Next Article in Journal
Liver Stiffness-Based Risk Prediction Model for Hepatocellular Carcinoma in Patients with Nonalcoholic Fatty Liver Disease
Next Article in Special Issue
Targeted Therapies and Druggable Genetic Anomalies in Acute Myeloid Leukemia: From Diagnostic Tools to Therapeutic Interventions
Previous Article in Journal
Combination of a 15-SNP Polygenic Risk Score and Classical Risk Factors for the Prediction of Breast Cancer Risk in Cypriot Women
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Therapeutic Targeting of Acute Myeloid Leukemia by Gemtuzumab Ozogamicin

by
Michele Gottardi
1,†,
Giorgia Simonetti
2,†,
Alessandra Sperotto
3,
Davide Nappi
4,
Andrea Ghelli Luserna di Rorà
2,*,
Antonella Padella
2,
Marianna Norata
5,
Maria Benedetta Giannini
5,
Gerardo Musuraca
5,
Francesco Lanza
6,
Claudio Cerchione
5,‡ and
Giovanni Martinelli
7,‡
1
Onco Hematology, Department of Oncology, Veneto Institute of Oncology IOV, IRCCS, 31033 Padua, Italy
2
Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola (FC), Italy
3
Hematology and Transplant Center Unit, Dipartimento di Area Medica (DAME), Udine University Hospital, 33100 Udine, Italy
4
Department of Hematology and Cell Bone Marrow Transplantation (CBMT), Ospedale di Bolzano, 39100 Bolzano, Italy
5
Hematology Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola (FC), Italy
6
Hematology Unit & Romagna Transplant Network, Ravenna Hospital, 48121 Ravenna, Italy
7
Scientific Directorate, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola (FC), Italy
*
Author to whom correspondence should be addressed.
M.G. and G.S. equal contribution.
C.C. and G.M. equal contribution.
Cancers 2021, 13(18), 4566; https://doi.org/10.3390/cancers13184566
Submission received: 19 August 2021 / Revised: 8 September 2021 / Accepted: 10 September 2021 / Published: 11 September 2021

Abstract

:

Simple Summary

Gemtuzumab Ozogamicin (GO) is a drug approved for the treatment of acute myeloid leukemia (AML). It targets leukemic cells that express the CD33 molecule on their surface and brings the toxic agent calicheamicin inside the cell to kill it. Several studies have shown that AML patients can benefit of the addition of GO to chemotherapy during induction regimens, pre- and post-transplantation. Moreover, some disease features have been addressed or are under investigation for their capacity to predict response to GO, with the future aim of selecting AML patients that can mostly benefit of GO treatment.

Abstract

Acute myeloid leukemia (AML) is a complex hematological malignancy characterized by genetic and clinical heterogeneity and high mortality. Despite the recent introduction of novel pharmaceutical agents in hemato-oncology, few advancements have been made in AML for decades. In the last years, the therapeutic options have rapidly changed, with the approval of innovative compounds that provide new opportunities, together with new challenges for clinicians: among them, on 1 September, 2017 the Food and Drug Administration granted approval for Gemtuzumab Ozogamicin (GO) in combination with daunorubicin and cytarabine for the treatment of adult patients affected by newly diagnosed CD33+ AML. Benefits of GO-based regimens were also reported in the pre- and post-transplantation settings. Moreover, several biomarkers of GO response have been suggested, including expression of CD33 and multidrug resistance genes, cytogenetic and molecular profiles, minimal residual disease and stemness signatures. Among them, elevated CD33 expression on blast cells and non-adverse cytogenetic or molecular risk represent largely validated predictors of good response.

1. Introduction

Many patients affected by acute myeloid leukemia (AML) benefit of chemotherapy regimens and hematopoietic stem cell transplant (HSCT). However, progress has been modest in this therapeutic setting and huge challenges remain, mainly related to off-target cytotoxicities and to different chemoresistance mechanisms.
Immunotherapy is an innovative biological cancer therapy that exploits patient natural immune defenses to identify and eradicate cancer cells. Different types of immunotherapy have been developed, including antibody drug conjugates (ADCs) [1,2]. The advances of ADCs is to combine the specificity of a monoclonal antibody with the therapeutically benefits of chemotherapy agents [3]. Indeed, in contrast to conventional chemotherapeutics, ADCs provide superior efficacy and specificity while showing low risk of off-target cytotoxicity. Indeed, the chemotherapy particles associated with ADC remain inactive while passing through blood flow and become active only after ADC internalization. As a consequence, ADCs can increase the therapeutic window by reducing the Minimum Effective Dose (MED) along with enhancing the Maximum Tolerated Dose (MTD) [4,5,6].
Great interest in AML has been raised by the sialic acid-binding immunoglobulin-like lectin (Siglec) CD33 as a therapeutic target. Indeed, CD33 became an ideal target for the development of new ADCs due to the fact that its expression is common on the surface of AML blast cells and almost absent in normal pluripotent hematopoietic stem cells. Gemtuzumab ozogamicin (GO) is a humanized anti-CD33 monoclonal antibody covalently linked to various molecules of the cytotoxic agent N-acetyl gamma calicheamicin.
When GO binds CD33 antigen on the cell surface, the GO-CD33 complex is internalized and calichemicin molecules are released inside the cytoplasm. Active calichemicin particles intercalate DNA, thus inducing DNA damages, which, if left unrepaired, lead to cell cycle arrest and leukemic cell apoptosis. Different clinical trials have highlighted the benefit of GO on patient survival. GO is also the first antibody drug conjugate approved by the U.S. Food and Drug Administration (FDA) and the increasing knowledge of the GO metabolic pathway has improved our understanding on biomarkers of response.
In this review we summarize the clinical results obtained on CD33 targeting by GO in AML as single agent and in combination with chemotherapy, its potential benefit pre- and post-transplantation and we discuss the predictive biomarkers of therapy response.

2. CD33 Structure and Expression: Rationale for a Targeted Therapy in AML

CD33 is a 67 kDa glycosylated transmembrane protein belonging to the Siglec family [7]. The downstream pathway and biological functions of CD33 are still poorly understood, however Siglecs family members may regulate cytokine production, dampen inflammatory and immune responses, modulate intracellular calcium mobilization, cell adhesion, apoptosis of leukemic cells and myeloid cell maturation [8,9]. From a structural point of view, CD33 cytoplasmic tail contains two conserved immunoreceptor tyrosine-based inhibition motifs (ITIM and ITIM-like motifs) which, upon phosphorylation, promote the recruitment and activation of Src homology 2 domain-containing phosphatases 1 and 2 (SHP-1 and SHP-2). These activated SHPs further dephosphorylate various signaling molecules and suppress cellular activation [8]. The suppressor of cytokine signaling 3 (SOCS3) kinase competes with SHP-1 or SHP-2 for binding to the ITIMs. After the interaction with ITIM motifs, SOCS3 promotes the proteasome-dependent degradation of CD33, resulting in myeloid cell activation and proliferation [9].
Recently, different isoforms of CD33 gene have been identified [10]. Among them, the one missing exon 2 (CD33∆E2) has been predicted to have a clinical impact [11,12,13]. Briefly, this isoform lacks of the V-set domain containing the immune-dominant epitopes that represent the binding site of most CD33 antibodies, including GO.
In the healthy population, binding of antibodies recognizing the V-set domain of CD33 showed that CD33 is displayed on the surface of cells committed to the myeloid lineage, from myeloblasts to monocytes and also myelocytes and is down-regulated as the normal cells mature towards terminally-differentiated granulocytes, while it is retained on macrophages and dendritic cells [14,15]. However, in AML myeloid cells fail to differentiate, thus indicating that a high number of binding sites for CD33-specific agents are preserved and can be therapeutically exploited.

3. Gemtuzumab Ozogamicin: Mechanism of Cytotoxicity

Exploratory clinical studies indicated that the pharmacological inhibition of CD33 using unconjugated anti-CD33 antibodies have limited activity against AML cells.
From a structural point of view, GO is a recombinant humanized immunoglobulin G4 (IgG4) kappa (named P67.6) which specifically targets the CD33 antigen, linked to N-acetyl gamma calicheamicin, via the acid-labile hybrid 4-(4’-acetylphenoxy) butanoic acid linker. To improve the clinical applicability, researchers grafted the complementary-determining regions of P67.6 into a human immunoglubluin G4 (IgG4) kappa framework (hP67.6). Moreover, to stabilize the drug and to prevent Fab-arm exchange with endogenous human IgG4, a core-hinge mutation (S228P) was introduce in the IgG4 sequence [16,17]. Unconjugated P67.6 and hP67.6 antibodies per se lack substantial anti-leukemic activity, but they are useful to deliver a chemotherapy (e.g., calicheamicin derivatives) to CD33+ cells.
As already mentioned, after binding to the CD33 antigen, the GO-CD33 complex is rapidly internalized (Figure 1). The 4-(4’-acetylphenoxy)butanoic acid linker that connects the antibody portion to calicheamicin is rapidly hydrolyzed in acid environment, such as the lysosomes of the myeloblast. Following the hydrolyzation, calicheamicin dimethyl hydrazide is released and reduced by the glutathione into highly reactive species, which then bind to the DNA in the minor groove. The addition of calicheamicin to the DNA structure causes site-specific damages and, in particular, double-stranded breaks which are extremely toxic for proliferating cells. Downstream, the activation of the DNA repair pathway is mediated by the ataxia-telangiectasia mutated (ATM)/ataxia-telangiectasia and Rad3-related (ATR) protein kinases [18,19] (Figure 1). In turn, ATM and ATR kinases phosphorylate CHK1 and CHK2 proteins, which induce G2/M cell cycle arrest. The DNA-dependent pathway (DNA-PK) kinase participates in the response to DNA damages by phosphorylating H2AX histones and consequently promoting the recruitment of other DNA repair mediators on the site of damage. Finally, the DNA damage is repaired throughout the homologous recombination (HR) repair or the non-homologous end joining (NHEJ) repair mechanisms. Hence, it has been showed that cancer cells defective of different DNA damage response genes (e.s. ATM or DNA-PK) are hypersensitive to calicheamicin [20,21]. If the DNA damage is not repaired, ATM/ATR kinases trigger apoptosis through the phosphorylation of two BCL2 family proteins (BAX and BAK), which release the cytochrome-c and activate caspase 9 and 3 [22,23] (Figure 1). Data from a phase II trial suggest that the inhibition of BCL2 functionality using a specific antisense (Oblimersen sodium) may enhance the induction of leukemic cell apoptosis in patients subjected to a concomitant treatment with GO [24].
Cytotoxicity analyses in the HL-60 human leukemia cell line showed a 2000-fold higher effect of the drug compared with the unconjugated calicheamicin alone [25]. In the cytoplasm, free calicheamicin can be pumped out through ATP-binding cassette (ABC) transmembrane transporters (ABCB1 and ABCC1), thus reducing its cytotoxic effect [26]. Independently from the action of transmembrane transporters, the sensitivity to calicheamicin varies significantly among AML patients [27], thus emphasizing the importance of defining the factors that regulate the anti-leukemia activity of GO. The reasons why calicheamicin sensitivity changes among patients are still unknown.

4. Clinical Trials and Clinical Experience of GO in AML

4.1. GO as Monotherapy for Newly Diagnosed or Relapsed/Refractory Adult AML

Three phase II trials (0903B1-201-US/CA [NCT00003131], 0903B1-202-EU, 0903B1-203-US/EU [NCT00003673]) assessed the safety and efficacy of GO administered as a monotherapy at 9 mg/mq on day 1 and day 14 in adult AML patients at first relapse: the results of these studies showed that GO induces a complete remission (CR) or CR with incomplete platelet recovery (CRp) in up to 25–35% of patients [26,28,29,30,31,32,33].
Based on these results on single-drug activity, on May 2000, FDA accelerated the regulatory approval of GO for CD33+ AML patients older than 60 years, in first relapse and unfit for intensive treatment [34].
Phase I studies identified a dose and schedule for GO (9 mg/mq every 2 weeks), able to reach complete or near complete CD33-binding site saturation while lacking dose-limiting non-hematological toxicity [26,35]. This drug schedule is however limited to the fact that the expression of new CD33 molecules is constantly displayed on blasts cell surface and thus the antigen levels, down-modulated by GO exposure, return to baseline after 72 h [35]. Fractionated GO dosing (3 mg/mq every 3 days) may therefore enhance intracellular calicheamicin delivery compared with higher dose schedules, which may be supra-saturating in some patients.
The sequential phase II/III EORTC-GIMEMA AML-19 trial first determined the best GO induction regimen. In the phase II trial GO was administered as monotherapy, 6 mg/mq on day 1 plus 3 mg/mq on day 8 versus (vs.) GO 3 mg/mq on day 1, 3 and 5, while the phase III trial compared GO to best supportive care in patients ≥61 years that were unsuitable for intensive chemotherapy [36]. A higher rate of disease non-progression, defined as CR/CRp rate or patients in stable disease at the end of induction course was reported in patients treated with the first drug schedule. Moreover, another phase III clinical trial revealed that the schedule 6 mg/mq on day 1 plus 3 mg/mq on day 8 of GO monotherapy significantly improved overall survival (OS) compared to best supportive care (1-year OS: 24.3% vs. 9.7%, hazard ratio (HR): 0.69, 95% confidence interval (CI): 0.53–0.90, p = 0.005) [37].

4.2. GO as Monotherapy for Relapsed/Refractory Pediatric AML

Regarding pediatric AML, the first phase I study of GO for compassionate use for children with relapse/refractory disease defined the effective dose of 4–9 mg/mq in up to 3 cycles in monotherapy [38]. Arceci and colleagues studied the effect of treatment with GO in monotherapy with doses of 6–9 mg/mq (2 doses, 2-week intervals) in children with relapse/refractory AML. Using this drug schedule, they found CR in 30% and 26% of AML patients with refractory and relapsed disease, respectively [39]. In 2010, a subsequent phase II study showed a significantly higher survival of children with advanced AML treated with two doses of 7.5 mg/mq of GO with 14-day intervals compared to children who did not receive the treatment (3-year probability of overall survival: 27.0% vs. 0.0%, respectively; p = 0.001) [40].

4.3. GO in Combination with Chemotherapy for Newly Diagnosed or Relapsed/Refractory Adult and Pediatric AML

During the last years, different clinical trials evaluated the efficacy of GO in combination with anti-leukemic drugs or with drug efflux pump inhibitors in both newly diagnosed or advanced AML patients. Unfortunately, due to the limited number of patients enrolled and/or absence of correct control arms, they failed to provide convincing proofs of the efficacy of GO in those settings.
Four randomized clinical trials evaluated the efficacy of GO in combination with the first cycle of intensive chemotherapy in AML patients:
SWOG S016: The Southwest Oncology Group conducted a phase III randomized trial to evaluate the clinical benefit of adding GO (6 mg/mq on day 4) to the standard 3 + 7 induction regimen in AML patients at first relapse. To balance toxicities patients allocated to the GO arm received lower dose of daunorubicin (45 mg/mq vs. 60 mg/mq) in comparison with patient treated with conventional induction regimen [41]. The interim analysis reported a higher number of fatal toxicities in the GO arm compared to the other, causing the premature end of the study and the withdrawal of GO from the market on June 2010. Moreover, the complete data analysis of the trial failed to demonstrate any clinical improvement by GO addition both in terms of relapse-free survival (RFS) and OS (GO vs. non-GO arm: 5-year RFS, 43.0% vs. 42.0%; p = 0.40; 5-year OS, 46.0% vs. 50.0%; p = 0.85).
Medical Research Council (MRC) AML15 and National Cancer Research Institute (NCRI) AML16: based on the results of a dose-finding trial that evaluated GO addition (3 mg/mq vs. 6 mg/mq) to intensive chemotherapy, with 3 mg/mq GO appearing effective and safe [42], two randomized phase III trials addressed the clinical consequences of adding GO 3 mg/mq to the induction regimen in young (predominantly ≤60 years, MRC AML15) [43] and older patients (NRCI AML16) [44]. In these trials, the GO arm showed an improved OS in elderly patients and in younger ones with favorable-risk AML.
Acute Leukemia French Association (ALFA)-0701: since the CD33 antigen is rapidly re-expressed on the surface of AML blasts after GO exposure, the acute leukemia French association tested the fractionated treatment (3 mg/mq on day 1, 4 and 7) in a phase I/II study in combination with chemotherapy for the treatment of relapsed AML [45,46]. Sixty-five to 75.0% of patients achieved CR/CRp. Based on these results, the randomized phase III ALFA-0701 trial compared fractionated GO (3 mg/mq on days 1,4 and 7 during induction and on day 1 of each consolidation course) plus standard chemotherapy vs. chemotherapy alone in newly diagnosed CD33+ AML patients aged 50 to 70 years [47]. The GO arm displayed an improvement of event free survival (EFS, median value 13.6 vs. −9.5 months, HR: 0.66; 95% CI: 0.49–0.89; p = 0.006), but no differences in terms of OS compared with the chemotherapy arm (HR: 0.81; 95% CI: 0.60–1.09; p = 0.16) [47,48].
Thanks to these results, on September 2017, FDA re-approved GO for adult newly diagnosed CD33+ AML patients and for pediatric relapsed/refractory CD33+ AML patients (aged ≥2 years). GO received the marketing authorization of the European Medicine Agency (EMA) on April 2018 for the treatment of newly diagnosed de novo CD33+ AML patients aged ≥2 years, in combination with daunorubicin and cytarabine.
A meta-analysis of 3325 AML patients treated in the above reported clinical trials showed that the addition of GO had no impact on the overall remission rate. However, it reduced the 5-year cumulative incidence of relapse with an odds ratio (OR) of 0.81 (95% CI: 0.73–0.90, p < 0.001) and it improved survival (OR for death = 0.90, 95% CI: 0.82–0.98, p = 0.01) [49].
A subsequent trial (NCRI AML17) evaluated the impact of GO dosing 3 mg/mq vs. 6 mg/mq combined with intensive chemotherapy in a cohort of 788 newly diagnosed AML patients. The result of the study showed no correlation between the increase of GO dosage and clinical benefit. Indeed, the increased GO dosing did improve neither the response rate nor the patients’ outcome (OS: HR: 1.10; 95% CI: 0.90–1.34; p = 0.3; RFS: HR: 1.11; 95% CI: 0.91–1.35; p = 0.30 [50].

4.4. GO-related Toxicities

Most of GO-related toxicities have been reported after first infusion. Acute infusion-related toxicities, such as chills, fever, low or high blood pressure, nausea/vomiting were the most frequently observed events. However, all these events were usually transient and could be resolved using standard interventions. The most common adverse event reported in AML patients treated with GO in monotherapy was bone marrow myelosuppression, resulting in grade 3–4 neutropenia and thrombocytopenia. In the ALFA-0701 trial, similar toxicities were seen in patients treated with GO in combination with chemotherapy. In these patients, myeloid recovery was not significantly delayed following induction with GO and chemotherapy while platelet recovery was prolonged for days [47,48]. Severe intra-vascular hemolysis have been reported in some pediatric cases treated with GO [51]. The biological reason of this phenomenon may be ascribed to impaired hemoglobin scavenging coming from the elimination of CD33+ monocytes/macrophages expressing the CD163 hemoglobin scavenger receptor [14]. Other transient toxicities have been reported following GO therapy and, in particular, transient alterations of liver enzymes levels including hyper-bilirubinemia and increased level of aspartate and/or alanine aminotransferase (AST/ALT) [52,53].
Among GO-related toxicities, the development of veno-occlusive disease (VOD) has the highest clinical impact. Different clinical trials highlighted that the risk of VOD in patients treated with GO is dose-dependent. Indeed, a relatively low risk of VOD has been reported in AML patients receiving doses of GO lower than 3 mg/mq and in combination with conventional therapy [50]. Accordingly, a recent study on 137 GO-treated patients and 548 matched control subjects demonstrated that GO exposure before myeloablative allogenic transplantation does not associate with higher frequency of VOD or death [54]. On the other hand, the risk of VOD increases when GO is administered in heavily pre-treated AML patients or when the doses were higher than 3 mg/mq [55,56]. The biological reason for the development of VOD are still unknown, however similar observations come from patients treated with inotuzumab-ozogamicin, an anti-CD22 calicheamicin conjugate, used for the treatment of acute lymphoblastic leukemia (ALL) patients [57]. This phenomenon suggested that the mechanism of GO-associated VOD is CD33 independent but may be related to the structure of the antibody or related to calicheamicin toxicity. More than one aspect might be involved and their effective contribution might depend on the level of CD33-binding sites saturation achieved in the blood. Defibrotide, a drug commonly used to treat VOS, provided some benefits in the treatment of GO-induced VOD, with 17/27 patients (63.0%) surviving and/or showing a response, with a safety profile comparable to the one reported in other defibrotide studies [58].

4.5. GO Treatment before and after Transplantation in Adult AML Patients

Risk stratification in AML is currently used to tailor patients’ post-remission treatment, which may include transplant (autologous, autoSCT or allogeneic, alloSCT) or continued chemotherapy. Post-remission treatment for AML patients should be adjusted according to an assessment of transplant-related mortality (TRM) along with leukemia characteristics and minimal residual disease (MRD).
In this context, GO could be considered before harvesting, in order to achieve and/or consolidate MRD negativity, and during transplant, for intensifying conditioning regimens: of course, a modulation of GO dosage (and of conditioning regimens) should be evaluated. A list of clinical trials including GO treatment before or after transplantation is reported in Table 1 (clinicaltrials.gov updated to 1 September 2021).
In the favorable risk core binding factor (CBF) subtype, GO was not able to reduce the residual leukemia initiating clone that survived the consolidation therapy, thus showing no benefit in the setting of autoSCT [59].
The retrospective analysis of post-transplant outcomes in subjects who received HSCT as follow-up therapy in the ALFA-0701 trial showed that fractionated-dose GO in the induction and consolidation regimen did not induce higher rate of post-transplant VOD/sinusoidal obstruction syndrome or mortality [60]. Post-transplant outcomes were comparable between arms and the study failed to demonstrate the survival benefit observed in the GO vs. control arm in patients who did not receive HSCT. Taking together, these data indicate that HSCT can follow GO-based regimen, as consolidation treatment. Accordingly, in a “real-life” setting, the combination of fractionated GO with cytarabine and mitoxantrone (MYLODAM scheme) confirmed that a GO-based intensive regimen can be applied as bridge to alloSCT in relapsed/refractory AML [61]. Moreover, the efficacy of GO combinations as a potential bridge to transplant was confirmed in a retrospective study of 24 high-risk AML patients who received fractionated GO in combination with intermediate-dose cytarabine and daunorubicin as salvage therapy [62]. A recent study also reported that relapsed AML patients may also benefit of GO monotherapy as a conditioning regimen before second alloSCT from the same donor used in the first transplantation [63]. After transplantation, the disease relapse remains the major cause of therapy failure for AML patients. Moreover, the therapeutic options to treat relapsed patients after transplant are extremely limited, due to the rising of disease resistance and a higher risk of toxicities. Therefore, there is a clinical need for therapeutic strategies able to prevent or manage disease relapse. The optimal pharmacological compound should have a safe toxicity profile, an anti-tumor effect and an immune profile, which can be used to boost GVL and reduce GVHD. Several cases of treatment with GO or GO plus donor lymphocyte infusion (DLI) for AML relapsing after alloSCT have been previously reported. The available experiences [64,65,66,67] suggest that GO treatment followed by DLI is more effective when administered soon after relapse or, if possible, even in a pre-emptive setting. A recent study reported encouraging results by fractionated GO combined with intensive chemotherapy in adult CD33+ AML patients relapsing after alloHCT, as salvage regimen, with an overall response rate of 72.0% and OS of 42.0% at 2 years [68]. Moreover, the combination with additional targeted therapies, when available, has to be taken into account.

4.6. GO Treatment before and after Transplantation in Pediatric AML

GO is currently being evaluated in this setting also in the pediatric population (Table 1). Data based on clinical experience showed that GO can be safely added (i) to a busulfan/cyclophosphamide conditioning regimen before alloSCT in children and adolescents affected by poor-risk AML [69]; (ii) to fludarabine and cytarabine (FLA) before HSCT for first-line refractory AML in children [70]; (iii) to fludarabine, cytarabine, granulocyte colony-stimulating factor and idarubicin (FLAG-IDA) as reinduction therapy before a KIR-ligand-mismatched cord blood transplant in pediatric relapsed/refractory AML [71]. Moreover, GO treatment, either as monotherapy or in combination with cytarabine or other agents, of relapsed/refractory pediatric AML patients enabled blast reduction [72] also to MRD negativity levels [73], thus allowing HSCT, without imposing major adverse events.
In the pediatric setting, GO consolidation (4.5 mg/mq to 9 mg/mq per dose) after reduced-intensity conditioning and alloSCT was safe in pediatric patients with CD33+ AML [74] in CR1/CR2, with OS probability at 1 and 5 years of 78.0% and 61.0%, respectively [75].

5. Biomarkers of Response to GO Therapy

Since the clinical benefit of GO is variable among patients and potential adverse effects have been reported, it is important to identify factors able to predict treatment response (Figure 2).

5.1. CD33 Expression

The anti-leukemic effect of GO is likely correlated with the cytoplasmic level of activated calicheamicin derivates and to the intrinsic sensitivity of the target cells to DNA damage. The concentration of calichemicin molecules inside the target cells can be affected by the density of CD33 molecules on the cell surface, the efficiency of 4-(4’-acetylphenoxy) butanoic acid linker hydrolysis and the mechanism of calicheamicin activation.
CD33 is heterogeneously expressed on the surface of leukemic cells [76,77]. Data from mathematical models suggested that the cytoplasmic concentration of calichaemicin, given a fixed dose of GO, depends on the absolute number of CD33+ target cells, the CD33 production rate and the drug efflux pumps (ATP-binding cassette transporter) activity rather than on CD33 density on leukemic cell membrane [78]. Nevertheless, it has been showed the CD33 density on AML cells can affect GO efficacy in specific settings. In vitro studies showed that the surface CD33 expression was relevant to the drug cytotoxic effect and CD33 expression levels positively correlated with GO binding activity and leukemic cell clearance [79].
Different clinical trials investigated the relationship between CD33 density, quantified as mean fluorescence intensity or percentage of CD33+ blasts, and GO efficacy (Table 2):
The EORTC-GIMEMA AML-19 trial reported that GO therapy improved survival in AML patients with a percentage of CD33+ blasts higher than 80% [37];
In the ALFA-0701 trial, a retrospective analysis revealed that GO improved EFS and RFS of patients expressing high CD33 surface levels and the prognostic relevance was maintained when adjusting for other predictive markers such as cytogenetics and NPM1/FLT3-ITD mutational status [80];
In the MRC/NCRI trial, a significant relationship between the risk of post-GO relapse and the percentage of CD33+ myeloblasts has been documented. In details, patients with the lowest expression of CD33+ blasts were at higher risk of relapse when treated with GO compared with subjects with the highest expression, measured according to quartiles [81];
The Children’s Oncology Group (COG) AAML0531 trial reported an association between CD33 mean fluorescent intensity and response to GO. Patients belonging to the second to fourth quartiles, compared with the lowest quartile of CD33 mean fluorescence intensity, had higher CR rates, lower MRD rates at the end of the first therapy cycle, lower risk of relapse and better disease free survival (DFS) across three cytogenetic/molecular risk group [76].
Taken together, this evidence indicates that CD33 expression is an important pre-treatment biomarker of GO response and it should be used to select patients that can significantly benefit of the treatment.

5.2. CD33 Single Nucleotide Polymorphism

Splicing variants of the CD33 gene generate alternative isoforms of the transmembrane receptor compromising GO binding. Molecular analysis conducted on patients from the COG AAML0531 trial indicated that CD33 genotypes can predict response to GO therapy [82] (Table 2). Indeed, patients carrying the rs12459419 CC genotype (51.0% of patients) had a significant lower risk of relapse and better EFS and DFS when treated with GO plus chemotherapy compared with chemotherapy only. Conversely, the rs12459419 C > T (Ala14Val) polymorphism, either in heterozygosis (39.0% of cases) or homozygosis (10.0% canceled the clinical benefit of GO. The results were recently confirmed in the adult NPM1-mutated (mut) AML population from the AMLSG 09-09 phase III study, which compared the clinical outcome of patients receiving GO plus induction (3 mg/mq on day 1) and consolidation chemotherapy (3 mg/mq on day 1 of the first consolidation cycle) [83]. Patients with the rs12459419 CC genotype showed a superior RFS in the GO arm compared with conventional therapy. The rs12459419 C > T polymorphism resulted in CD33 exon 2 skipping and, consequently, in the generation a shorter CD33 isoform [84]. From a structural point of view, the above mentioned isoform lacks the immunoglobulin-like V-set domain, which is the specific antigen detected by GO and which is generally used for the quantification of CD33 level of expression in flow cytometry [82]. As a consequence, patients displaying the TT genotype, had a significantly lower CD33 expression compared to those with CT or CC genotype (TT < T < CC: p < 0.001) [85,86]. Similar data were reported for the rs3865444 single nucleotide polymorphism (SNP), which localizes in the non-coding, promoter region of the CD33 gene and frequently occurs in linkage disequilibrium with rs12459419 [82,87]. In a multivariate analysis including the CD33 genotype, cytogenetic/molecular risk and CD33 surface expression, the rs12459419 CC genotype was still independently associated with lower RR and better DFS, while CD33 expression lost its significance in the evaluation of GO response [85]. This association, which deserves further investigation, could explain the differences in GO response observed among races, since the rs12459419 SNP CC genotype is more frequent in African-Americans compared with the European population [85]. Five additional CD33 SNPs identified in AML patients, namely rs1803254 (G > C; 3’UTR), rs35112940 (G > A; Arg304Gly), rs2455069 (A > G; Arg69Gly), rs61736475 (T > C; Ser305Pro) and rs201074739 (CCGG deletion), can impact on GO response [88]. A reduced RR was observed in GO treated patients carrying rs1803254 GG (p = 0.009), rs35112940 GG (p < 0.001), rs2455069 GG (p = 0.005), rs1736475 TT (p = 0.002) and rs201074739 CCGG/CCGG (p = 0.002).
Interestingly, a recent composite score, namely the CD33_PGx6_Score, based on six CD33 SNPs (rs12459419, rs2455069, rs201074739, rs35112940, rs61736475 and rs1803254), has been established to understand the impact of CD33 SNPs on CD33 expression and on GO efficacy [88]. The analysis, conducted on 938 patients from the COG AAML0531 cohort, showed that a CD33_PGx6_Score higher than 0 associated with high CD33 expression, better RFS (5-year RFS: 62.5% vs. 46.8% in the GO arm compared to the control arm; p = 0.008) and lower RR (5-year RR: 28.3% vs. 49.9% in the GO arm compared to the non-GO arm; p < 0.001). Conversely, the addition of GO did not affected on the outcome of AML patients having a CD33_PGx6_Score below 0.

5.3. Cytogenetic Alterations

Data from randomized clinical trials evaluating the effect of GO and intensive chemotherapy in adults AML patients highlighted that the efficacy is correlated with the cytogenetic risk (Table 2). In details, the addition of GO was associated with a survival benefit of 20.7% and 5.7% in patients from good (OR: 0.47; 95% CI: 0.31–0.73, p < 0.001) and intermediate cytogenetic risks (OR: 0.84; 95% CI: 0.75–0.95, p = 0.005) groups, respectively [49]. Conversely, GO did not improve clinical outcome in the adverse cytogenetic group, that generally express lower levels of CD33, with an absolute survival benefit at 6 years of 2.2% (OR: 0.99; 95% CI: 0.83–1.18, p = 0.90). Several studies corroborated the clinical benefit of GO addition in non-adverse cytogenetic risk patients [37,43,47,89].
CBF and lysine methyltransferase 2A (KMT2A)-rearranged AML, that belong to the good and intermediate/high risk classes, respectively, deserve further discussion.
CBF-AMLs are characterized by low CD33 expression [90], which can be predictive of poor GO response. The low CD33 expression is likely related to the cellular stage of differentiation. Indeed, t(8;21)/inv(16)/t(16;16) arise in preleukemic CD33 cells at a very early stage of differentiation [91] and this cells may be spared by GO treatment. Conversely, additional alterations inducing a proliferative status occur late in CD33 expressing cells, that can be effectively targeted by GO. However, a phase II trial studying the efficacy in adult CBF-AML patients of the FLAG induction regimen as frontline therapy in combination with GO 3 mg/mq at induction day 1 and post-remission course 1 and 2 day 1 (FLAG-GO), showed that FLAG-GO induced a higher remission rate (95.0%) and it was associated with a 3-year OS and RFS of 78.0% and 85.0%, respectively [92]. In particular, the addition of GO to standard chemotherapy was demonstrated to mitigate the risk of relapse in CBF cases carrying mutations in the exon17 of the KIT gene [93]. The benefit of GO addition was also confirmed by comparing FLAG-GO with FLAG-Idarubicin, in terms of molecular response rate (76.0% vs. 42.0%, p = 0.002) and 5-year RFS (87.0% vs. 68.0%, p = 0.02) [94].
11q23/KMT2A-rearrangements, which characterize 4% of adult [95] and 15–20% of pediatric AML [96], associate with high CD33 expression in leukemic cells [97]. Despite being classified as adverse risk leukemia, in most cases KMT2A-rearranged AML had a good response to GO in relapsed/refractory patients [98,99]. The analysis of 215 KMT2A-rearranged AML from the COG AAML0531 trial revealed that patients treated with GO in combination with chemotherapy achieved higher EFS compared with those receiving chemotherapy alone (5-year EFS: 48.0% vs. 28.0%, p = 0.002) [100].
These differences may be due to diverse blast sensitivity to calicheamicin (e.g., high sensitivity in CBF cells) and, potentially, drug uptake into AML cells [92,97]. Indeed, ABC transporter activity is generally high in elderly AML patients and in adults with adverse-risk cytogenetics [101,102]. This may explain the poor response to GO therapy of adult patients characterized by adverse-risk cytogenetics. In this scenario, the poor response rate may be also relate to the lower cell surface density of CD33 in these cases. Notably, the cytogenetic/molecular risk did not affect OS and DFS in a phase II multicenter clinical trial that enrolled 130 patients, aged < 65, treated with FLAI-GO induction regimen. The study showed a CR rate of 82.0% after induction [103]. The probability of 1, 2, and 5-year DFS was 77.0%, 58.0% and 52.0%, respectively, with a median follow-up of 54 months. Age and molecular remission after FLAI-GO and alloSCT predicted prolonged DFS in a Cox multivariate analysis. These data raises new hopes on the combination of GO with induction and consolidation chemotherapy regimens (other than cytarabine and doxorubicin only) for the treatment of high-risk patients.

5.4. Molecular Profile

The molecular profile is commonly used to stratify AML patients into prognostic subgroups when receiving standard chemotherapy [95]. Similar approaches can be exploited also when using newly approved antileukemic drugs.
Recently, Fournier and colleagues evaluated the predictive value of molecular alterations on the efficacy of combining GO with standard frontline chemotherapy [104]. By analyzing data from the ALFA-0701 trial, they confirmed that only patients classified into favorable (HR 0.54; 95% CI: 0.30–0.98) and intermediate (HR 0.57; 95% CI: 0.33–1.00) risk categories according to the European LeukemiaNet (ELN) 2017 risk stratification could benefit of GO combined with conventional chemotherapy. Conversely, the outcome of patients belonging to the adverse risk group was not affected by GO (HR 0.93; 95% CI: 0.61–1.43), in line with data obtained by cytogenetic risk classification [49,91] (Table 2).
When focusing on individual gene mutations, data from the literature indicate that NPM1-mut (25–35% of AML patients in general and 45–60% of cytogenetically normal AML) or FLT3-ITD patients (~20% of cases) had a higher CD33 expression compared to wildtype (wt) cases [80,105,106] and may benefit from GO treatment. In the ALFA-0701 trial, a clinical benefit was observed on 2-year EFS, RFS and OS in the NPM1-mut cohort [47]. The AMLSG 09-09 study did not confirm the result in terms of 2-year EFS (HR: 0.83, 95% CI: 0.65–1.04; p = 0.1), however GO treatment reduced the incidence of relapse in patients achieving CR/CR with incomplete hematologic recovery (HR: 0.66; 95% CI: 0.49–0.88; p = 0.005) [107]. In this cohort, GO also improved 2-year EFS of FLT3 wild-type, but not FLT3-ITD mut patients (HR: 0.72; 95% CI: 0.56–0.95 vs. HR: 1.53; 95% CI: 0.95–2.48, respectively; p = 0.002). Similar results regarding FLT3 mutational status and response to GO were obtained in adult AML patients from the MRC AML15 and NCRI AML16 trials, in which GO failed to demonstrate a clinical improvement in FLT3-ITD cases [49]. In this cohort, GO benefit on NPM1-mut cases was not seen either. This outcome may be partially attributed to the administration of a single GO dose in the MRC AML15 and NCRI AML16 trials. In contrast, other studies reported an improved OS, EFS and RFS in adult AML patients with FLT3-ITD by the addition of GO [47,89,104]. The analysis of FLT3-ITD patients from the COG AAML03P1 and AAML0531 studies revealed a decreased RR in those treated with GO-based regimens (37.0% vs. 59.0%, p = 0.02) [108]. In particular, the incidence of relapse was significantly reduced in patients that were exposed to GO prior of undergoing a HSCT in first CR (22.0% vs. 56.0%, p = 0.003). Interestingly, the cohort of poor prognosis patients harboring a high FLT3-ITD allelic ratio, that was exposed to GO prior to transplant, had a lower RR (15.0% vs. 53.0%, p = 0.007).
In the ALFA-0701 trial the benefit of GO was not limited to FLT3-ITD patients, but it was extended to all cases with activating signaling mutations (FLT3-ITD, FLT3-TKD, KRAS, NRAS, S, PTPN11, JAK2, RIT1, CBL, HR 0.43; 95% CI: 0.28–0.65), which correlated with higher CD33 expression levels [104]. Notably, mutant PTPN11 was shown to confer resistance to the BCL-2 inhibitor venetoclax via upregulating MCL1, pMCL1, and BCL-xL [109], thus indicating that GO-based regimens may be a valuable therapeutic strategy for this subgroup of patients. Elevated CD33 expression was also measured in patients carrying co-occurrent mutations in epigenetic modifiers and signaling genes (98.0% vs. 60.0% of CD33+ cells in patients carrying both mutation types and altered epigenetic regulators only, respectively; p < 0.001). No association was reported between CD33 expression and mutations of NPM1 or spliceosome genes in this study.
The link between activating signaling mutation and GO benefit is not clear. This observation is substantiated by the good response of CBF patients [92,94], that frequently harbors mutations in signaling genes [110], to GO treatment, despite the low CD33 expression [90]. As already mentioned, CD33 is a transmembrane glycoprotein receptor whose downstream pathway results in an inhibitory effect on myeloid cells [8]. Therefore, the co-occurrence with mutations in signaling genes may be an oncogenic mechanism adopted by leukemic cells to overcome CD33 downstream effects [111].
In contrast to the above results, the GOELAMS/FILO AML 2006-IR trial, enrolling younger AML patients (<60 years) with intermediate cytogenetic risk, failed to achieve a clinical improvement by the addition of GO to chemotherapy, even by stratifying cases according to their molecular profile [112]. Unsupervised analysis of patients according to the mutational status of seven genes (NPM1, FLT3-ITD, CEBPA, DNMT3A, IDH1, IDH2, ASXL1) identified six mutational clusters defining three major outcome groups (group A: NPM1-mut, FLT3-ITD-wt or biallelic CEBPA-mut; group B: no mutations, or NPM1-mut FLT3-ITD or NPM1-mut, FLT3-ITD-wt, epigenetic mutations; group C: NPM1-wt, FLT3-ITD). Although the results may not be conclusive, due to GO-related toxicity that led to early closure, group C showed the worst OS, DFS and EFS, followed by group B, while group A displayed the best outcome, when considering either patients receiving standard chemotherapy or those treated by chemotherapy plus GO.
Overall, these data suggest that the benefit of GO can be dependent on the disease genetic background, with patients carrying mutations in signaling genes and high CD33 expression being candidate for a better response. The analysis of the mutational status of a large panel of AML-related genes may help define genetic backgrounds predicting a clinical benefit related to GO addition to chemotherapy regimens and may guide novel tailored treatment strategies. For example, current trials are evaluating the relevance of the use of GO in combination with targeted therapies [33], including FLT3 inhibitors (NCT03900949, NCT04385290, NCT04293562) and PARP inhibitor (NCT04207190).
In addition to the genetic profile, the transcript levels of the anticoagulant factor ANXA5 were shown to predict better OS and EFS in multivariate analysis in pediatric AML receiving GO in combination with conventional chemotherapy [113]. By dividing the cohort according to the median ANXA5 expression, patients showing high ANXA5 levels had significantly better OS (p = 0.0012) and EFS (p < 0.001) compared with the other group.

5.5. Multidrug Resistance

The function of the ABCB1 multidrug resistance protein has been reported as a mechanisms dampening in vitro GO-mediated apoptosis [114,115,116]. Despite its pro-apoptotic effects, it is not surprising that free calicheamicin may be a substrate of the ABCC1 transporter [115,117,118]. Although similar results were obtained when GO is administered in combination with intensive chemotherapy, formal proofs are still lacking. 58% of AML patients expresses ABCB1 and its frequency in blasts cells varies from 19.0% to 75.0% of positive cells [118,119,120]. Several studies reported an association between ABCB1 and poor in vivo GO response, including failure to clear bone marrow blasts and to achieve CR [118,121,122,123], or dismal outcome, as indicated by OS and EFS [115,119,124,125] (Table 2). This data was confirmed in a cohort of pediatric relapsed/refractory AML patients, in which the relative degree of ex vivo drug efflux was predictive of GO response in the clinical setting. Indeed, 5 out of 8 patients with low drug efflux ratios achieved CR/CRp, that was not in patients with high drug efflux levels [39]. Other studies suggested that GO response may be also shaped by the inter-individual variability of calicheamicin efflux level [27]. Moreover, the ABCB1 genotype was shown to have a clinical impact on GO response, by acting on the accumulation of calicheamicin. A recent analysis on 942 patients from the COG AAML0531 cohort reported that GO recipients with rs1045642 (C > T; Ile1145Ile) CT or TT genotype displayed a better outcomes compared with those displaying the CC genotype (CT or TT vs. CC, 5-year EFS: p = 0.022; 5–year RR: p = 0.007), which correlated with an increased intracellular calicheamicin retention [123].

5.6. Minimal Residual Disease

Monitoring MRD after induction even in patients achieving morphological CR has prognostic impact in AML patients [126,127,128]. Therefore, different molecular or immunophenotypic markers have been studied to monitor MRD also in response to GO treatment. In the NCRI AML16 trial, the MRD level was measured by flow cytometry in order to evaluate its role as independent prognostic factor of response. No significant correlation has been identify between MRD negativity and the administration of GO in comparison to the control arm (57.0% vs. 48.0%; p = 0.18) [128].
NPM1 mutation and Wilms’ tumor 1 (WT1) gene expression are suitable prognostic molecular markers for MRD analysis by quantitative reverse-transcription polymerase chain reaction [129]. The ALFA-0701 study showed that negativity for NPM1 mutation MRD was frequently achieved in GO-treated patients compared to the control arm, both at the end of induction (39.0% vs. 7.0%; p = 0.006) and end of treatment (91.0% vs. 61.0%; p = 0.028) [129]. Conversely, GO provided no advantage when MRD was measured by WT1 transcript level at both time points (MRD negativity, after induction: 75.0% vs. 65.0%; p = 0.29; at the end of treatment 82.0% vs. 80.0%; p = 1.0).
Recently, the prognostic impact of NPM1 mutation MRD in AML patients receiving GO was determined in the phase III AMLSG 09-09 trial, that analyzed 7526 samples, including peripheral blood and bone marrow, from 469 patients [130]. At the end of cycle 2 and at the end of treatment, a reduction ≥1000 of NPM1 mutation transcript combined with MRD negativity was predictive of lower incidence of relapse. Patients receiving GO plus chemotherapy achieved lower levels of transcript level of NPM1 mutation across all treatment cycles compared with those treated with standard chemotherapy, thus resulting in a higher frequency of MRD negative cases at the end of treatment (56.0% vs. 41.0%; p = 0.01). Moreover, MRD positive patients belonging to the GO arm showed lower levels of NPM1 mutation transcript after two treatment cycles, which in turn led to lower RR (29.3% vs. 45.7% at 4-years; p = 0.009).
Overall, these data indicate that MRD is a valuable tool to measure GO response and that a combination strategy based on GO and chemotherapy can increase the rate of patients achieving MRD negativity (Table 2).

5.7. Stemness Signature

Leukemic stem cells (LSC) are the main determinants of AML refractoriness and relapse. The main features responsible for therapy resistance include cell cycle quiescence, self-renewal and high levels of drug efflux [131]. Recently, a 17-gene expression signature for LSC has been established in AML, named the LSC17 score, that has prognostic value [132]. In particular, its application to the ALFA-0701 trial data revealed that GO addition improved the outcome of patients having low LSC17 score (EFS: HR: 0.42; p = 0.001; RFS: HR: 0.53; p = 0.03), but not those with high signature score. These results indicate that the LSC17 score can serve as biomarker to predict response of AML patients to GO (Table 2).
Table 2. Main evidence of in vivo GO efficacy in AML subgroups defined according to biomarkers of response.
Table 2. Main evidence of in vivo GO efficacy in AML subgroups defined according to biomarkers of response.
BiomarkerMain ObservationsStudyReferences
CD33 expressionGO improved OS of patients with >80% CD33+ blastsEORTC-GIMEMA AML-19[37]
GO improved EFS and RFS of patients expressing high CD33 surface levelsALFA-0701[80]
Low percentage of CD33+ blasts associated with higher risk of relapse after GOMRC AML15, NCRI AML16[81]
Patients in the 2nd to 4th quartiles of CD33 surface expression had higher CR and lower MRD rates at the end of the first cycle, lower risk of relapse and better DFSCOG AAML0531[76]
CD33 SNPsrs12459419 CC genotype: lower risk of relapse and better EFS and DFS in the GO armCOG AAML0531[82]
rs12459419 C > T SNP: no GO benefitCOG AAML0531[82]
NPM1-mut patients with the rs12459419 CC genotype showed a superior RFS in the GO arm AMLSG 09-09[83]
Patients carrying rs1803254 GG, rs35112940 GG, rs2455069 GG, rs1736475 TT and rs201074739 CCGG/CCGG had reduced RR to GOCOG AAML0531[88]
CD33_PGx6_Score >0 associated with high CD33 expression, better RFS and lower RR in the GO armCOG AAML0531[88]
Cytogenetic alterationsGO provided a survival benefit in patients with good and intermediate cytogenetic risks, but not in the adverse cytogenetic groupVarious[37,43,47,52,89,91]
The addition of GO to standard chemotherapy reduced the risk of relapse in CBF cases carrying KIT mutationsFLAG-GO[93]
The addition of GO to standard chemotherapy induced higher EFS in KMT2A-rearranged AMLCOG AAML0531[100]
Molecular profileGO provided a survival benefit in patients from favorable and intermediate, but not adverse molecular risk categories (ELN 2017)ALFA-0701[49,91]
GO provided EFS, RFS and OS benefit in NPM1-mut AML and reduced the incidence of relapse in NPM1-mut patients achieving CR/CRiALFA-0701, AMLSG 09-09[50,107]
GO improved EFS of FLT3-ITD-wt, but not FLT3-ITD-mut patientsAMLSG 09-09, MRC AML15, NCRI AML16[52,107]
GO improved OS, EFS and RFS and reduced the RR in adult FLT3-ITD-mut patientsCOG AAML03P1, COG AAML0531[50,92,107,108]
GO provided clinical benefit to patients with activating signaling mutationsALFA-0701[104]
The mutational status of seven genes identified a group characterized by NPM1-mut, FLT3-ITD-wt or biallelic CEBPA-mut that displayed the best outcome in the GO armGOELAMS/FILO AML 2006-IR[112]
Multidrug resistanceABCB1 expression associated with failure to clear bone marrow blasts and to achieve CR or poos OS and EFSVarious[115,118,119,121,122,123,124,125]
ABCB1 rs1045642 CT or TT genotype associated with better outcomes in GO recipientsCOG AAML0531[123]
MRDGO-treated patients frequently achieved negativity for NPM1 mutation MRD and a reduction 1000 of NPM1 mutation transcript combined with MRD negativity was predictive of lower RRALFA-0701, AMLSG 09-09[130,131]
LSC signatureGO addition improved the outcome of patients having low LSC17 score but not those with high signature scoreALFA-0701[132]
CR: complete remission; CRi: complete remission with incomplete hematologic recovery; DFS: disease free survival; EFS: event free survival; LSC: leukemia stem cell; MRD: minimal residual disease; mut: mutated; OS: overall survival; RFS; relapse free survival; RR: relapse rate; SNP: single nucleotide polymorphism.

6. Novel Preclinical GO-based Therapeutic Combinations in AML

In addition to strategies based on standard chemotherapy regimens, GO is under clinical investigation in association with novel drugs (e.g., CPX-351 liposome), the demethylating agent azacytidine and/or targeted therapies, including venetoclax, avelumab, FLT3 inhibitors (midostaurin, gilteritinib), glasdegib, bortezomib, pacrinostat, talazoparib, that are variably combined between each other and with nucleoside analogs and/or anthracyclines (Figure 3) [33].
Moreover, insights for novel and future therapeutic combinations come from preclinical studies. Non-cytotoxic concentrations of the PP242 [133] or the combination with AZD2014 [134], two mTORC1/2 dual kinase inhibitors, enhanced GO cytotoxicity in AML cells, by potentiating lysosomal functions and suppressing GO-induced CHK1 activation, thus promoting cell cycle progression with damaged DNA and, ultimately, cell death. DNA damage accumulation is also at the base of the successful combination of GO with the farnesyltransferase inhibitor tipifarnib/zarnestra. It has been showed that farnesyltransferase inhibitors induce DNA damage generating reactive oxygen species [135]. In particular, the CD34+CD38 phenotype and cell dormancy, that impaired DNA damage resolution, were predictive of higher chemosensitivity to the drug combination ex vivo [136].
Great interest is currently raising around the combination of GO with DNA repair inhibitors. Since calicheamicin induces DNA double strand breaks, agents that block the non-homologous end joining (NHEJ) pathway are expected to synergize with GO treatment. This hypothesis has been confirmed by combining GO with the inhibitor of DNA-PK (M3814) [137] or with the PARP inhibitors olaparib [138] and talazoparib [139].

7. Conclusions

CD33 represents a bona fide target in AML therapy, since it is highly expressed in the majority of leukemic cells. GO, targeting CD33, is not a perfect drug, due its conjugation technology and susceptibility to depotentiation by ABC transporter activity. The clinical history of GO in oncology has been long and unusual. After an accelerated approval for AML patients, GO was withdrawn by the company in 2010 following the confirmatory phase III clinical trial, in which GO did not show a clear clinical benefit in comparison to control arms. Indeed, the study was stopped prematurely after the high number of early deaths occurred in the arm receiving GO in comparison with those receiving chemotherapy alone. However, in light of the widespread CD33 expression on AML cells and its specificity for the myeloid lineage, GO was an interesting drug in the field. GO came back into the market seven year later after the promising results of the spontaneous ALFA-0701 trial, in which novel GO drug schedule and concentration demonstrated superiority in favorable and intermediate risk patients. A better knowledge of the disease biology and its molecular subtypes was crucial for the definition of a target population that could benefit of GO-based regimens.
Over the past years, the advancements of sequencing technologies, strategies for MRD monitoring, genotyping studies and the increased knowledge of the mechanisms of therapy resistance have expanded the number and type of biomarkers able to predict GO response. Future studies on large cohorts should try to combine the diverse biomarkers to improve the prediction accuracy, thus paving the way for precision medicine in the real-life clinical practice.
Moreover, the development of novel targeted agents paves the way to new combinatory therapies that may enhance GO efficacy. Indeed, ongoing studies are investigating the role of GO in in patients’ subpopulations and in newer therapy combinations, towards more personalized and, hopefully, less toxic, approaches. Currently, studies open for enrollment are mainly focused on GO treatment in elderly patients, in combination with demethylating agents and other targeted drugs. In this clinical setting, drug benefits, that display a high level of variability among patients, according to known biomarkers and target-specific toxicity affecting normal tissue, which may be a limit to GO administration, have to be taken into account carefully. The regulatory history of GO is a hallmark of the potential pitfalls that may occur in the drug development process towards clinical application.

Author Contributions

M.G., G.S., A.S., D.N., A.G.L.d.R., A.P. and C.C. have drafted the manuscript and created the figures and tables. M.N., M.B.G., G.M. (Gerardo Musuraca), and F.L. critically revised the manuscript, G.M. (Giovanni Martinelli) supervised the work and acquired funding. All authors have approved the submitted version and agree to be personally accountable for the author’s own contributions and for ensuring that questions related to the accuracy or integrity of any part of the work, even ones in which the author was not personally involved, are appropriately investigated, resolved, and documented in the literature. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by Treviso AIL (Associazione Italiana Leucemie).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Farkona, S.; Diamandis, E.P.; Blasutig, I.M. Cancer immunotherapy: The beginning of the end of cancer? BMC Med. 2016, 14, 73. [Google Scholar] [CrossRef] [Green Version]
  2. Marin-Acevedo, J.A.; Soyano, A.E.; Dholaria, B.; Knutson, K.L.; Lou, Y. Cancer immunotherapy beyond immune checkpoint inhibitors. J. Hematol. Oncol. 2018, 11, 1–25. [Google Scholar] [CrossRef]
  3. Kim, E.G.; Kim, K.M. Strategies and advancement in antibody-drug conjugate optimization for targeted cancer therapeutics. Biomol. Ther. 2015, 23, 493–509. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Schumacher, D.; Hackenberger, C.P.R.; Leonhardt, H.; Helma, J. Current Status: Site-Specific Antibody Drug Conjugates. J. Clin. Immunol. 2016, 36, 100–107. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Teicher, B.A.; Chari, R.V.J. Antibody conjugate therapeutics: Challenges and potential. Clin. Cancer Res. 2011, 17, 6389–6397. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  6. Lin, J.H.; Guo, Y.; Wang, W. Challenges of Antibody Drug Conjugates in Cancer Therapy: Current Understanding of Mechanisms and Future Strategies. Curr. Pharmacol. Rep. 2018, 4, 10–26. [Google Scholar] [CrossRef]
  7. Von Gunten, S.; Bochner, B.S. Basic and clinical immunology of Siglecs. Ann. N. Y. Acad. Sci. 2008, 1143, 61–82. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  8. Lajaunias, F.; Dayer, J.M.; Chizzolini, C. Constitutive repressor activity of CD33 on human monocytes requires sialic acid recognition and phosphoinositide 3-kinase-mediated intracellular signaling. Eur. J. Immunol. 2005, 35, 243–251. [Google Scholar] [CrossRef]
  9. Orr, S.J.; Morgan, N.M.; Elliott, J.; Burrows, J.F.; Scott, C.J.; McVicar, D.W.; Johnston, J.A. CD33 responses are blocked by SOCS3 through accelerated proteasomal-mediated turnover. Blood 2007, 109, 1061–1068. [Google Scholar] [CrossRef]
  10. Gbadamosi, M.O.; Shastri, V.M.; Hylkema, T.; Papageorgiou, I.; Pardo, L.; Cogle, C.R.; Doty, A.; Loken, M.R.; Meshinchi, S.; Lamba, J.K. Novel CD33 antibodies unravel localization, biology and therapeutic implications of CD33 isoforms. Future Oncol. 2021, 17, 263–277. [Google Scholar] [CrossRef]
  11. Hernández-Caselles, T.; Martínez-Esparza, M.; Pérez-Oliva, A.B.; Quintanilla-Cecconi, A.M.; García-Alonso, A.; Alvarez-López, D.M.R.; García-Peñarrubia, P. A study of CD33 (SIGLEC-3) antigen expression and function on activated human T and NK cells: Two isoforms of CD33 are generated by alternative splicing. J. Leukoc. Biol. 2006, 79, 46–58. [Google Scholar] [CrossRef]
  12. Pérez-Oliva, A.B.; Martínez-Esparza, M.; Vicente-Fernández, J.J.; Corral-San Miguel, R.; García-Peñarrubia, P.; Hernández-Caselles, T. Epitope mapping, expression and post-translational modifications of two isoforms of CD33 (CD33M and CD33m) on lymphoid and myeloid human cells. Glycobiology 2011, 21, 757–770. [Google Scholar] [CrossRef] [Green Version]
  13. Laszlo, G.S.; Harrington, K.H.; Gudgeon, C.J.; Beddoe, M.E.; Fitzgibbon, M.P.; Ries, R.E.; Lamba, J.K.; McIntosh, M.W.; Meshinchi, S.; Walter, R.B. Expression and functional characterization of CD33 transcript variants in human acute myeloid leukemia. Oncotarget 2016, 7, 43281–43294. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Griffin, J.D.; Linch, D.; Sabbath, K.; Larcom, P.; Schlossman, S.F. A monoclonal antibody reactive with normal and leukemic human myeloid progenitor cells. Leuk. Res. 1984, 8, 521–534. [Google Scholar] [CrossRef]
  15. Andrews, R.G.; Singer, J.W.; Bernstein, I.D. Precursors of colony-forming cells in humans can be distinguished from colony-forming cells by expression of the CD33 and CD34 antigens and light scatter properties. J. Exp. Med. 1989, 169, 1721–1731. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  16. Hamann, P.R.; Hinman, L.M.; Hollander, I.; Beyer, C.F.; Lindh, D.; Holcomb, R.; Hallett, W.; Tsou, H.R.; Upeslacis, J.; Shochat, D.; et al. Gemtuzumab ozogamicin, a potent and selective anti-CD33 antibody-Calicheamicin conjugate for treatment of acute myeloid leukemia. Bioconjug. Chem. 2002, 13, 47–58. [Google Scholar] [CrossRef]
  17. Labrijn, A.F.; Buijsse, A.O.; Van Den Bremer, E.T.J.; Verwilligen, A.Y.W.; Bleeker, W.K.; Thorpe, S.J.; Killestein, J.; Polman, C.H.; Aalberse, R.C.; Schuurman, J.; et al. Therapeutic IgG4 antibodies engage in Fab-arm exchange with endogenous human IgG4 in vivo. Nat. Biotechnol. 2009, 27, 767–771. [Google Scholar] [CrossRef] [PubMed]
  18. Amico, D.; Barbui, A.M.; Erba, E.; Rambaldi, A.; Introna, M.; Golay, J. Differential response of human acute myeloid leukemia cells to gemtuzumab ozogamicin in vitro: Role of Chk1 and Chk2 phosphorylation and caspase 3. Blood 2003, 101, 4589–4597. [Google Scholar] [CrossRef] [Green Version]
  19. Mårtensson, S.; Nygren, J.; Osheroff, N.; Hammarsten, O. Activation of the DNA-dependent protein kinase by drug-induced and radiation-induced DNA strand breaks. Radiat. Res. 2003, 160, 291–301. [Google Scholar] [CrossRef]
  20. Elmroth, K.; Nygren, J.; Mårtensson, S.; Ismail, I.H.; Hammarsten, O. Cleavage of cellular DNA by calicheamicin γ1. DNA Repair 2003, 2, 363–374. [Google Scholar] [CrossRef]
  21. Sullivan, N.; Lyne, L. Sensitivity of fibroblasts derived from ataxia-telangiectasia patients to calicheamicin γ1I. Mutat. Res. Lett. 1990, 245, 171–175. [Google Scholar] [CrossRef]
  22. Prokop, A.; Wrasidlo, W.; Lode, H.; Herold, R.; Lang, F.; Henze, G.; Dörken, B.; Wieder, T.; Daniel, P.T. Induction of apoptosis by enediyne antibiotic calicheamicin θII proceeds through a caspase-mediated mitochondrial amplification loop in an entirely Bax-dependent manner. Oncogene 2003, 22, 9107–9120. [Google Scholar] [CrossRef] [Green Version]
  23. Haag, P.; Viktorsson, K.; Lindberg, M.L.; Kanter, L.; Lewensohn, R.; Stenke, L. Deficient activation of Bak and Bax confers resistance to gemtuzumab ozogamicin-induced apoptotic cell death in AML. Exp. Hematol. 2009, 37, 755–766. [Google Scholar] [CrossRef] [PubMed]
  24. Moore, J.; Seiter, K.; Kolitz, J.; Stock, W.; Giles, F.; Kalaycio, M.; Zenk, D.; Marcucci, G. A Phase II study of Bcl-2 antisense (oblimersen sodium) combined with gemtuzumab ozogamicin in older patients with acute myeloid leukemia in first relapse. Leuk. Res. 2006, 30, 777–783. [Google Scholar] [CrossRef] [PubMed]
  25. McGavin, J.K.; Spencer, C.M. Gemtuzumab ozogamicin. Drugs 2001, 61, 1317–1322. [Google Scholar] [CrossRef] [PubMed]
  26. Cowan, A.J.; Laszlo, G.S.; Estey, E.H.; Walter, R.B. Antibody-based therapy of acute myeloid leukemia with gemtuzumab ozogamicin. Front. Biosci. 2013, 18, 1311–1334. [Google Scholar] [CrossRef] [Green Version]
  27. Goemans, B.F.; Zwaan, C.M.; Vijverberg, S.J.H.; Loonen, A.H.; Creutzig, U.; Hählen, K.; Reinhardt, D.; Gibson, B.E.S.; Cloos, J.; Kaspers, G.J.L. Large interindividual differences in cellular sensitivity to calicheamicin may influence gemtuzumab ozogamicin response in acute myeloid leukemia. Leukemia 2008, 22, 2284–2285. [Google Scholar] [CrossRef] [Green Version]
  28. Pagano, L.; Fianchi, L.; Caira, M.; Rutella, S.; Leone, G. The role of Gemtuzumab Ozogamicin in the treatment of acute myeloid leukemia patients. Oncogene 2007, 26, 3679–3690. [Google Scholar] [CrossRef] [Green Version]
  29. Breccia, M.; Lo-Coco, F. Gemtuzumab ozogamicin for the treatment of acute promyelocytic leukemia: Mechanisms of action and resistance, safety and efficacy. Expert Opin. Biol. Ther. 2011, 11, 225–234. [Google Scholar] [CrossRef] [PubMed]
  30. Htter, M.L.; Schlenk, R.F. Gemtuzumab ozogamicin in non-acute promyelocytic acute myeloid leukemia. Expert Opin. Biol. Ther. 2011, 11, 1369–1380. [Google Scholar] [CrossRef]
  31. Takeshita, A. Efficacy and resistance of gemtuzumab ozogamicin for acute myeloid leukemia. Int. J. Hematol. 2013, 97, 703–716. [Google Scholar] [CrossRef] [Green Version]
  32. Thol, F.; Schlenk, R.F. Gemtuzumab ozogamicin in acute myeloid leukemia revisited. Expert Opin. Biol. Ther. 2014, 14, 1185–1195. [Google Scholar] [CrossRef] [PubMed]
  33. Gottardi, M.; Sperotto, A.; Di Rorà, A.G.L.; Padella, A.; Cangini, D.; Giannini, M.B.; Simonetti, G.; Martinelli, G.; Cerchione, C. Gemtuzumab ozogamicin in acute myeloid leukemia: Past, present and future. Minerva Med. 2020, 111, 395–410. [Google Scholar] [CrossRef] [PubMed]
  34. Bross, P.F.; Beitz, J.; Chen, G.; Chen, X.H.; Duffy, E.; Kieffer, L.; Roy, S.; Sridhara, R.; Rahman, A.; Williams, G.; et al. Approval Summary: Gemtuzumab ozogamicin in relapsed acute myeloid leukemia. Clin. Cancer Res. 2001, 7, 1490–1496. [Google Scholar] [PubMed]
  35. Laszlo, G.S.; Estey, E.H.; Walter, R.B. The past and future of CD33 as therapeutic target in acute myeloid leukemia. Blood Rev. 2014, 28, 143–153. [Google Scholar] [CrossRef]
  36. Amadori, S.; Suciu, S.; Selleslag, D.; Stasi, R.; Alimena, G.; Baila, L.; Rizzoli, V.; Borlenghi, E.; Gaidano, G.; Magro, D.; et al. Randomized trial of two schedules of low-dose gemtuzumab ozogamicin as induction monotherapy for newly diagnosed acute myeloid leukaemia in older patients not considered candidates for intensive chemotherapy. A phase II study of the EORTC and GIMEMA leuka. Br. J. Haematol. 2010, 149, 376–382. [Google Scholar] [CrossRef] [PubMed]
  37. Amadori, S.; Venditti, A.; Voso, M.T.; Annino, L.; De Fabritiis, P.; Alimena, G.; Mancini, M.; Paoloni, F.; Vignetti, M.; Fazi, P.; et al. Gemtuzumab ozogamicin versus best supportive care in older patients with newly diagnosed acute myeloid leukemia unsuitable for intensive chemotherapy: Results of the randomized phase III EORTC-GIMEMA AML-19 Trial. J. Clin. Oncol. 2016, 34, 972–979. [Google Scholar] [CrossRef]
  38. Zwaan, C.M.; Reinhardt, D.; Corbacioglu, S.; Van Wering, E.R.; Bökkerink, J.P.M.; Tissing, W.J.E.; Samuelsson, U.; Feingold, J.; Creutzig, U.; Kaspers, G.J.L. Gemtuzumab ozogamicin: First clinical experiences in children with relapsed/refractory acute myeloid leukemia treated on compassionate-use basis. Blood 2003, 101, 3868–3871. [Google Scholar] [CrossRef] [Green Version]
  39. Arceci, R.J.; Sande, J.; Lange, B.; Shannon, K.; Franklin, J.; Hutchinson, R.; Vik, T.A.; Flowers, D.; Aplenc, R.; Berger, M.S.; et al. Safety and efficacy of gemtuzumab ozogamicin in pediatric patients with advanced CD33+ acute myeloid leukemia. Blood 2005, 106, 1183–1188. [Google Scholar] [CrossRef] [Green Version]
  40. Zwaan, C.M.; Reinhardt, D.; Zimmerman, M.; Hasle, H.; Stary, J.; Stark, B.; Dworzak, M.; Creutzig, U.; Kaspers, G.J.L. Salvage treatment for children with refractory first or second relapse of acute myeloid leukaemia with gemtuzumab ozogamicin: Results of a phase II study. Br. J. Haematol. 2010, 148, 768–776. [Google Scholar] [CrossRef]
  41. Malfuson, J.V.; Konopacki, J.; Thepenier, C.; Eddou, H.; Foissaud, V.; De Revel, T. Fractionated doses of gemtuzumab ozogamicin combined with 3+7 induction chemotherapy as salvage treatment for young patients with acute myeloid leukemia in first relapse. Ann. Hematol. 2012, 91, 1871–1877. [Google Scholar] [CrossRef]
  42. Kell, W.J.; Burnett, A.K.; Chopra, R.; Yin, J.A.L.; Clark, R.E.; Rohatiner, A.; Culligan, D.; Hunter, A.; Prentice, A.G.; Milligan, D.W. A feasibility study of simultaneous administration of gemtuzumab ozogamicin with intensive chemotherapy in induction and consolidation in younger patients with acute myeloid leukemia. Blood 2003, 102, 4277–4283. [Google Scholar] [CrossRef] [PubMed]
  43. Burnett, A.K.; Hills, R.K.; Milligan, D.; Kjeldsen, L.; Kell, J.; Russell, N.H.; Yin, J.A.L.; Hunter, A.; Goldstone, A.H.; Wheatley, K. Identification of Patients with Acute Myeloblastic Leukemia Who Benefit from the Addition of Gemtuzumab Ozogamicin: Results of the MRC AML15 Trial. J. Clin. Oncol. 2011, 29, 369–377. [Google Scholar] [CrossRef] [PubMed]
  44. Burnett, A.K.; Russell, N.H.; Hills, R.K.; Kell, J.; Freeman, S.; Kjeldsen, L.; Hunter, A.E.; Yin, J.; Craddock, C.F.; Dufva, I.H.; et al. Addition of gemtuzumab ozogamicin to induction chemotherapy improves survival in older patients with acute myeloid leukemia. J. Clin. Oncol. 2012, 30, 3924–3931. [Google Scholar] [CrossRef] [PubMed]
  45. Farhat, H.; Reman, O.; Raffoux, E.; Berthon, C.; Pautas, C.; Kammoun, L.; Chantepie, S.; Gardin, C.; Rousselot, P.; Chevret, S.; et al. Fractionated doses of gemtuzumab ozogamicin with escalated doses of daunorubicin and cytarabine as first acute myeloid leukemia salvage in patients aged 50–70-year old: A phase 1/2 study of the acute leukemia French association. Am. J. Hematol. 2012, 87, 62–65. [Google Scholar] [CrossRef] [PubMed]
  46. Pilorge, S.; Rigaudeau, S.; Rabian, F.; Sarkozy, C.; Taksin, A.L.; Farhat, H.; Merabet, F.; Ghez, S.; Raggueneau, V.; Terré, C.; et al. Fractionated gemtuzumab ozogamicin and standard dose cytarabine produced prolonged second remissions in patients over the age of 55 years with acute myeloid leukemia in late first relapse. Am. J. Hematol. 2014, 89, 399–403. [Google Scholar] [CrossRef] [PubMed]
  47. Castaigne, S.; Pautas, C.; Terré, C.; Raffoux, E.; Bordessoule, D.; Bastie, J.N.; Legrand, O.; Thomas, X.; Turlure, P.; Reman, O.; et al. Effect of gemtuzumab ozogamicin on survival of adult patients with de-novo acute myeloid leukaemia (ALFA-0701): A randomised, open-label, phase 3 study. Lancet 2012, 379, 1508–1516. [Google Scholar] [CrossRef]
  48. Lambert, J.; Pautas, C.; Terré, C.; Raffoux, E.; Turlure, P.; Caillot, D.; Legrand, O.; Thomas, X.; Gardin, C.; Gogat-Marchant, K.; et al. Gemtuzumab ozogamicin for de novo acute myeloid leukemia: Final efficacy and safety updates from the open-label, phase III ALFA-0701 trial. Haematologica 2019, 104, 113–119. [Google Scholar] [CrossRef] [Green Version]
  49. Hills, R.K.; Castaigne, S.; Appelbaum, F.R.; Delaunay, J.; Petersdorf, S.; Othus, M.; Estey, E.H.; Dombret, H.; Chevret, S.; Ifrah, N.; et al. The addition of gemtuzumab ozogamicin to induction chemotherapy in adult patients with acute myeloid leukaemia: A meta-analysis of individual patient data from randomised controlled trials. Lancet Oncol. 2014, 15, 986–996. [Google Scholar] [CrossRef] [Green Version]
  50. Burnett, A.; Cavenagh, J.; Russell, N.; Hills, R.; Kell, J.; Jones, G.; Nielsen, O.J.; Khwaja, A.; Thomas, I.; Clark, R. Defining the dose of gemtuzumab ozogamicin in combination with induction chemotherapy in acute myeloid leukemia: A comparison of 3 mg/m2 with 6 mg/m2 in the NCRI AML17 trial. Haematologica 2016, 101, 724–731. [Google Scholar] [CrossRef] [Green Version]
  51. Maniecki, M.B.; Hasle, H.; Friis-Hansen, L.; Lausen, B.; Nielsen, O.J.; Bendix, K.; Moestrup, S.K.; Møller, H.J. Impaired CD163-mediated hemoglobin-scavenging and severe toxic symptoms in patients treated with gemtuzumab ozogamicin. Blood 2008, 112, 1510–1514. [Google Scholar] [CrossRef] [Green Version]
  52. Tsimberidou, A.; Estey, E.; Cortes, J.; Thomas, D.; Faderl, S.; Verstovsek, S.; Garcia-Manero, G.; Keating, M.; Albitar, M.; O’Brien, S.; et al. Gemtuzumab, fludarabine, cytarabine, and cyclosporine in patients with newly diagnosed acute myelogenous leukemia or high-risk myelodysplastic syndromes. Cancer 2003, 97, 1481–1487. [Google Scholar] [CrossRef] [PubMed]
  53. Larson, R.A.; Sievers, E.L.; Stadtmauer, E.A.; Löwenberg, B.; Estey, E.H.; Dombret, H.; Theobald, M.; Voliotis, D.; Bennett, J.M.; Richte, M.; et al. Final report of the efficacy and safety of gemtuzumab ozogamicin (Mylotarg) in patients with CD33-positive acute myeloid leukemia in first recurrence. Cancer 2005, 104, 1442–1452. [Google Scholar] [CrossRef]
  54. Ho, V.T.; Martin, A.S.; Pérez, W.S.; Steinert, P.; Zhang, M.J.; Chirnomas, D.; Hoang, C.J.; Loberiza, F.R.; Saber, W. Prior Gemtuzumab Ozogamicin Exposure in Adults with Acute Myeloid Leukemia Does Not Increase Hepatic Veno-Occlusive Disease Risk after Allogeneic Hematopoietic Cell Transplantation: A Center for International Blood and Marrow Transplant Research Analysis. Biol. Blood Marrow Transplant. 2020, 26, 884–892. [Google Scholar] [CrossRef] [PubMed]
  55. Selby, C.; Yacko, L.R.; Glode, A.E. Gemtuzumab Ozogamicin: Back Again. J. Adv. Pract. Oncol. 2019, 10, 68–82. [Google Scholar] [CrossRef] [PubMed]
  56. De Witte, T.; Suciu, S.; Meert, L.; Halkes, C.; Selleslag, D.; Bron, D.; Amadori, S.; Willemze, R.; Muus, P.; Baron, F. Idarubicin and cytarabine in combination with gemtuzumab ozogamicin (IAGO) for untreated patients with high-risk MDS or AML evolved from MDS: A phase II study from the EORTC and GIMEMA Leukemia Groups (protocol 06013). Ann. Hematol. 2015, 94, 1981–1989. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  57. Wynne, J.; Wright, D.; Stock, W. Inotuzumab: From preclinical development to success in B-cell acute lymphoblastic leukemia. Blood Adv. 2019, 3, 96–104. [Google Scholar] [CrossRef]
  58. Richardson, P.G.; Corbacioglu, S. Veno-occlusive disease/sinusoidal obstruction syndrome in patients with prior gemtuzumab ozogamicin: Literature analysis of survival after defibrotide treatment. Blood Cancer J. 2020, 10. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  59. Van Der Reijden, B.A.; Simons, A.; Luiten, E.; Van Der Poel, S.C.; Hogenbirk, P.E.; Tönnissen, E.; Valk, P.J.M.; Löwenberg, B.; De Greef, G.E.; Breuning, M.H.; et al. Minimal residual disease quantification in patients with acute myeloid leukaemia and inv(16)/CBFB-MYH11 gene fusion. Br. J. Haematol. 2002, 118, 411–418. [Google Scholar] [CrossRef]
  60. Pautas, C.; Raffoux, E.; Lambert, J.; Legrand, O.; Chantepie, S.; Gastaud, L.; Marolleau, J.P.; Thomas, X.; Turlure, P.; Benner, R.J.; et al. Outcomes following hematopoietic stem cell transplantation in patients treated with standard chemotherapy with or without gemtuzumab ozogamicin for acute myeloid leukemia. Bone Marrow Transplant. 2021, 56, 1474–1477. [Google Scholar] [CrossRef]
  61. Debureaux, P.E.; Labopin, M.; Mamez, A.C.; Lapusan, S.; Isnard, F.; Adaeva, R.; Bonnin, A.; Hirsch, P.; Delhommeau, F.; Battipaglia, G.; et al. Fractionated gemtuzumab ozogamicin in association with high dose chemotherapy: A bridge to allogeneic stem cell transplantation in refractory and relapsed acute myeloid leukemia. Bone Marrow Transplant. 2020, 55, 452–460. [Google Scholar] [CrossRef] [PubMed]
  62. Paubelle, E.; Ducastelle-Leprêtre, S.; Labussière-Wallet, H.; Nicolini, F.E.; Barraco, F.; Plesa, A.; Salles, G.; Wattel, E.; Thomas, X. Fractionated gemtuzumab ozogamicin combined with intermediate-dose cytarabine and daunorubicin as salvage therapy in very high-risk AML patients: A bridge to reduced intensity conditioning transplant? Ann. Hematol. 2017, 96, 363–371. [Google Scholar] [CrossRef] [PubMed]
  63. Sumiyoshi, R.; Tashiro, H.; Saito, S.; Matsuo, T.; Yamamoto, T.; Matsumoto, K.; Ooi, J.; Shirafuji, N. Gemtuzumab ozogamicin monotherapy prior to stem cell infusion induces sustained remission in a relapsed acute myeloid leukemia patient after allogeneic stem cell transplantation: A case report. Medicine 2020, 99, e22064. [Google Scholar] [CrossRef] [PubMed]
  64. Mosna, F.; Papayannidis, C.; Martinelli, G.; Di Bona, E.; Bonalumi, A.; Tecchio, C.; Candoni, A.; Capelli, D.; Piccin, A.; Forghieri, F.; et al. Complex karyotype, older age, and reduced first-line dose intensity determine poor survival in core binding factor acute myeloid leukemia patients with long-term follow-up. Am. J. Hematol. 2015, 90, 515–523. [Google Scholar] [CrossRef] [Green Version]
  65. Terwijn, M.; Kelder, A.; Huijgens, P.C.; Dräger, A.M.; Oussoren, Y.J.M.; Scholten, W.J.; Snel, A.N.; Ossenkoppele, G.J.; Schuurhuis, G.J.; Biemond, B.J.; et al. High prognostic impact of flow cytometric minimal residual disease detection in acute myeloid leukemia: Data from the HOVON/SAKK AML 42A study. J. Clin. Oncol. 2013, 31, 3889–3897. [Google Scholar] [CrossRef]
  66. Walter, R.B.; Gooley, T.A.; Wood, B.L.; Milano, F.; Fang, M.; Sorror, M.L.; Estey, E.H.; Salter, A.I.; Lansverk, E.; Chien, J.W.; et al. Impact of pretransplantation minimal residual disease, as detected by multiparametric flow cytometry, on outcome of myeloablative hematopoietic cell transplantation for acute myeloid leukemia. J. Clin. Oncol. 2011, 29, 1190–1197. [Google Scholar] [CrossRef] [Green Version]
  67. Pollard, J.A.; Alonzo, T.A.; Loken, M.; Gerbing, R.B.; Ho, P.A.; Bernstein, I.D.; Raimondi, S.C.; Hirsch, B.; Franklin, J.; Walter, R.B.; et al. Correlation of CD33 expression level with disease characteristics and response to gemtuzumab ozogamicin containing chemotherapy in childhood AML. Blood 2012, 119, 3705–3711. [Google Scholar] [CrossRef] [Green Version]
  68. Genthon, A.; Brissot, E.; Malard, F.; Van de Wyngaert, Z.; Bonnin, A.; Banet, A.; Marjanovic, Z.; Ikhlef, S.; Lapusan, S.; Sestili, S.; et al. Gemtuzumab Ozogamicin Combined With Intensive Chemotherapy in Patients With Acute Myeloid Leukemia Relapsing After Allogenic Stem Cell Transplantation. Clin. Lymphoma Myeloma Leuk. 2020, 20, 791–796. [Google Scholar] [CrossRef]
  69. Satwani, P.; Bhatia, M.; Garvin, J.H.; George, D.; Dela Cruz, F.; Le Gall, J.; Jin, Z.; Schwartz, J.; Duffy, D.; Van de Ven, C.; et al. A Phase I Study of Gemtuzumab Ozogamicin (GO) in Combination with Busulfan and Cyclophosphamide (Bu/Cy) and Allogeneic Stem Cell Transplantation in Children with Poor-Risk CD33+ AML: A New Targeted Immunochemotherapy Myeloablative Conditioning (MAC) Regim. Biol. Blood Marrow Transplant. 2012, 18, 324–329. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  70. Penel-Page, M.; Plesa, A.; Girard, S.; Marceau-Renaut, A.; Renard, C.; Bertrand, Y. Association of fludarabin, cytarabine, and fractioned gemtuzumab followed by hematopoietic stem cell transplantation for first-line refractory acute myeloid leukemia in children: A single-center experience. Pediatr. Blood Cancer 2020, 67, e28305. [Google Scholar] [CrossRef] [PubMed]
  71. Toyama, D.; Matsuno, R.; Sugishita, Y.; Kaneko, R.; Okamoto, N.; Koganesawa, M.; Fujita, S.; Akiyama, K.; Isoyama, K.; Yamamoto, S. Successful Treatment of Pediatric Refractory/Relapsed AML with KIR-Ligand-Mismatched Cord Blood Transplant after FLAG-IDA Reinduction Therapy with or without the GO Regimen. Case Rep. Hematol. 2020. [Google Scholar] [CrossRef]
  72. Niktoreh, N.; Lerius, B.; Zimmermann, M.; Gruhn, B.; Escherich, G.; Bourquin, J.P.; Dworzak, M.; Sramkova, L.; Rossig, C.; Creutzig, U.; et al. Gemtuzumab ozogamicin in children with relapsed or refractory acute myeloid leukemia: A report by Berlin-Frankfurt-Münster study group. Haematologica 2019, 104, 120–127. [Google Scholar] [CrossRef] [Green Version]
  73. O’Hear, C.; Inaba, H.; Pounds, S.; Shi, L.; Dahl, G.; Paul Bowman, W.; Taub, J.W.; Pui, C.H.; Ribeiro, R.C.; Coustan-Smith, E.; et al. Gemtuzumab ozogamicin can reduce minimal residual disease in patients with childhood acute myeloid leukemia. Cancer 2013, 119, 4036–4043. [Google Scholar] [CrossRef] [Green Version]
  74. Roman, E.; Cooney, E.; Harrison, L.; Militano, O.; Wolownik, K.; Hawks, R.; Foley, S.; Satwani, P.; Unal, E.; Bhatia, M.; et al. Preliminary results of the safety of immunotherapy with gemtuzumab ozogamicin following reduced intensity allogeneic stem cell transplant in children with CD33+ acute myeloid leukemia. Clin. Cancer Res. 2005, 11, 7164s–7170s. [Google Scholar] [CrossRef] [Green Version]
  75. Zahler, S.; Bhatia, M.; Ricci, A.; Roy, S.; Morris, E.; Harrison, L.; van de Ven, C.; Fabricatore, S.; Wolownik, K.; Cooney-Qualter, E.; et al. A Phase I Study of Reduced-Intensity Conditioning and Allogeneic Stem Cell Transplantation Followed by Dose Escalation of Targeted Consolidation Immunotherapy with Gemtuzumab Ozogamicin in Children and Adolescents with CD33+ Acute Myeloid Leukemia. Biol. Blood Marrow Transplant. 2016, 22, 698–704. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  76. Pollard, J.A.; Loken, M.; Gerbing, R.B.; Raimondi, S.C.; Hirsch, B.A.; Aplenc, R.; Bernstein, I.D.; Gamis, A.S.; Alonzo, T.A.; Meshinchi, S. CD33 expression and its association with gemtuzumab ozogamicin response: Results from the randomized phase III children’s oncology group trial AAML0531. J. Clin. Oncol. 2016, 34, 747–755. [Google Scholar] [CrossRef] [PubMed]
  77. Van Der Velden, V.H.J.; Te Marvelde, J.G.; Hoogeveen, P.G.; Bernstein, I.D.; Houtsmuller, A.B.; Berger, M.S.; Van Dongen, J.J.M. Targeting of the CD33-calicheamicin immunoconjugate Mylotarg (CMA-676) in acute myeloid leukemia: In vivo and in vitro saturation and internalization by leukemic and normal myeloid cells. Blood 2001, 97, 3197–3204. [Google Scholar] [CrossRef]
  78. Jager, E.; Van der Velden, V.H.J.; Te Marvelde, J.G.; Walter, R.B.; Agur, Z.; Vainstein, V. Targeted drug delivery by gemtuzumab ozogamicin: Mechanism-Based mathematical model for treatment strategy improvement and therapy individualization. PLoS ONE 2011, 6. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  79. Walter, R.B.; Raden, B.W.; Kamikura, D.M.; Cooper, J.A.; Bernstein, I.D. Influence of CD33 expression levels and ITIM-dependent internalization on gemtuzumab ozogamicin-induced cytotoxicity. Blood 2005, 105, 1295–1302. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  80. Olombel, G.; Guerin, E.; Guy, J.; Perrot, J.Y.; Dumezy, F.; de Labarthe, A.; Bastie, J.N.; Legrand, O.; Raffoux, E.; Plesa, A.; et al. The level of blast CD33 expression positively impacts the effect of gemtuzumab ozogamicin in patients with acute myeloid leukemia. Blood 2016, 127, 2157–2160. [Google Scholar] [CrossRef] [PubMed]
  81. Khan, N.; Hills, R.K.; Virgo, P.; Couzens, S.; Clark, N.; Gilkes, A.; Richardson, P.; Knapper, S.; Grimwade, D.; Russell, N.H.; et al. Expression of CD33 is a predictive factor for effect of gemtuzumab ozogamicin at different doses in adult acute myeloid leukaemia. Leukemia 2017, 31, 1059–1068. [Google Scholar] [CrossRef] [Green Version]
  82. Lamba, J.K.; Chauhan, L.; Shin, M.; Loken, M.R.; Pollard, J.A.; Wang, Y.C.; Ries, R.E.; Aplenc, R.; Hirsch, B.A.; Raimondi, S.C.; et al. CD33 splicing polymorphism determines gemtuzumab ozogamicin response in de novo acute myeloid leukemia: Report from randomized phase III children’s oncology group trial AAML0531. J. Clin. Oncol. 2017, 35, 674–2682. [Google Scholar] [CrossRef]
  83. Teich, K.; Krzykalla, J.; Kapp-Schwoerer, S.; Gaidzik, V.I.; Schlenk, R.F.; Paschka, P.; Weber, D.; Fiedler, W.; Kühn, M.W.M.; Schroeder, T.; et al. Cluster of differentiation 33 single nucleotide polymorphism rs12459419 is a predictive factor in patients with nucleophosmin1 mutated acute myeloid leukemia receiving gemtuzumab ozogamicin. Haematologica 2021. [Google Scholar] [CrossRef] [PubMed]
  84. Raj, T.; Ryan, K.J.; Replogle, J.M.; Chibnik, L.B.; Rosenkrantz, L.; Tang, A.; Rothamel, K.; Stranger, B.E.; Bennett, D.A.; Evans, D.A.; et al. CD33: Increased inclusion of exon 2 implicates the Ig V-set domain in Alzheimer’s disease susceptibility. Hum. Mol. Genet. 2014, 23, 2729–2736. [Google Scholar] [CrossRef] [Green Version]
  85. Mortland, L.; Alonzo, T.A.; Walter, R.B.; Gerbing, R.B.; Mitra, A.K.; Pollard, J.A.; Loken, M.R.; Hirsch, B.; Raimondi, S.; Franklin, J.; et al. Clinical significance of CD33 nonsynonymous single-nucleotide polymorphisms in pediatric patients with acute myeloid leukemia treated with gemtuzumab-ozogamicin-containing chemotherapy. Clin. Cancer Res. 2013, 19, 1620–1627. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  86. Lamba, J.K.; Voigt, A.P.; Chauhan, L.; Shin, M.; Aplenc, R.; Eidenschink Brodersen, L.; Gamis, A.S.; Meshinchi, S.; Loken, M.R. CD33 splicing SNP regulates expression levels of CD33 in normal regenerating monocytes in AML patients. Leuk. Lymphoma 2018, 59, 2250–2253. [Google Scholar] [CrossRef]
  87. Malik, M.; Simpson, J.F.; Parikh, I.; Wilfred, B.R.; Fardo, D.W.; Nelson, P.T.; Estus, S. CD33 Alzheimer’s risk-altering polymorphism, CD33 expression, and exon 2 splicing. J. Neurosci. 2013, 33, 13320–13325. [Google Scholar] [CrossRef] [Green Version]
  88. Chauhan, L.; Shin, M.; Wang, Y.-C.; Loken, M.; Pollard, J.; Aplenc, R.; Hirsch, B.A.; Raimondi, S.; Ries, R.E.; Bernstein, I.D.; et al. CD33_PGx6_Score Predicts Gemtuzumab Ozogamicin Response in Childhood Acute Myeloid Leukemia: A Report From the Children’s Oncology Group. JCO Precis. Oncol. 2019, 3, 1–15. [Google Scholar] [CrossRef]
  89. Renneville, A.; Abdelali, R.B.; Chevret, S.; Nibourel, O.; Cheok, M.; Pautas, C.; Duléry, R.; Boyer, T.; Cayuela, J.M.; Hayette, S.; et al. Clinical impact of gene mutations and lesions detected by SNP-array karyotyping in acute myeloid leukemia patients in the context of gemtuzumab ozogamicin treatment: Results of the ALFA-0701 trial. Oncotarget 2014, 5, 916–932. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  90. Krupka, C.; Kufer, P.; Kischel, R.; Zugmaier, G.; Bögeholz, J.; Köhnke, T.; Lichtenegger, F.S.; Schneider, S.; Metzeler, K.H.; Fiegl, M.; et al. CD33 target validation and sustained depletion of AML blasts in long-term cultures by the bispecific T-cell-engaging antibody AMG 330. Blood 2014, 123, 356–365. [Google Scholar] [CrossRef] [PubMed]
  91. Walter, R.B.; Appelbaum, F.R.; Estey, E.H.; Bernstein, I.D. Acute myeloid leukemia stem cells and CD33-targeted immunotherapy. Blood 2012, 119, 6198–6208. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  92. Borthakur, G.; Cortes, J.E.; Estey, E.E.; Jabbour, E.; Faderl, S.; O’Brien, S.; Garcia-Manero, G.; Kadia, T.M.; Wang, X.; Patel, K.; et al. Gemtuzumab ozogamicin with fludarabine, cytarabine, and granulocyte colony stimulating factor (FLAG-GO) as front-line regimen in patients with core binding factor acute myelogenous leukemia. Am. J. Hematol. 2014, 89, 964–968. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Tarlock, K.; Alonzo, T.A.; Wang, Y.C.; Gerbing, R.B.; Ries, R.; Loken, M.R.; Pardo, L.; Hylkema, T.; Joaquin, J.; Sarukkai, L.; et al. Functional properties of KIT mutations are associated with differential clinical outcomes and response to targeted therapeutics in CBF acute myeloid leukemia. Clin. Cancer Res. 2019, 25, 5038–5048. [Google Scholar] [CrossRef] [PubMed]
  94. Borthakur, G.M.; Cortes, J.E.; Ravandi, F.; Garcia-Manero, G.; Kadia, T.M.; Jabbour, E.; Patel, K.; Issa, G.C.; Daver, N.G.; Ohanian, M.N.; et al. Fludarabine, Cytarabine, G-CSF and Gemtuzumab Ozogamicin (FLAG-GO) Regimen Results in Better Molecular Response and Relapse-Free Survival in Core Binding Factor Acute Myeloid Leukemia Than FLAG and Idarubicin (FLAG-Ida). Blood 2019, 134. [Google Scholar] [CrossRef]
  95. Döhner, H.; Estey, E.; Grimwade, D.; Amadori, S.; Appelbaum, F.R.; Büchner, T.; Dombret, H.; Ebert, B.L.; Fenaux, P.; Larson, R.A.; et al. Diagnosis and management of AML in adults: 2017 ELN recommendations from an international expert panel. Blood 2017, 129, 424–447. [Google Scholar] [CrossRef] [Green Version]
  96. Balgobind, B.V.; Zwaan, C.M.; Pieters, R.; Van Den Heuvel-Eibrink, M.M. The heterogeneity of pediatric MLL-rearranged acute myeloid leukemia. Leukemia 2011, 25, 1239–1248. [Google Scholar] [CrossRef] [Green Version]
  97. Muñoz, L.; Nomdedéu, J.F.; Villamor, N.; Guardia, R.; Colomer, D.; Ribera, J.M.; Torres, J.P.; Berlanga, J.J.; Fernández, C.; Llorente, A.; et al. Acute myeloid leukemia with MLL rearrangements: Clinicobiological features, prognostic impact and value of flow cytometry in the detection of residual leukemic cells. Leukemia 2003, 17, 76–82. [Google Scholar] [CrossRef] [Green Version]
  98. Tamai, H.; Shioi, Y.; Yamaguchi, H.; Okabe, M.; Wakita, S.; Mizuki, T.; Nakayama, K.; Inokuchi, K.; Tajika, K.; Dan, K. Treatment of relapsed acute myeloid leukemia with MLL/AF6 fusion after allogeneic hematopoietic stem cell transplantation with gemtuzumab ozogamicin with a long interval followed by donor lymphocyte infusion. Leukemia 2008, 22, 1273–1274. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  99. Asano, H.; Yamamoto, G.; Hosoi, M.; Takahashi, T.; Hangaishi, A.; Kurokawa, M. Complete molecular remission in refractory acute myeloid leukemia with MLL/AF9 treated with gemtuzumab ozogamicin. Leuk. Res. 2010, 34. [Google Scholar] [CrossRef]
  100. Pollard, J.A.; Guest, E.; Alonzo, T.A.; Gerbing, R.B.; Loken, M.R.; Brodersen, L.E.; Kolb, E.A.; Aplenc, R.; Meshinchi, S.; Raimondi, S.C.; et al. Gemtuzumab Ozogamicin Improves Event-Free Survival and Reduces Relapse in Pediatric KMT2A-Rearranged AML: Results From the Phase III Children’s Oncology Group Trial AAML0531. J. Clin. Oncol. 2021. [Google Scholar] [CrossRef]
  101. Legrand, O.; Zompi, S.; Perrot, J.Y.; Faussat, A.M.; Benderra, Z.; Chaoui, D.; Marie, J.P. P-glycoprotein and multidrug resistance associated pr otein-1 activity in 132 acute myeloid leukemias according to FAB subtypes and cytogenetic risk groups. Haematologica 2004, 89, 34–41. [Google Scholar] [PubMed]
  102. Seedhouse, C.H.; Grundy, M.; White, P.; Li, Y.; Fisher, J.; Yakunina, D.; Moorman, A.V.; Hoy, T.; Russell, N.; Burnett, A.; et al. Sequential influences of leukemia-specific and genetic factors on P-glycoprotein expression in blasts from 817 patients entered into the National Cancer Research Network acute myeloid leukemia 14 and 15 trials. Clin. Cancer Res. 2007, 13, 7059–7066. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  103. Candoni, A.; Papayannidis, C.; Martinelli, G.; Simeone, E.; Gottardi, M.; Iacobucci, I.; Gherlinzoni, F.; Visani, G.; Baccarani, M.; Fanin, R. Flai (fludarabine, cytarabine, idarubicin) plus low-dose Gemtuzumab Ozogamicin as induction therapy in CD33-positive AML: Final results and long term outcome of a phase II multicenter clinical trial. Am. J. Hematol. 2018, 93, 655–663. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. Fournier, E.; Duployez, N.; Ducourneau, B.; Raffoux, E.; Turlure, P.; Caillot, D.; Thomas, X.; Marceau-Renaut, A.; Chantepie, S.; Malfuson, J.V.; et al. Mutational profile and benefit of gemtuzumab ozogamicin in acute myeloid leukemia. Blood 2020, 135, 542–546. [Google Scholar] [CrossRef]
  105. De Propris, M.S.; Raponi, S.; Diverio, D.; Milani, M.L.; Meloni, G.; Falini, B.; Foà, R.; Guarini, A. High CD33 expression levels in acute myeloid leukemia cells carrying the nucleophosmin (NPM1) mutation. Haematologica 2011, 96, 1548–1551. [Google Scholar] [CrossRef] [Green Version]
  106. Ehninger, A.; Kramer, M.; Röllig, C.; Thiede, C.; Bornhäuser, M.; Von Bonin, M.; Wermke, M.; Feldmann, A.; Bachmann, M.; Ehninger, G.; et al. Distribution and levels of cell surface expression of CD33 and CD123 in acute myeloid leukemia. Blood Cancer J. 2014, 4. [Google Scholar] [CrossRef] [Green Version]
  107. Schlenk, R.F.; Paschka, P.; Krzykalla, J.; Weber, D.; Kapp-Schwoerer, S.; Gaidzik, V.I.; Leis, C.; Fiedler, W.; Kindler, T.; Schroeder, T.; et al. Gemtuzumab ozogamicin in NPM1-mutated acute myeloid leukemia: Early results from the prospective randomized AMLSG 09-09 Phase III study. J. Clin. Oncol. 2020, 38, 623–632. [Google Scholar] [CrossRef]
  108. Tarlock, K.; Alonzo, T.A.; Gerbing, R.B.; Raimondi, S.C.; Hirsch, B.A.; Sung, L.; Pollard, J.A.; Aplenc, R.; Loken, M.R.; Gamis, A.S.; et al. Gemtuzumab ozogamicin reduces relapse risk in FLT3/ITD acute myeloid leukemia: A report from the children’s oncology group. Clin. Cancer Res. 2016, 22, 1951–1957. [Google Scholar] [CrossRef] [Green Version]
  109. Zhang, H.; Nakauchi, Y.; Köhnke, T.; Stafford, M.; Bottomly, D.; Thomas, R.; Wilmot, B.; McWeeney, S.K.; Majeti, R.; Tyner, J.W. Integrated analysis of patient samples identifies biomarkers for venetoclax efficacy and combination strategies in acute myeloid leukemia. Nat. Cancer 2020, 1, 826–839. [Google Scholar] [CrossRef]
  110. Itzykson, R.; Duployez, N.; Fasan, A.; Decool, G.; Marceau-Renaut, A.; Meggendorfer, M.; Jourdan, E.; Petit, A.; Lapillonne, H.; Micol, J.B.; et al. Clonal interference of signaling mutations worsens prognosis in core-binding factor acute myeloid leukemia. Blood 2018, 132, 187–196. [Google Scholar] [CrossRef]
  111. Balaian, L.; Zhong, R.K.; Ball, E.D. The inhibitory effect of anti-CD33 monoclonal antibodies on AML cell growth correlates with Syk and/or ZAP-70 expression. Exp. Hematol. 2003, 31, 363–371. [Google Scholar] [CrossRef]
  112. Bouvier, A.; Hamel, J.; Delaunay, J.; Delabesse, E.; Dumas, P.; Ledoux, M.; Peterlin, P.; Luquet, I.; Roth Guepin, G.; Bulabois, C.E.; et al. Molecular classification and prognosis in younger adults with acute myeloid leukemia and intermediate-risk cytogenetics treated or not by gemtuzumab ozogamycin: Final results of the GOELAMS/FILO acute myeloid leukemia 2006-intermediate-risk trial. Eur. J. Haematol. 2021. [Google Scholar] [CrossRef]
  113. Zhang, N.; Zhang, Y.; Zhang, P.; Lou, S.; Chen, Y.; Li, H.; Zeng, H.; Shen, Y.; Deng, J. Overexpression of annexin A5 might guide the gemtuzumab ozogamicin treatment choice in patients with pediatric acute myeloid leukemia. Ther. Adv. Med. Oncol. 2020, 12. [Google Scholar] [CrossRef]
  114. Matsumoto, T.; Jimi, S.; Hara, S.; Takamatsu, Y.; Suzumiya, J.; Tamura, K. Importance of inducible multidrug resistance 1 expression in HL-60 cells resistant to gemtuzumab ozogamicin. Leuk. Lymphoma 2012, 53, 1399–1405. [Google Scholar] [CrossRef]
  115. Walter, R.B.; Raden, B.W.; Hong, T.C.; Flowers, D.A.; Bernstein, I.D.; Linenberger, M.L. Multidrug resistance protein attenuates gemtuzumab ozogamicin-induced cytotoxicity in acute myeloid leukemia cells. Blood 2003, 102, 1466–1473. [Google Scholar] [CrossRef] [Green Version]
  116. Matsui, H.; Takeshita, A.; Naito, K.; Shinjo, K.; Shigeno, K.; Maekawa, M.; Yamakawa, Y.; Tanimoto, M.; Kobayashi, M.; Ohnishi, K.; et al. Reduced effect of gemtuzumab ozogamicin (CMA-676) on P-glycoprotein and/or CD34-positive leukemia cells and its restoration by multidrug resistance modifiers. Leukemia 2002, 16, 813–819. [Google Scholar] [CrossRef] [Green Version]
  117. Walter, R.B.; Raden, B.W.; Thompson, J.; Flowers, D.A.; Kiem, H.P.; Bernstein, I.D.; Linenberger, M.L. Breast cancer resistance protein (BCRP/ABCG2) does not confer resistance to gemtuzumab ozogamicin and calicheamicin-γ1 in acute myeloid leukemia cells. Leukemia 2004, 18, 1914–1917. [Google Scholar] [CrossRef] [PubMed]
  118. Walter, R.B.; Gooley, T.A.; Van Der Velden, V.H.J.; Loken, M.R.; Van Dongen, J.J.M.; Flowers, D.A.; Bernstein, I.D.; Appelbaum, F.R. CD33 expression and P-glycoprotein-mediated drug efflux inversely correlate and predict clinical outcome in patients with acute myeloid leukemia treated with gemtuzumab ozogamicin monotherapy. Blood 2007, 109, 4168–4170. [Google Scholar] [CrossRef] [PubMed]
  119. Van Den Heuvel-Eibrink, M.M.; Van Der Holt, B.; Te Boekhorst, P.A.W.; Pieters, R.; Schoester, M.; Löwenberg, B.; Sonneveld, P. MDR 1 expression is an independent prognostic factor for response and survival in de novo acute myeloid leukaemia. Br. J. Haematol. 1997, 99, 76–83. [Google Scholar] [CrossRef] [PubMed]
  120. Leith, C.P.; Kopecky, K.J.; Chen, I.M.; Eijdems, L.; Slovak, M.L.; McConnell, T.S.; Head, D.R.; Weick, J.; Grever, M.R.; Appelbaum, F.R.; et al. Frequency and clinical significance of the expression of the multidrug resistance proteins MDR1/P-glycoprotein, MRP1, and LRP in acute myeloid leukemia. A Southwest Oncology Group study. Blood 1999, 94, 1086–1099. [Google Scholar] [CrossRef] [PubMed]
  121. Linenberger, M.L.; Hong, T.; Flowers, D.; Sievers, E.L.; Gooley, T.A.; Bennett, J.M.; Berger, M.S.; Leopold, L.H.; Appelbaum, F.R.; Bernstein, I.D. Multidrug-resistance phenotype and clinical responses to gemtuzumab ozogamicin. Blood 2001, 98, 988–994. [Google Scholar] [CrossRef] [Green Version]
  122. Del Poeta, G.; Venditti, A.; Aronica, G.; Stasi, R.; Cox, M.C.; Buccisano, F.; Bruno, A.; Tamburini, A.; Suppo, G.; Simone, M.D.; et al. P-glycoprotein expression in de novo acute myeloid leukemia. Leuk. Lymphoma 1997, 27, 257–274. [Google Scholar] [CrossRef]
  123. Rafiee, R.; Chauhan, L.; Alonzo, T.A.; Wang, Y.C.; Elmasry, A.; Loken, M.R.; Pollard, J.; Aplenc, R.; Raimondi, S.; Hirsch, B.A.; et al. ABCB1 SNP predicts outcome in patients with acute myeloid leukemia treated with Gemtuzumab ozogamicin: A report from Children’s Oncology Group AAML0531 Trial. Blood Cancer J. 2019, 9. [Google Scholar] [CrossRef]
  124. Michieli, M.; Damiani, D.; Ermacora, A.; Masolini, P.; Raspadori, D.; Visani, G.; Scheper, R.J.; Baccarani, M. P-glycoprotein, lung resistance-related protein and multidrug resistance associated protein in de novo acute non-lymphocytic leukaemias: Biological and clinical implications. Br. J. Haematol. 1999, 104, 328–335. [Google Scholar] [CrossRef]
  125. Boyer, T.; Gonzales, F.; Barthélémy, A.; Marceau-Renaut, A.; Peyrouze, P.; Guihard, S.; Lepelley, P.; Plesa, A.; Nibourel, O.; Delattre, C.; et al. Clinical significance of ABCB1 in acute myeloid leukemia: A comprehensive study. Cancers 2019, 11, 1323. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  126. Balsat, M.; Renneville, A.; Thomas, X.; De Botton, S.; Caillot, D.; Marceau, A.; Lemasle, E.; Marolleau, J.P.; Nibourel, O.; Berthon, C.; et al. Postinduction minimal residual disease predicts outcome and benefit from allogeneic stem cell transplantation in acute myeloid leukemia with NPM1 mutation: A study by the acute leukemia French association group. J. Clin. Oncol. 2017, 35, 185–193. [Google Scholar] [CrossRef] [PubMed]
  127. Dillon, R.; Hills, R.; Freeman, S.; Potter, N.; Jovanovic, J.; Ivey, A.; Kanda, A.S.; Runglall, M.; Foot, N.; Valganon, M.; et al. Molecular MRD status and outcome after transplantation in NPM1-mutated AML. Blood 2020, 135, 680–688. [Google Scholar] [CrossRef] [Green Version]
  128. Freeman, S.D.; Virgo, P.; Couzens, S.; Grimwade, D.; Russell, N.; Hills, R.K.; Burnett, A.K. Prognostic relevance of treatment response measured by flow cytometric residual disease detection in older patients with acute myeloid leukemia. J. Clin. Oncol. 2013, 31, 4123–4131. [Google Scholar] [CrossRef] [PubMed]
  129. Lambert, J.; Lambert, J.; Nibourel, O.; Pautas, C.; Hayette, S.; Cayuela, J.M.; Terré, C.; Rousselot, P.; Dombret, H.; Chevret, S.; et al. MRD assessed by WT1 and NPM1 transcript levels identifies distinct outcomes in AML patients and is influenced by gemtuzumab ozogamicin. Oncotarget 2014, 5, 6280–6288. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  130. Kapp-Schwoerer, S.; Weber, D.; Corbacioglu, A.; Gaidzik, V.I.; Paschka, P.; Krönke, J.; Theis, F.; Rücker, F.G.; Teleanu, M.V.; Panina, E.; et al. Impact of gemtuzumab ozogamicin on MRD and relapse risk in patients with NPM1-mutated AML: Results from the AMLSG 09-09 trial. Blood 2020, 136, 3041–3050. [Google Scholar] [CrossRef] [PubMed]
  131. Thomas, D.; Majeti, R. Biology and relevance of human acute myeloid leukemia stem cells. Blood 2017, 129, 1577–1585. [Google Scholar] [CrossRef]
  132. Ng, S.W.K.; Mitchell, A.; Kennedy, J.A.; Chen, W.C.; McLeod, J.; Ibrahimova, N.; Arruda, A.; Popescu, A.; Gupta, V.; Schimmer, A.D.; et al. A 17-gene stemness score for rapid determination of risk in acute leukaemia. Nature 2016, 540, 433–437. [Google Scholar] [CrossRef] [PubMed]
  133. Maimaitili, Y.; Inase, A.; Miyata, Y.; Kitao, A.; Mizutani, Y.; Kakiuchi, S.; Shimono, Y.; Saito, Y.; Sonoki, T.; Minami, H.; et al. An mTORC1/2 kinase inhibitor enhances the cytotoxicity of gemtuzumab ozogamicin by activation of lysosomal function. Leuk. Res. 2018, 74, 68–74. [Google Scholar] [CrossRef] [PubMed]
  134. Mizutani, Y.; Inase, A.; Maimaitili, Y.; Miyata, Y.; Kitao, A.; Matsumoto, H.; Kawaguchi, K.; Higashime, A.; Goto, H.; Kurata, K.; et al. An mTORC1/2 dual inhibitor, AZD2014, acts as a lysosomal function activator and enhances gemtuzumab ozogamicin-induced apoptosis in primary human leukemia cells. Int. J. Hematol. 2019, 110, 490–499. [Google Scholar] [CrossRef]
  135. Pan, J.; She, M.; Xu, Z.X.; Sun, L.; Yeung, S.C.J. Farnesyltransferase inhibitors induce DNA damage via reactive oxygen species in human cancer cells. Cancer Res. 2005, 65, 3671–3681. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  136. Jawad, M.; Yu, N.; Seedhouse, C.; Tandon, K.; Russell, N.H.; Pallis, M. Targeting of CD34+CD38- cells using Gemtuzumab ozogamicin (Mylotarg) in combination with tipifarnib (Zarnestra) in acute Myeloid Leukaemia. BMC Cancer 2012, 12. [Google Scholar] [CrossRef] [Green Version]
  137. Carr, M.I.; Zimmermann, A.; Chiu, L.Y.; Zenke, F.T.; Blaukat, A.; Vassilev, L.T. DNA-PK Inhibitor, M3814, as a New Combination Partner of Mylotarg in the Treatment of Acute Myeloid Leukemia. Front. Oncol. 2020, 10. [Google Scholar] [CrossRef] [PubMed]
  138. Yamauchi, T.; Uzui, K.; Nishi, R.I.; Shigemi, H.; Ueda, T. Gemtuzumab Ozogamicin and Olaparib Exert Synergistic Cytotoxicity in CD33-positive HL-60 Myeloid Leukemia Cells. Anticancer Res. 2014, 34, 5487–5494. [Google Scholar] [PubMed]
  139. Portwood, S.M.; Cantella, M.C.; Cronin, T.L.; Wang, E.S. Addition of the PARP Inhibitor, Talazoparib, to Gemtuzumab Ozogamicin Significantly Enhances Anti-Leukemic Activity in Human CD33+ Acute Myeloid Leukemia. Blood 2019, 134, 1371. [Google Scholar] [CrossRef]
Figure 1. Mechanisms of action of GO.
Figure 1. Mechanisms of action of GO.
Cancers 13 04566 g001
Figure 2. Biomarkers predictive of GO response in AML.
Figure 2. Biomarkers predictive of GO response in AML.
Cancers 13 04566 g002
Figure 3. Targeted agents that are under clinical or preclinical investigation in combination with GO.
Figure 3. Targeted agents that are under clinical or preclinical investigation in combination with GO.
Cancers 13 04566 g003
Table 1. Clinical trials of GO used before or after transplantation (clinicaltrials.gov updated to 1 September 2021).
Table 1. Clinical trials of GO used before or after transplantation (clinicaltrials.gov updated to 1 September 2021).
NCT NumberInterventionConditionsAgePhaseTrial Status
NCT00044733GO at relapse after auto or alloSCTAMLchild, adult, older adultIICompleted
NCT02221310GO+chemotherapy followed by alloSCThigh-risk AML/MDSup to 25 yearsIIRecruiting
NCT00669890GO+Busulfan and Cyclophosphamid before alloSCThigh-risk AML/MDS/JMMLup to 30 yearsITerminated
NCT02117297GO consolidation after alloSCTaverage-risk AML/MDSup to 25 yearsIIRecruiting
NCT01020539GO consolidation after alloSCTaverage-risk AML/MDS/JMMLup to 30 yearsIActive, not recruiting
NCT00460447GO before alloSCT at relapseAML18–70 yearsI/IIUnknown
NCT00038831GO+Melphalan+Fludarabine before alloSCT in older or medically infirm patientsAML/MDS/CLL12–75 yearsI/IICompleted
NCT00476541GO consolidation after SCTAMLup to 18 yearsIIICompleted
NCT00008151GO+fludarabine+total-body irradiation before alloSCTadvanced AML/MDSchild, adult, older adultIICompleted
NCT00038805GO+nonmyeloablative preparative regimen before mini-alloSCT in older or medically infirm patientsAML/ALL/CML/MDS55–75 yearsII/IIITerminated
NCT01723657GO “in vivo purging” before autoSCT in patients with favorable/intermediate characteristics and without matched related donorAML18–70 yearsIICompleted
NCT00070174GO in remission induction, intensification therapy before alloSCTAMLchild, adult, older adultIICompleted
Allo: allogenic; AML: acute myeloid leukemia; auto: autologous; CLL: chronic lymphocytic leukemia; CML: chronic myeloid leukemia; GO: gemtuzumab ozogamicin, JMML: juvenile myelomonocytic leukemia; MDS: myelodysplastic syndrome.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Gottardi, M.; Simonetti, G.; Sperotto, A.; Nappi, D.; Ghelli Luserna di Rorà, A.; Padella, A.; Norata, M.; Giannini, M.B.; Musuraca, G.; Lanza, F.; et al. Therapeutic Targeting of Acute Myeloid Leukemia by Gemtuzumab Ozogamicin. Cancers 2021, 13, 4566. https://doi.org/10.3390/cancers13184566

AMA Style

Gottardi M, Simonetti G, Sperotto A, Nappi D, Ghelli Luserna di Rorà A, Padella A, Norata M, Giannini MB, Musuraca G, Lanza F, et al. Therapeutic Targeting of Acute Myeloid Leukemia by Gemtuzumab Ozogamicin. Cancers. 2021; 13(18):4566. https://doi.org/10.3390/cancers13184566

Chicago/Turabian Style

Gottardi, Michele, Giorgia Simonetti, Alessandra Sperotto, Davide Nappi, Andrea Ghelli Luserna di Rorà, Antonella Padella, Marianna Norata, Maria Benedetta Giannini, Gerardo Musuraca, Francesco Lanza, and et al. 2021. "Therapeutic Targeting of Acute Myeloid Leukemia by Gemtuzumab Ozogamicin" Cancers 13, no. 18: 4566. https://doi.org/10.3390/cancers13184566

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop