical evidence supporting the long-term
`effect of these dose modifications.
`A major limitation for venetoclax-containing regimens is
`venetoclax resistance, as lack of response to venetoclax-based
`treatment is usually associated with prolonged cytopenias.
`Moreover, as other options for first-line treatment are avail-
`able, prediction of response to venetoclax-based therapy will
`become very important. Venetoclax resistance is driven
`mainly by overexpression or mutations in MCL1 and BCL-
`XL, the two other anti-apoptotic proteins of the BCL-2 fam-
`ily.35 Recent studies suggested that high BCL2 expression or
`BCL2/MCL1 ratio can predict for good response to veneto-
`clax. It was also shown that HMA and chemotherapy sup-
`press MCL1
`expression
`and
`thus
`synergise with
`venetoclax.36,37 A few recently-published articles demonstrate
`that AML cells with monocytic differentiation respond poorly
`to venetoclax, possibly because of low BCL2/MCL1 ratio, and
`that CD14 can serve as a marker for venetoclax sensitivity.37–
`40 Other pathways/mechanisms which confer venetoclax
`resistance are currently being actively investigated, as are
`other venetoclax combinations, such as those with MEK
`inhibitor, MDM2 inhibitor and FLT3-ITD inhibitors.41–44
`
`Glasdegib
`
`Glasdegib is a specific inhibitor of Smoothened (SMO), a
`receptor regulating the hedgehog pathway. SMO inhibition
`in cell lines is found to reduce the percentage of G0 cells,
`especially in leukaemic stem cells, and to abrogate cytarabine
`resistance. In mouse model, SMO inhibition is demonstrated
`to attenuate leukaemia-initiation potential of AML cells, pre-
`sumably by targeting leukaemic stem cells.45 Based on these
`results, a phase II study was conducted, comparing LDAC to
`LDAC + glasdegib in AML patients and high-risk myelodys-
`plastic syndrome (MDS) patients. The glasdegib + LDAC
`combination was found to have an OS benefit
`in AML
`patients (median OS 8 8 months, compared to 4 9 months
`with LDAC alone), which was consistent in all cytogenetic
`risk groups, albeit less pronounced in high-risk cytogenetics.
`Despite the survival advantage, only 27% patients achieved
`CR/CRi. Notably, 17% of patients who received the combi-
`nation were previously treated with HMAs. Most adverse
`events more common with glasdegib were gastrointestinal
`symptoms, mostly grade I/II.46 Based on these results, glas-
`degib was approved by the FDA for relapsed/refractory
`patients over 75 years or those with comorbidities, and a
`phase III trial for newly-diagnosed patients combining glas-
`degib with either intensive chemotherapy or HMAs was initi-
`ated in 2018.
`
`Treatment of Older Patients with AML
`
`Our priority for this patient is a combination of veneto-
`clax with azacytidine. While LDAC is a perfectly reasonable
`option, we prefer the combination with HMA due to the
`lower dose of venetoclax, used with less risk of drug interac-
`tions.
`
`Case 3: Treatment of an unfit AML patient with
`IDH1/2 mutation
`
`A 77-year-old female presents with normal karyotype,
`NPM1wt/FLT3-ITD-negative/IDH1-positive de novo AML.
`She is known to have hypertension, treated with valsartan/
`amlodipine. She had a hip fracture 4 months ago, for which
`she underwent total hip replacement. She is assisted with a
`walker, and her ECOG PS is 2. On CBC, she is pancytopenic
`with WBC of 2500/ll, absolute neutrophil count (ANC) of
`1200/ll, 72 000/ll platelets, haemoglobin 7 9 g/dl. The
`patient is a widow, and her children live abroad. She is afraid
`that she would not be able to attend frequent visits to the
`clinic.
`Is a total ambulatory care possible in this patient?
`
`IDH1/2 inhibitors
`
`IDH1 and IDH2 mutations are present in about 20% of
`AML cases, predominantly in normal karyotype AML. The
`IDH1 inhibitor ivosidenib and the IDH2 inhibitor enasidenib
`will be discussed together, since their safety profiles and effi-
`cacies are very similar. In relapsed/refractory AML, each of
`these IDH inhibitors is effective as a single agent, with as
`much as 30% CR + CRi rates and a median survival of over
`1 year for CR/CRi responders. Also, many patients who
`achieved only partial response (PR) had prolonged survival
`and became transfusion-independent.47,48 Based on these
`results,
`the
`IDH inhibitors gained FDA approval
`for
`relapsed/refractory AML. Interestingly, patients with FLT3-
`ITD co-mutation and RAS co-mutations respond poorly to
`IDH inhibition. For newly-diagnosed older patients, both
`IDH inhibitors were tested in phase I studies as single agents.
`Ivosidenib treatment achieved 55% ORR and 42% CR/CRh
`rates, with more
`than 60% of
`responses
`lasting for
`12 months. Based on these data, ivosidenib was approved by
`the FDA for this indication.49 Enasidenib achieved 31%
`ORR, with median duration response not reached.50 Adverse
`effects with these drugs are minor. Leukocytosis associated
`with differentiation syndrome is unique but uncommon, and
`possible QT prolongation, particularly with co-administra-
`tion of CYP3A4 inhibitors such as azoles, may also occur.
`IDH inhibitors were also tested in combination with aza-
`cytidine, demonstrating relatively high CR rates of 50–57%
`and ORR of 68–78%. Grade 3–4 neutropenia occurred in
`about 30%, whereas grade 3–4 febrile neutropenia occurred
`in 12% of patients treated with enasidenib and 39% of
`patients treated with ivosedenib combinations.13,51
`
`ª 2020 British Society for Haematology and John Wiley & Sons Ltd
`British Journal of Haematology, 2020, 191, 682–691
`
`685
`
`

`

`Frisch et al.
`
`Given that IDH inhibitors are oral anti-leukaemic agents
`with a remarkable safety profile, this would easily be our
`preference for older unfit patients who clearly elect to avoid
`hospitalisations. The sustained response and easy administra-
`tion of single agent ivosidenib make this an exciting thera-
`peutic strategy. However, in cases where immobilisation is
`less of a problem, venetoclax combinations, very effective
`also in IDH1/2-mutated AML, would be offered to this
`patient, given the overall greater likelihood of achieving a
`longer disease-free interval.
`
`Case 4: Treating an older patient with FLT3-ITD-
`mutated AML
`
`A 75-year-old obese male, known to have diabetes and
`restrictive lung disease with hypoventilation syndrome, pre-
`sents to the emergency room with dyspnoea. On examina-
`tion, his O2 saturation is 86% in ambient air, and chest
`radiography demonstrates bilateral pulmonary infiltrates. His
`CBC shows a WBC count of 140 000/ll with 85% circulating
`monoblasts, 60 000/ll platelets and haemoglobin 8 4 g/dl.
`He is
`treated with supplement oxygen, broad-spectrum
`antibiotics and hydroxyurea 6 g/day. Two days later, his
`WBC is 25 000/ll and his lungs are clear. The molecular
`AML work-up comes back with the diagnosis of normal
`karyotype, NPM1 wt/FLT3-ITD-mutated (allelic ratio 1 5)
`AML.
`FLT3 is a transmembrane tyrosine kinase receptor, which,
`upon binding to its ligand, promotes proliferation and sur-
`vival of normal haematopoietic progenitors through various
`intracellular pathways. Mutations in FLT3, resulting in con-
`stitutive activation of FLT3, are among the most common in
`AML in younger patients and are less frequent in advanced
`age. These mutations can be sub-divided into mutations in
`the juxtamembrane domain (FLT3-ITD mutation) and the
`tyrosine kinase domain (typically D835 point mutation).52
`FLT3-ITD is a well-known adverse prognostic marker, and is
`associated with high proliferation index and reduced RFS
`and OS.53 The poor prognosis is more pronounced if high
`allelic ratio (AR) FL3-ITD mutation (defined as >0 5) is
`noted.54 Guidelines of the National Comprehensive Cancer
`Network and the European LeukemiaNet categorise AML
`with FLT3-ITD mutation as poor-risk AML, based on the
`high relapse risk after intensive chemotherapy, and recom-
`mend allo-SCT as the preferred post-remission treatment. As
`for FLT3-TKD mutations, their prognostic impact is more
`uncertain due to conflicting evidence,55–57 and therefore they
`are not considered currently as poor prognostic markers.
`FLT3-ITD-mutated AML patients treated with low-inten-
`sity regimens with no FLT3 inhibitors have poor responses.
`CR rates of 44% for venetoclax + LDAC and 33% for glas-
`degib + LDAC were reported, which were lower than results
`in patients with no FLT3 mutations. The OS was also infe-
`rior, with only one-third of patients treated with veneto-
`clax + LDAC being alive after a year. Similar results were
`
`686
`
`recently published, demonstrating 53 3% CR/CRi rates and
`median OS of 12 4 months for FLT3-ITD-mutated AML
`receiving venetoclax-based combinations.58 Thus,
`patients
`FLT3-ITD-mutated AML in older patients should be consid-
`ered separately with novel agents. FLT3 inhibitors have a
`substantial role in treatment of AML patients presenting with
`FLT3 mutations. Midosturin, a first-generation FLT3 inhibi-
`tor, was the first to be approved as first-line therapy, in com-
`bination with 7 + 3 intensive chemotherapy, based on CR
`and survival benefit demonstrated in a prospective phase III
`study.59,60 Sorafenib, another first-generation FLT3 inhibitor,
`which is biologically active only against FLT3-ITD and not
`against TKD mutations, was also tested prospectively in com-
`bination with either intensive chemotherapy or azacytidine.
`Addition of sorafenib to chemotherapy during induction and
`consolidation, with use as a single agent in 1-year mainte-
`nance, was found to improve the CR and survival rates in
`FLT3-ITD-mutated young AML patients.61 The same survival
`benefit was demonstrated for FLT3-ITD-mutated AML
`patients aged 60–69, but not in a more elderly group, pre-
`sumably because of adverse effects.62 This is in line with a
`prior report regarding increased early mortality for patients
`>60 years treated with chemotherapy combined with sorafe-
`nib.63
`The combination of sorafenib and azacytidine was tested
`in a phase II trial for newly-diagnosed AML and relapsed/re-
`fractory patients and resulted in considerable response rates
`of 32% for the relapsed patients and 67% for newly-diag-
`nosed ones. Notably, about half of responses to sorafenib
`combined with azacytidine were CRi and not CR.64,65
`Next-generation FLT3 inhibitors are more specific and
`more potent than first-generation inhibitors. Quizartinib was
`tested against chemotherapy for relapsed/refractory FLT3-
`ITD + AML patients in a phase III
`trial, demonstrating
`improved OS with manageable
`toxicity. Over 20% of
`patients
`receiving quizartinib achieved only PR as best
`response considered an event, which might have contributed
`to a less-pronounced EFS advantage.66 Due to the very short
`EFS with this drug, the FDA has not approved it for use yet.
`Gilteritinib is another next-generation FLT3 inhibitor, which
`was tested as monotherapy against intensive and non-inten-
`sive salvage chemotherapy in a phase III trial. Based on
`improved OS in this setting, it was approved by the FDA for
`relapsed/refractory AML. Interestingly, only 21% of patients
`achieved CR/CRh, but nevertheless many others became
`transfusion-independent and/or could be referred to allo-
`SCT. It remains to be seen whether adding chemotherapy or
`a targeted agent to gilteritinib will improve the outcome in
`advanced FLT3-positive AML. These two next-generation
`inhibitors are currently being explored in various combina-
`tions in other scenarios,
`including newly-diagnosed AML
`patients.
`This patient has serious comorbidities that preclude him
`from standard chemotherapy. He also has a highly prolifera-
`tive AML with high allelic-ratio FLT3-ITD mutation, which
`
`ª 2020 British Society for Haematology and John Wiley & Sons Ltd
`British Journal of Haematology, 2020, 191, 682–691
`
`

`

`would argue for the addition of an FLT3 inhibitor. There are
`no good options here, and the therapeutic intent is limited.
`In the absence of a clinical trial and the lack of mature
`data using second-generation FLT3 inhibitors in newly-diag-
`nosed AML, our preferred choice would be to treat him
`with the combination of sorafenib and azacytidine. The
`importance of FLT3 inhibition should be appreciated.
`Recently, DiNardo et al. reported treatment failure in AML
`patients receiving venetoclax combinations, frequently attrib-
`uted to expansion of kinase bearing clones, with domination
`of FLT3 mutation.12 Interestingly, a recent report67 described
`the feasibility of adding an FLT3 inhibitor to venetoclax-
`based regimens, without additive toxicity and with improved
`outcomes, but the results are not mature enough to allow
`clear recommendation. In any event, the monocytic pheno-
`type described in this case may predict
`for venetoclax
`failure.
`
`Case 5: An older patient with treatment-related
`AML
`
`A 73-year-old active female (ECOG PS-1) is diagnosed with
`AML. Eight years ago, she had triple-negative breast cancer,
`for which she received 4 cycles of adriamycin and cyclophos-
`phamide followed by taxol
`for 12 successive weeks. After
`completion of neo-adjuvant therapy, she underwent mastec-
`tomy with negative sentinel lymph node biopsy. She has had
`routine annual oncologic surveillance with no evidence of
`disease. In the past year, she developed mild leukopenia and
`deteriorating anaemia, for which she did not undergo any
`investigation. She now presents with WBC of 5600/ll, 30%
`of which are circulating blasts, 23 000/ll platelets and hae-
`moglobin 6 1 g/dl. Molecular analysis is negative for NPM1,
`FLT3 and IDH mutations. Cytogenetic analysis reveals 47XX
`with trisomy 8. On geriatric evaluation, she is considered fit
`for intensive chemotherapy.
`For this fit patient, is 3 + 7 the best option?
`This case illustrates a fit patient with a secondary AML
`based on prior exposure to anthracyclines and alkylators.
`Her cumulative anthracycline dose is 240 mg/m2, which is
`low enough to allow further anthracycline exposure. The
`probability to achieve CR with intensive chemotherapy is
`estimated to be 50% at most in treatment-related AML4,68
`but with a 2-year OS of only 20%.4
`
`CPX351
`
`formulation of cytarabine and
`CPX351 is a liposomal
`daunorubicin at a fixed molar ratio of 1:5. A phase II trial
`comparing CPX351 to 7 + 3 demonstrated higher response
`rates, particularly more CRi without increased toxicity. In
`secondary AML patients,
`it also demonstrated a survival
`advantage.68 A successive phase III trial conducted in patients
`aged 60–75 years
`reported that
`therapy with CPX351
`achieved better outcomes in treatment-related AML patients,
`
`ª 2020 British Society for Haematology and John Wiley & Sons Ltd
`British Journal of Haematology, 2020, 191, 682–691
`
`Treatment of Older Patients with AML
`
`as well as in AML patients with antecedent MDS/chronic
`myelomonocytic leukaemia and AML with complex kary-
`otype. Toxicity profile of CPX351 is similar to that of inten-
`sive chemotherapy, but early mortality rate is reduced. Time
`will tell if CPX351 is indeed superior to 3 + 7, or if this
`advantage merely reflects optimised anthracycline/cytarabine
`ratio.69 In a prespecified subgroup analysis, two subgroups,
`including patients previously treated with HMAs and patients
`with FLT3-mutated AML, seemed to have no benefit from
`CPX351, the latter despite an acceptable CR rate.70 However,
`the standard deviation in these groups was wide and no
`definitive conclusions could be made. As FLT3 inhibitors
`were prohibited in that study, it is possible that, if used, the
`combination of CPX351 and FLT3 inhibitor would have the
`same advantage in FLT3-mutated AML patients. CPX351 is
`currently the only drug found to be better than standard IC
`for secondary AML patients in a phase III trial.
`Following cardiac evaluation with echocardiography, con-
`firming normal heart function, the patient can be offered
`treatment with CPX351. On

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