Next Article in Journal
Advances in Sol-Gel-Based Superhydrophobic Coatings for Wood: A Review
Previous Article in Journal
The Sigma Factor AsbI Is Required for the Expression of Acinetobactin Siderophore Transport Genes in Aeromonas salmonicida
Previous Article in Special Issue
A Combination of the Immunotherapeutic Drug Anti-Programmed Death 1 with Lenalidomide Enhances Specific T Cell Immune Responses against Acute Myeloid Leukemia Cells
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

A Direct Comparison, and Prioritisation, of the Immunotherapeutic Targets Expressed by Adult and Paediatric Acute Myeloid Leukaemia Cells: A Systematic Review and Meta-Analysis

1
Department of Chemistry and Biochemistry, University of Hull, Kingston upon Hull HU6 7RX, UK
2
Hull York Medical School, University of Hull, Kingston upon Hull HU6 7RX, UK
3
Centre for Biomedicine, Hull York Medical School, Kingston upon Hull HU6 7RX, UK
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2023, 24(11), 9667; https://doi.org/10.3390/ijms24119667
Submission received: 26 April 2023 / Revised: 27 May 2023 / Accepted: 29 May 2023 / Published: 2 June 2023
(This article belongs to the Special Issue Molecular Mechanisms and Therapies of Myeloid Leukaemia 2.0)

Abstract

:
Acute myeloid leukaemia (AML) is characterized by impaired myeloid differentiation resulting in an accumulation of immature blasts in the bone marrow and peripheral blood. Although AML can occur at any age, the incidence peaks at age 65. The pathobiology of AML also varies with age with associated differences in incidence, as well as the frequency of cytogenetic change and somatic mutations. In addition, 5-year survival rates in paediatrics are 60–75% but fall to 5–15% in older AML patients. This systematic review aimed to determine whether the altered genes in AML affect the same molecular pathways, indifferent of patient age, and, therefore, whether patients could benefit from the repurposing drugs or the use of the same immunotherapeutic strategies across age boundaries to prevent relapse. Using a PICO framework and PRISMA-P checklist, relevant publications were identified using five literature databases and assessed against an inclusion criteria, leaving 36 articles, and 71 targets for therapy, for further analysis. QUADAS-2 was used to determine the risk of bias and perform a quality control step. We then priority-ranked the list of cancer antigens based on predefined and pre-weighted objective criteria as part of an analytical hierarchy process used for dealing with complex decisions. This organized the antigens according to their potential to act as targets for the immunotherapy of AML, a treatment that offers an opportunity to remove residual leukaemia cells at first remission and improve survival rates. It was found that 80% of the top 20 antigens identified in paediatric AML were also within the 20 highest scoring immunotherapy targets in adult AML. To analyse the relationships between the targets and their link to different molecular pathways, PANTHER and STRING analyses were performed on the 20 highest scoring immunotherapy targets for both adult and paediatric AML. There were many similarities in the PANTHER and STRING results, including the most prominent pathways being angiogenesis and inflammation mediated by chemokine and cytokine signalling pathways. The coincidence of targets suggests that the repurposing of immunotherapy drugs across age boundaries could benefit AML patients, especially when used in combination with conventional therapies. However, due to cost implications, we would recommend that efforts are focused on ways to target the highest scoring antigens, such as WT1, NRAS, IDH1 and TP53, although in the future other candidates may prove successful.

1. Introduction

Acute myeloid leukaemia (AML) is an aggressive cancer that occurs due to the clonal expansion of immature white blood cells, known as blasts [1]. The accumulation of blasts occurs due to cytogenetic and molecular abnormalities that impair myeloid stem cell differentiation within the bone marrow [2]. Due to the impairment of normal haemopoiesis, the increased proliferation can cause blood-related problems, such as anaemia and haemorrhage [3].
The frequency, pathophysiology, treatment approaches, and survival rates vary between adult and paediatric AML, with age being a major factor in the frequency of AML diagnosis. In patients below 65 years of age, there are ~1.3 cases per 100,000 compared to ~12.2 cases per 100,000 in those above 65 years of age [2]. The increased AML rates in the older population are likely caused by the random accumulation of somatic mutations over time. Additionally, age seems to be a factor in the type of AML that patients develop. AML in the vast majority of paediatric patients develops de novo, whilst in adult patients AML more frequently develops from myeloproliferative neoplasms or myelodysplastic syndromes [1]. Along with age, there is often an increase in the activation of oncogenes, such as FLT3 [4], inactivation of tumour suppressor genes, such as WT1 [5], aberration of epigenetic modulators, such as TET2 [4], and decreased immune response [1]. Survival rates differ significantly between adult and paediatric patients, with one study showing a 5-year survival of ~17% in adult patients over 60 years of age [6], while survival rates in paediatric patients were around 60–75% [1]. The capacity of older and younger adults and children to tolerate the rigours of cancer treatment may explain why to date adult and paediatric patients have been treated in quite independent ways. Despite the differences in pathophysiology and initial survival rates, one thing AML has in common is that both adult and paediatric patients are at a high risk of death through a combination of relapse and treatment-related mortality [1]. Current treatment methods that include both induction and consolidation therapy often fail to eradicate AML cells that are protected by the bone marrow microenvironment and lead to relapse (reviewed in [7]). Thus, a new approach for AML treatment is essential to try to minimise the death toll caused by relapse.
Recently there has been a shift towards antigen-targeting immunotherapy in the treatment of cancers, including AML, to trigger anti-leukaemic responses [8]. In 2007, a National Cancer Institute Immunotherapy agent workshop [9] was held to rank immunotherapy targets based on the likelihood of cancer therapy efficacy. This well vetted list with broad and substantive input from academia, industry, and the government identified a number of immunotherapeutic agents that were effective cancer vaccine components in preclinical models. A subsequent National Cancer Institute pilot project sought to prioritise cancer antigens [10] based on their potential to act as translated targets for therapy. The methodology for prioritization was based on the analytical hierarchy process (AHP) [11], a mathematical model that helps teams deal with complex decisions that involve human perception and judgement. It has been used in a variety of settings, including government, industry, healthcare, and education, and helps by breaking down complex problems into pairwise comparable subproblems that are easier to quantify even when people have very different areas of specialisation. The model for prioritizing cancer antigens was developed through the combined efforts of experts in the field, and they assigned weighting to each of the criteria they ranked as being important for cancer vaccine development. Although with some limitations, this allowed for a uniform approach to the assessment of potential immunotherapeutic targets and a way to rank the most promising. There are multiple immunotherapeutic targets on the list of cancer antigens that are relevant to leukaemia and show promise in clinical trials. For example, both Phase I and II trials of a vaccine targeting PR1, a peptide derived from the leukaemia-associated antigen proteinase 3 [12], have been successful [13]. Recent vaccine developments have allowed for simultaneous targeting of multiple leukaemic antigens through the fusion of patient-derived AML cells to autologous dendritic cells, which induced an expansion of tumour-specific T cells, leading to extended remission times [14].
We wanted to determine whether the molecular pathways affected in AML were the same in adults and paediatrics, and whether the same immunotherapy targets could be used to treat AML across age groups. AML is diagnosed by molecular findings as described in the 2016 revision of the World Health Organisation (WHO) classification of myeloid neoplasms and acute leukaemia [15] which were published in the “blue book” monographs in 2001 and 2008, as third and fourth editions, respectively. However, we also wanted to include earlier articles that had used the French–American British (FAB) system of subtyping AML [16] and so we focused on studies of AML patients without cytogenetic abnormalities (such as t(8;21), inv(16) or t(15;17)), the FAB M4 subtype of AML known as acute myelomonocytic leukaemia, which has the highest prominence in the UK, and FAB M5, which includes acute monoblastic/monocytic leukaemia [17]. Our study examined all the available literature on antigen expression in paediatrics and adults with AML, since the inception of the literature databases until the current time. We have included a detailed meta-analysis that examines the commonality and differences in antigen expression and the pathways involved, and delivers a prioritisation of the top 20 antigens in each age-group as targets for immunotherapy clinical trials. By doing this we have established a large overlap in immunotherapeutic targets for adult and paediatric AML, illustrating the potential for a unified treatment approach.

2. Methods

2.1. Systematic Review

Standard systematic review methods were used and reported according to the PRISMA guidelines [18], including the development of a protocol (Supplementary Table S1). Google Scholar was used to ensure that this type of review of current research had not been undertaken recently, and the search strategy was developed based on index terms found in three to six sentinel articles that were identified in an initial screening of the literature using PubMed. The articles were identified using MeSH terms in articles, without limitation on when they were published. We wanted to show that the search terms identified relevant articles and that their own key words were encompassed by our search terms.
The study inclusion criteria were composed of both study and report characteristics (Table 1). The PICO system was used to design the research question, as follows: population—AML patients, intervention—expression of antigen targets in patient samples, comparison—expression in samples from healthy donors, outcome—potential as targets for therapy.

2.2. Search Strategy

Two independent reviewers (V.M. and H.G.) performed the search of the literature and screening process. Any disagreements were then settled by a third independent reviewer (B.G.) as and when required. MeSH terms and Boolean operators were used to search five databases (Scopus, the Cochrane library, CINAHL, MEDLINE, and PubMed) (Table 2) from inception until 4 March 2023. To ensure that all the relevant literature was used, alternative spellings of certain terms, such as leukaemia, were used. Studies using non-human models, animal models, comments, opinions, protocols, and non-English language publications were excluded.

2.3. Study Selection

The studies found within each database were exported into a single page of a Microsoft Excel Office 2019 sheet, combined, and duplicates were removed (Supplementary Table S2). The remaining articles were screened by title and then abstract, and articles were excluded if they did not meet the inclusion criteria. An in-depth screen of the articles was then performed. At the stage of abstract reading the search excluded books, systematic reviews, meta-analyses, and conference papers. The assessment of whether an article met the inclusion/exclusion criteria was performed by two independent researchers (V.M. and H.G.) and each reviewer were blinded to each other’s exact decisions until screening was complete. Any uncertainties in respect to the relevance of content were discussed with B.G.
Review articles were only removed once the cited articles in all selected manuscripts had been screened against the inclusion/exclusion criteria, as detailed above. This “backward snowballing” step helps ensure that relevant literature is successfully found as part of a systematic review [19].

2.4. Quality Assessment

A study specific version of QUADAS-2 [20] was applied to all articles at the final stage to factor in the risk of bias (Supplementary Table S3). Any articles that had a significant risk of bias were then excluded. Two independent reviewers (V.M. and H.G.) were used to minimise the risk of reviewer bias, and any disagreements were resolved through the involvement of a third reviewer (B.G.).

2.5. Data Extraction and Synthesis

From the final list of articles after quality assurance and risk of bias analysis, the potential targets of interest were separated into two data collection files, one for the antigens found in adult AML tissues and one for the antigens found in paediatric AML tissues (Supplementary Table S4). The NCBI number for each target was listed, as well as which articles it was listed within. Cancer antigens were then prioritized using the system devised by Cheever et al. [10], developed to rank vaccine targets based on predefined and pre-weighted objective criteria. The criteria in descending order of importance (followed by weighting in parenthesis) were: (a) therapeutic function (0.32); (b) immunogenicity (0.17); (c) role of the antigen in oncogenicity (0.15); (d) specificity (0.15); (e) expression level and percent of antigen-positive cells (0.07); (f) stem cell expression (0.05); (g) number of patients with antigen-positive cancers (0.04); (h) number of antigenic epitopes (0.04); (i) cellular location of antigen expression (0.02). None of the antigens were expected to have all the characteristics of an ideal antigen, but had one been perfect it would have scored 1.0.

2.6. Meta Analyses

PANTHER was used to illustrate which pathways are affected by events within the genome through predictions of gene interactions/functions. The 20 potential targets with the highest prioritization scores for both adult and paediatric AML underwent PANTHER analysis independently to produce a bar chart of the affected pathways.
STRING was also utilized to analyse the 20 highest scoring targets in both adult and paediatric AML separately. This created networks of known and predicted protein–protein interactions within Homo sapiens from a wide range of sources for both diseases. For both networks, the confidence levels of interactions were set to 0.7 to increase the accuracy of the network without excluding predicted connections. The database has in-built quality control and provides enrichment analysis to ensure the accuracy and relevance of the networks produced [21]. The identified inter-relationships can suggest which interactions define the behaviour of the biological system [22].

3. Results

3.1. Study Selection and Quality Assessment

The search strategy used identified a total number of 8577 articles (Supplementary Table S2). Of these, 5785 remained after the removal of duplicates. Articles were discarded in stages according to the exclusion criteria as shown in the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (Prisma-P) flow diagram (Figure 1). A total of 36 articles remained at the end of the process, which were then subjected to QUADAS-2 assessment to check for risk of bias [20]. The QUADAS-2 scores (Supplementary Table S3) were then applied to each of the 71 potential targets (Table 3) which led to the elimination of eight targets from the final list of potentially viable targets for immunotherapy.

3.2. Data Extraction

A total of 90.7% of the genes identified as altered in paediatric patients were also identified in the literature describing altered gene expression in adults with AML (Figure 2; Supplementary Table S4). However, a further 21 genes were identified as having altered expression in adult AML tissues only.

3.3. Prioritisation of Tumour Antigens as Targets for Immunotherapy

We identified and assessed 63 targets and found that they ranged in score from 0.85 (WT1) down to the lowest scoring target at 0.17 (VEGFR-2) (Figure 3; Supplementary Table S5). A total of 20 antigens scored above 0.7, with all scoring highly for immunogenicity and most scoring low for expression levels and % positive cells. Overall, despite the highest value, 1, being assigned to the majority of targets for the number of epitopes identified due to each category being weighted differently, this category did not significantly contribute to the overall ratings.

3.4. Pathway and Protein–Protein Interaction Analysis

To determine which pathways were predominantly affected within adult and paediatric AML cells, the NCBI numbers of the top 20 targets, as identified by the modified Cheever model (in Section 3.3), were independently entered into Protein Analysis Through Evolutionary Relationships (PANTHER) [59]. PANTHER contains 176 different pathways within its database, of which 25 pathways were found to include the AML antigens identified in this study. There were notable similarities in the pathways involved as well as their relative involvement in AML between adult and paediatric patients (Figure 4). Only one pathway, the Alzheimer’s disease presenilin pathway, differed between the two age groups of AML patients, as it was only present within the top 20 for paediatric AML. The two pathways with the highest number of targets involved in both adult and paediatric AML age groups were angiogenesis and inflammation mediated by the chemokine and cytokine signalling pathway.
Inputting the top 20 targets into the Search Tool for the Review of Interacting Genes/Protein (STRING) for paediatric and adult AML patients showed their inter-relationships (Figure 5). STRING networks contain different coloured connections corresponding to the type of prediction; for example, experimentally determined interactions are connected in pink. This allowed for not only the number of different interactions to be determined but also for the strength of current evidence available to be indicated. The confidence levels of predicted interactions were set to 0.7 so that only highly likely interactions were identified. BIRC5 had the most relationships in paediatric AML, while PTPN11 had the most in adult AML.

4. Discussion

The databases selected meant that a large number of articles were systematically reviewed. This is because the Cochrane Library includes articles from the CINAHL database and MEDLINE® includes results from PubMed. It is possible that relevant studies may have been missed if they were not written entirely in English or if the title or abstract did not make it clear that the outcome of the article was focussed on antigen expression in AML. However, the use of “reverse snowballing”, whereby citations in the selected articles were also subjected to the systematic review process, was used to mitigate this risk [19].
The databases used for the literature search have built in quality assessment. For example, Scopus openly advertises “a rigorous evaluation and selection process to ensure it meets the high-quality title selection criteria required for acceptance” and that journals “must demonstrate their ability to maintain quality status each year” with a “multi-step re-evaluation process” [60]. Additionally, the use of two independent reviewers combined with a third to settle any disputes allowed for an extra level of quality assurance. Before data extraction took place, the articles chosen from the initial searches underwent a quality assurance (QUADAS-2) step to ensure all included antigenic targets that achieved a sufficient quality of evidence to support their further analysis [20]. QUADAS-2 has two separate components to it: one focuses on the risk of bias while the other focuses on applicability to the study. Therefore, each of these components were taken into account when including or excluding studies. For example, if the patients studied had AML M6, they would have a low rating in the applicability section of the assessment. After assessing all selected papers using QUADAS, the total number of antigenic targets dropped from 72 to 63.
The criteria used excluded animal models. Traditionally animal models, predominantly rodent, have been utilized to investigate the underlying mechanisms of biological processes in a multiorgan setting, and can avoid some of the ethical and practical concerns that are associated with human studies. However, there are several drawbacks to the use of animal models within cancer research, which has called for a shift in how these models are used. The average success rate within clinical trials of the translation from animal models and patients is stated to be less than 8% [61]. This illustrates the severe limitations of animal models to mimic human carcinogenesis, pathophysiology, and disease progression. There are new methods being investigated to mitigate the differences seen between human and rodent models, including humanized mouse models and even humanized larger animal models of cancer [62,63]. Another potential limitation of animal models is their reduced life span that may fail to mimic human AML where age is a significant factor in both patient survival and treatment choice. Thus, it would be much harder to distinguish between adult and paediatric targets within animal studies, because of the very short time rodents remain paediatric.
Significantly fewer articles described the testing of paediatric patient samples, probably reflecting their relative scarcity compared with adult AML, and this may have contributed to the lower number of targets identified in younger AML patients. In addition, the reduced number of articles describing genetic change in paediatric AML may reflect historical concerns with the use of samples from paediatric patients in clinical trials due to the age of consent being 18 years old. However, it was recently stated that treatment intended for paediatrics should only be tested on that age group due to pathophysiological differences between adults and children [64]. In addition, survival rates for paediatric AML are much higher than for adult AML patients, which in addition to the elevated frequency of adult AML, makes adult AML more accessible to studies, while the need for new therapies, particularly for older adults, is a priority area for research. Some articles found in this study that had examined paediatric AML patients included patients in their late teens only (>15 years of age) [29,52]. This extremely narrow age-range could have skewed the results obtained as older paediatric patients have sometimes been observed to have an adult AML profile [65]. Most of the targets affected by this, such as WT1, also appeared in other articles from the literature search, mitigating this issue.
There were a vast number of similarities seen between adult and paediatric AML with 90.7% of antigens in paediatric samples also being described in adult AML. In fact, only four targets identified in paediatric AML were not shared with adult AML (CD44, CLEC12A, DARS-AS1, and TSC2). The large number of similarities observed between adult and paediatric AML supported the possibility of treatment repurposing, such as targeting BIRC5 (survivin) that had high expression levels in both adult and paediatric AML [66,67]. BIRC5 has been linked to multiple biological pathways including cell proliferation, angiogenesis, and metastasis [68], while above median levels of BIRC5 were associated with worse survival in adult AML patients with inv(16) [38]. FL118 targeting the BIRC5 promotor has been linked to transcriptional inhibition, and anti-tumour activity, in multiple cancer types, including cervical and pancreatic cancer and leukaemia [68,69,70].
The Initial extraction of data identified a large amount of targets; therefore, we chose the Cheever et al. [10] model of antigen prioritization to identify which antigens we would recommend for further studies. The Cheever model, however, does have limitations in that it has not been updated since its first publication in 2009. This means that there may be new important factors that the model did not take into consideration that may alter the relative weighting of each category. There is unfortunately no recent update on this or any other model that can give a predefined system for the ranking of potential targets. One example of a limitation that a more updated version may have mitigated was that the therapeutic function category focused on vaccine development while a wider range of immunotherapeutic approaches may be more appropriate. This led to some targets that have clear evidence of efficacy in other immunotherapy methods, such as combinational CAR T cells or tandem diabodies, (not vaccines per se) being scored lower despite having high therapeutic function. The biggest limitation of the scoring system was that there was no way to give novel antigenic targets the same weighting as those that had been previously investigated, thus, prioritising the more investigated rather than the more promising targets. Despite its limitations, the Cheever model of antigen prioritization offered a way to determine which antigens had the most promise as vaccine targets by considering rigorously determined criteria, and their relative weighting as set out by a panel of experts within the field. One study showed that the median cost of developing a single cancer drug was $648 million [71]; thus, it is clear that due to cost limitations only the targets with the most potential can be prioritized for development by funding agencies. Therefore, the method of scoring potential targets by Cheever et al. needs to be considered when deciding to move forwards with a target. However, to ensure the inclusion of the most promising targets for approaches other than just vaccines, the scores cannot be solely relied upon to make that decision. One example of this would be that CD123, which fell outside of the highest scoring targets after Cheever, has been used to target CLL in combination with CD3 via dual-affinity retargeting antibodies (DARTs) [72,73]. This suggests it has a higher immunotherapeutic potential than its original score for AML if the response initially seen in this CLL trial was to translate into AML patients.
Looking at both the amount of the literature that focussed on it and its top position after prioritization, WT1 was the most promising immunotherapeutic target for AML patients. This was the same outcome found by Cheever et al. [10] in a study that focussed more on data from patients with solid tumours by virtue of their predominance in the literature and in clinics. There has been previous success in preventing AML relapse through TCR targeting of WT1 with 100% relapse-free survival observed after 44 months [74]. However, this study only included adult patients and was implemented post-transplant. WT1 is an established target, but not novel. To identify a novel target, it may be best to rule out the therapeutic function part of the prioritization to instead focus on potential targets, not established ones.
CD13 could prove a viable target due to the high scores it achieved in key categories, such as immunogenicity [75] and the number of patients whose AML blasts express it [76]. One study investigated the influence of CD13 on the growth and survival of AML cells in vitro and found that CD13 could serve as a target for inducing caspase-dependent apoptosis within AML [77]. Despite being an in vitro study, these findings confirm that CD13 has the potential to be an effective target for a unified treatment approach. Additionally, Ubenimex is a CD13 inhibitor that was able to show promising effects from in vitro studies to clinical trials; in the Phase I clinical trials it was observed to prolong survival of AML patients but to also promote graft versus leukaemia effects in patients post-transplant [78]. Similar findings have been observed through targeting CD13 and TIM3 with bispecific and split CAR T cells to eradicate AML with reduced toxicity to the stem cells in preclinical models [75]. Therefore, despite CD13 not initially scoring as highly in the Cheever model as other potential targets, possibly due to the model’s focus on vaccine development, CD13 would be a very promising target for future therapeutics studies.
PANTHER was used to determine which pathways were affected by the 20 highest scoring targets in both adult and paediatric AML patients independently. In addition to paediatric AML sharing 96% of their immunotherapeutic targets with adults, both adult and paediatric AML have the most targets involved in angiogenesis and inflammation mediated by the cytokine and chemokine pathway(s). Angiogenesis is the generation of new blood vessels, which are key for cancer survival through the fresh supply of oxygen and nutrients to cancer cells [79]. Usually, angiogenesis decreases with age due to vascular ageing [80], so to have angiogenesis equally involved in both adult and paediatric AML was surprising. Two targets implicated in angiogenesis for both adult and paediatric AML were WT1, a key angiogenesis regulator [81], and BIRC5 [82]. Expression levels of BIRC5 varied depending on AML subtype but, within M4 and M5, the focus of this review, they were elevated. Even within M4/5 there was a potential issue with angiogenesis targeting other pathways, such as the platelet-derived growth factor (PDGF) signalling pathway, which can provide “escape” mechanisms, allowing cancer to resume growth [83]. Therefore, the most effective immunotherapy approach may be to simultaneously target multiple antigens and/or their pathways to limit the chance of relapse due to the escape of drug resistant clones [84].
Inflammation mediated by the cytokine and chemokine pathway was involved in both adult and paediatric AML development and had an equal number of targets involved in angiogenesis. Cytokines that are secreted within the bone marrow microenvironment are crucial in modulating cell survival, proliferation, differentiation, and the immune response [85]. The exact role of inflammatory cytokines in leukaemic cell expansion and the progression of AML are not yet fully understood; however, previous studies have indicated that pro-inflammatory cytokines contribute to an inflammatory microenvironment and have a growth-promoting effect on AML [86]. Both CXCR4 and JAK2 are linked specifically to inflammation via cytokines and chemokines, and are expressed by adult and paediatric AML cells, but JAK2 scored higher during prioritization. Chemokines’ primary function is the coordination and recruitment of immune cells and, therefore, they have a vital role in tumour development, aiding the formation of immunosuppressive and protective tumour microenvironments [87]. There has been previous interest in targeting cytokines and chemokines for immunotherapeutic treatment. Interferon-α was first approved for use on hairy cell leukaemia in 1986 and has since been used for the treatment of several haematological malignancies at high doses to exploit their pro-apoptotic activity on cancerous cells [88]. However, due to its relatively low efficacy and high cytotoxicity, its clinical use is now being reduced. Other cytokines and chemokines show promise, such as the anti-inflammatory mediators TGF-β and IL-10, which appear to impede AML progression [89,90].
The adult AML STRING network was created using the 20 highest scoring targets after Cheever prioritization to ensure the network only included the most relevant targets. TP53 was the target that displayed the most connections within the adult network. It has been predicted that ~10% of AML patients have mutations in TP53, which are associated with poor prognosis [91]. TP53 mutations are associated with older age in AML, affecting up to 25% of elderly individuals [92], and were within the top five ranked targets for both adult and paediatric AML. However, clinical trials targeting p53, directly or indirectly, remain a significant unmet need [93]. TP53 has been examined as a target for the treatment of AML using microRNA (miRNA) therapy, including miR-223, which is currently in preclinical trials. Despite there being little data on miRNA therapy in AML, the fact that miR-223 has been linked to all FAB subtypes does give promise that, with the correct delivery system, it could provide an effective targeted treatment for a wide range of patients [94]. Similarly to other targeting techniques, toxicity issues would need to be addressed, which is where both delivery system and potential combinational approaches would need careful consideration.
In paediatric patients, STRING indicated that the target with the most connections was PTPN11. PTPN11 also had a lot of evidence types within the STRING network which supports its potential success as a target despite not being frequently represented within this literature review. PTPN11 was found to be mutated in both adult and paediatric patients with AML and, therefore, could provide a unified approach. Mutations within PTPN11 were found in ~12% of adult AML patients and only 4% of paediatric AML patients [95,96]. Recent clinical trials illustrated that, despite the relatively low number of patients with mutated PTPN11, carrying the mutation was strongly associated with adverse clinical outcomes and understanding the mechanisms that caused this affect may facilitate the development of treatments [97]. Due to PTPN11′s low mutation rates, especially in paediatric patients, it may be beneficial to consider a combinational approach if it were to be selected for development. This would entail pairing it up with a more common target to try and increase the treatment’s overall efficacy so that it does not have to be ruled out entirely based on its infrequent subversion in AML alone.
In concurrence with PANTHER, STRING networks showed vast similarities between adult and paediatric patients when considering the top 20 antigenic targets in each age group. In adults, BMI1, CA9, PRAME, and TERT only featured in adults with AML, while CD44, CD244, CLEC12A, and IL-3RA only featured in paediatrics. One of the most connected genes within the top 20 for paediatric AML was JAK2, which, as previously mentioned, is part of the cytokine-mediated inflammatory pathway. JAK2 has been previously utilized as an immunotherapeutic target for breast cancer; however, there is little evidence of its successful targeting in AML so far, although it could provide a novel therapeutic approach [98,99]. This is possibly because JAK2 mutations are seemingly rare in AML, with 11 out of 339 AML patients having JAK2 mutations [100]. Thus, despite JAK2 mutations being associated with de novo AML [101], they may not prove a viable target in terms of broad applicability. Another promising target within the top 20 in both adult and paediatric AML was NPM1. This target not only scored highly within the Cheever prioritization model but also had a high number of connections within the STRING network. NPM1 was also evidenced as being crucial to AML development by a team who used CRISPR dropout screening to identify genetic vulnerability within five different cell lines [102]. This study illustrated that NPM1 was depleted in four of the five AML cell lines, which identified it as an AML-specific vulnerability [102]. One potential target that lies outside of the top 20 highest scoring targets was DNMT3A, which was found in a larger proportion of AML patients than JAK2, with 22.1% (62/281) of patients having alterations in DNMT3A [103]. The current literature also links DNMT3A and isocitrate dehydrogenase 1 enzyme (IDH1) to both adult and paediatric AML, which does suggest that their targeting could prove an effective approach across a broad range of age groups. Already, hypomethylating agents, such as azacytidine and decitabine, and IDH inhibitors, are the main therapeutic treatments for older AML patients who are unable to tolerate chemotherapy and relapsed/refractory AML patients harbouring an IDH mutation, respectively (recently reviewed in [104]).
From the information presented thus far, there are a number of potential targets identified for the therapy of AML; however, due to the high costs of developing treatments and the length of time that the approval of a treatment takes, it may be beneficial to focus on a few prioritised targets for treatment development over the next few years. In order to decide which targets should be selected, there are additional factors to consider including broad applicability across subtypes and age-boundaries. FLT3 mutations have been found in ~25% of paediatric patients and ~30% of adult patients [105], with some studies suggesting that successful targeting of unmutated FLT3 could lead to ~80% of AML patients being treated with one approach [106,107]. Previous targeting of FLT3 initially showed high promise as an effective treatment, but resistance to FLT3 inhibitors is already becoming a pressing issue that would need to be addressed for this to be effective long-term. Current ideas suggest that there are two forms of resistance (primary and secondary) and that these are caused by different underlying mechanisms [108]. One example of a primary resistance causing mechanism is that the upregulation of CXCL12 and FGF2 within the bone marrow microenvironment can shield AML blasts from FLT3 inhibitors [109,110]. Meanwhile secondary resistance has numerous causative mechanisms, but one of the major ones is epigenetic dysregulation caused by mutations in epigenetic modification genes including DNM3TA, TET2, and IDH1/2 [111]. A reduction in resistance to FLT3 inhibitors could be achieved through either the development of novel inhibitors or by considering a multitarget approach. It is also worth noting that recent publications demonstrated methods of expression amplification in FLT3. This has not yet been applied to AML, but, in breast cancer, hormone receptor antagonism was able to enhance the efficacy of targeted immunotherapy by amplifying target expression [112]. If this could be applied to AML, then it would allow for targets that may have lower expression to be utilized rather than focusing future work solely on targets with elevated expression levels.
Another key factor to consider when selecting which of the targets should be pursued are the toxicity issues caused by a lack of specificity. There are ways to mitigate this type of toxicity either through a combinational CAR T cell approach [113] or the development of built-in “safety switches” that would eliminate the CAR T cells after use [114]. With the advent of next generation immunotherapeutic strategies including Boolean gating, reversing T cell exhaustion, and split/SUPRA CARs (recently reviewed in [115]), we see an opportunity to address the current challenges limiting the safety and efficacy of CAR-T cells for cancer treatment. It is likely that a combinational approach can provide a safe delivery method for immunotherapeutic targeting of AML, and, indeed, recent studies suggest the combination of an immune checkpoint inhibitor and antigen targeting, as well as immune modulation, could further enhance anti-leukaemic responses [116,117].

5. Conclusions

AML is a rare but progressive form of blood cancer, which continues to have poor prognosis in both adult and paediatric patients. Low survival rates indicate a clear need for the development of new treatment approaches that will universally improve prognosis. The most promising approach are immunotherapeutic methods that target specific antigens that have a key role in the development and survival of AML cells. This could create an approach with higher efficacy and fewer side effects than high-dose chemotherapy alone. Adult and paediatric AML are currently treated as different conditions due to differences in underlying pathophysiology; however, immunotherapy offers one way to cross age boundaries by targeting common antigens. This review has identified antigens that could or do offer an opportunity to develop a unified treatment approach. Throughout all analyses in this review, there were significant overlaps between the antigens expressed in adult and paediatric AML, including within the most promising targets. A few examples of the overlapping targets that stood out at multiple stages were WT1, TP53, BIRC5, and JAK2, with some of these already being effectively targeted within clinical trials using a variety of delivery systems. However, there are a lot more, such as CD13 and PTPN11, that have been identified as potentially viable novel targets for a future unified approach. With the correct target selection and delivery method it may be possible to mitigate a lot of the side effects associated with the immunotherapeutic treatment of AML, such as toxicity caused by the lack of specificity. It is likely that there are ways to develop a unified approach of immunotherapy that could treat both adult and paediatric patients simultaneously and benefit from the cost savings that drug repurposing offers.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/ijms24119667/s1.

Author Contributions

B.-a.G. designed the study; V.S.M. and H.G. performed the systematic review; V.S.M. and D.M.F. analysed the data and made the figures; V.S.M. and B.-a.G. wrote the paper. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by the INSPIRE grant generously awarded to the Hull York Medical School by the Academy of Medical Sciences through the Wellcome Trust [Ref: IR5\1018].

Institutional Review Board Statement

Ethical approval for this study was not required as the data employed did not reveal patient identifiers.

Informed Consent Statement

Not applicable.

Data Availability Statement

All data generated or analysed during this study are included in this published article and its Supplementary Materials files.

Acknowledgments

We would like to thank the librarians of the Brynmor Jones Library at the University of Hull.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

AMLAcute myeloid leukaemia
BIRC5Baculoviral IAP repeat containing 5
CAR T cellsChimeric antigen receptor T cells
CD13Alanine aminopeptidase
CD3Cluster of differentiation 3
CD33Sialic acid binding Ig-like lectin 3 (SIGLEC-3)
CD44CD44—phagocytic glycoprotein-1
CINAHLCumulative Index to Nursing and Allied Health Literature
CLEC12AC-type lectin domain family 12 member A
CXCR4C-X-C chemokine receptor type 4
DARS-AS1Aspartyl-tRNA synthetase antisense RNA 1
DNMT3ADNA methyltransferase 3 alpha
FABFrench–American–British
FLT3FMS-like tyrosine kinase 3
HLAHuman leukocyte antigen
IDH1Isocitrate dehydrogenase 1 enzyme
JAK2Janus kinase 2
M4Acute myelomonocytic leukaemia
M5Acute monocytic leukaemia
PANTHERProtein Analysis Through Evolutionary Relationships
PRISMAPreferred Reporting Items for Systematic Reviews and Meta-analyses
PTPN11Protein tyrosine phosphatase non-receptor type 11
QUADAS-2Quality Assessment of Diagnostic Accuracy Studies
STRINGSearch Tool for the Retrieval of Interacting Genes/Proteins
TCRT-cell receptor
TP53Tumour protein p53
TSC2Tuberous sclerosis complex 2
VEGFR-2Vascular endothelial growth factor 2
WT1Wilms’ tumour gene 1
AMLAcute myeloid leukaemia

References

  1. Chaudhury, S.; O’Connor, C.; Canete, A.; Bittencourt-Silvestre, J.; Sarrou, E.; Prendergast, A.; Choi, J.; Johnston, P.; Wells, C.A.; Gibson, B.; et al. Age-specific biological and molecular profiling distinguishes paediatric from adult acute myeloid leukaemias. Nat. Commun. 2018, 9, 5280. [Google Scholar] [CrossRef] [Green Version]
  2. De Kouchkovsky, I.; Abdul-Hay, M. Acute myeloid leukemia: A comprehensive review and 2016 update. Blood Cancer J. 2016, 6, e441. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Short, N.J.; Rytting, M.E.; Cortes, J.E. Acute myeloid leukaemia. Lancet 2018, 392, 593–606. [Google Scholar] [CrossRef] [PubMed]
  4. Feinberg, A.P.; Koldobskiy, M.A.; Gondor, A. Epigenetic modulators, modifiers and mediators in cancer aetiology and progression. Nat. Rev. Genet. 2016, 17, 284–299. [Google Scholar] [CrossRef]
  5. Wallwitz, J.; Aigner, P.; Stoiber, D. Tumor suppressors in acute myeloid leukemia. Leuk. Lymphoma 2021, 62, 2320–2330. [Google Scholar] [CrossRef]
  6. Rowe, J.M. Will new agents impact survival in AML? Best Pract. Res. Clin. Haematol. 2019, 32, 101094. [Google Scholar] [CrossRef]
  7. Pollyea, D.A.; Jordan, C.T. Therapeutic targeting of acute myeloid leukemia stem cells. Blood 2017, 129, 1627–1635. [Google Scholar] [CrossRef] [PubMed]
  8. Greiner, J. The Important Role of Immunotherapies in Acute Myeloid Leukemia. J. Clin. Med. 2019, 8, 2054. [Google Scholar] [CrossRef] [Green Version]
  9. NCI Immunotherapy Agent Workshop. 2009. Available online: http://dcb.nci.nih.gov/ImmunAgentWork (accessed on 25 April 2023).
  10. Cheever, M.A.; Allison, J.P.; Ferris, A.S.; Finn, O.J.; Hastings, B.M.; Hecht, T.T.; Mellman, I.; Prindiville, S.A.; Viner, J.L.; Weiner, L.M.; et al. The prioritization of cancer antigens: A national cancer institute pilot project for the acceleration of translational research. Clin. Cancer Res. 2009, 15, 5323–5337. [Google Scholar] [CrossRef] [Green Version]
  11. Saaty, T.L. The Analytic Hierarchy Process: Planning, Priority Setting, Resource Allocation; McGraw-Hill: New York, NY, USA, 1980. [Google Scholar]
  12. Qazilbash, M.H.; Wieder, E.; Thall, P.F.; Wang, X.; Rios, R.; Lu, S.; Kanodia, S.; Ruisaard, K.E.; Giralt, S.A.; Estey, E.H.; et al. PR1 peptide vaccine induces specific immunity with clinical responses in myeloid malignancies. Leukemia 2017, 31, 697–704. [Google Scholar] [CrossRef] [Green Version]
  13. Ivy, K.S.; Brent Ferrell, P., Jr. Disordered Immune Regulation and its Therapeutic Targeting in Myelodysplastic Syndromes. Curr. Hematol. Malig. Rep. 2018, 13, 244–255. [Google Scholar] [CrossRef] [PubMed]
  14. Stroopinsky, D.; Liegel, J.; Bhasin, M.; Cheloni, G.; Thomas, B.; Bhasin, S.; Panchal, R.; Ghiasuddin, H.; Rahimian, M.; Nahas, M.; et al. Leukemia vaccine overcomes limitations of checkpoint blockade by evoking clonal T cell responses in a murine acute myeloid leukemia model. Haematologica 2021, 106, 1330–1342. [Google Scholar] [CrossRef] [PubMed]
  15. Arber, D.A.; Orazi, A.; Hasserjian, R.; Thiele, J.; Borowitz, M.J.; Le Beau, M.M.; Bloomfield, C.D.; Cazzola, M.; Vardiman, J.W. The 2016 revision to the World Health Organization classification of myeloid neoplasms and acute leukemia. Blood 2016, 127, 2391–2405. [Google Scholar] [CrossRef] [PubMed]
  16. Bennett, J.M.; Catovsky, D.; Daniel, M.T.; Flandrin, G.; Galton, D.A.; Gralnick, H.R.; Sultan, C. Proposals for the classification of the acute leukaemias. French-American-British (FAB) co-operative group. Br. J. Haematol. 1976, 33, 451–458. [Google Scholar] [CrossRef] [PubMed]
  17. McNally, R.J.; Rowland, D.; Roman, E.; Cartwright, R.A. Age and sex distributions of hematological malignancies in the U.K. Hematol. Oncol. 1997, 15, 173–189. [Google Scholar] [CrossRef]
  18. Moher, D.; Liberati, A.; Tetzlaff, J.; Altman, D.G.; Group, P. Preferred reporting items for systematic reviews and meta-analyses: The PRISMA statement. PLoS Med. 2009, 6, e1000097. [Google Scholar] [CrossRef] [Green Version]
  19. Wohlin, C. Guidelines for snowballing in systematic literature studies and a replication in software engineering. In Proceedings of the 18th International Conference on Evaluation and Assessment in Software Engineering, London, UK, 13–14 May 2014; Volume 38, pp. 1–10. [Google Scholar] [CrossRef]
  20. Whiting, P.F.; Rutjes, A.W.; Westwood, M.E.; Mallett, S.; Deeks, J.J.; Reitsma, J.B.; Leeflang, M.M.; Sterne, J.A.; Bossuyt, P.M.; QUADAS-2. QUADAS-2: A revised tool for the quality assessment of diagnostic accuracy studies. Ann. Intern. Med. 2011, 155, 529–536. [Google Scholar] [CrossRef]
  21. Szklarczyk, D.; Morris, J.H.; Cook, H.; Kuhn, M.; Wyder, S.; Simonovic, M.; Santos, A.; Doncheva, N.T.; Roth, A.; Bork, P.; et al. The STRING database in 2017: Quality-controlled protein-protein association networks, made broadly accessible. Nucleic Acids Res. 2017, 45, D362–D368. [Google Scholar] [CrossRef] [Green Version]
  22. Klinke, D.J., 2nd. A multiscale systems perspective on cancer, immunotherapy, and Interleukin-12. Mol. Cancer 2010, 9, 242. [Google Scholar] [CrossRef] [Green Version]
  23. Dos Santos, C.; Demur, C.; Bardet, V.; Prade-Houdellier, N.; Payrastre, B.; Recher, C. A critical role for Lyn in acute myeloid leukemia. Blood 2008, 111, 2269–2279. [Google Scholar] [CrossRef]
  24. Xu, Z.; Wang, M.; Wang, L.; Wang, Y.; Zhao, X.; Rao, Q.; Wang, J. Aberrant expression of TSC2 gene in the newly diagnosed acute leukemia. Leuk. Res. 2009, 33, 891–897. [Google Scholar] [CrossRef] [PubMed]
  25. Darwish, N.H.; Sudha, T.; Godugu, K.; Elbaz, O.; Abdelghaffar, H.A.; Hassan, E.E.; Mousa, S.A. Acute myeloid leukemia stem cell markers in prognosis and targeted therapy: Potential impact of BMI-1, TIM-3 and CLL-1. Oncotarget 2016, 7, 57811–57820. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Xiao, H.; Cheng, Q.; Wu, X.; Tang, Y.; Liu, J.; Li, X. ADAR1 may be involved in the proliferation of acute myeloid leukemia cells via regulation of the Wnt pathway. Cancer Manag. Res. 2019, 11, 8547–8555. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  27. Wang, R.Q.; Chen, C.J.; Jing, Y.; Qin, J.Y.; Li, Y.; Chen, G.F.; Zhou, W.; Li, Y.H.; Wang, J.; Li, D.W.; et al. Characteristics and prognostic significance of genetic mutations in acute myeloid leukemia based on a targeted next-generation sequencing technique. Cancer Med. 2020, 9, 8457–8467. [Google Scholar] [CrossRef]
  28. Xu, J.; Zhang, Y.; Hu, J.; Ren, Y.; Wang, H. Clinical features and prognosis of normal karyotype acute myeloid leukemia pediatric patients with WT1 mutations: An analysis based on TCGA database. Hematology 2020, 25, 79–84. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  29. Haubner, S.; Perna, F.; Kohnke, T.; Schmidt, C.; Berman, S.; Augsberger, C.; Schnorfeil, F.M.; Krupka, C.; Lichtenegger, F.S.; Liu, X.; et al. Coexpression profile of leukemic stem cell markers for combinatorial targeted therapy in AML. Leukemia 2019, 33, 64–74. [Google Scholar] [CrossRef]
  30. Abdellateif, M.S.; Kassem, A.B.; El-Meligui, Y.M. Combined Expression of CD34 and FLT3-Internal Tandem Duplication Mutation Predicts Poor Response to Treatment in Acute Myeloid Leukemia. Int. J. Gen. Med. 2020, 13, 867–879. [Google Scholar] [CrossRef]
  31. Vollmer, M.; Li, L.; Schmitt, A.; Greiner, J.; Reinhardt, P.; Ringhoffer, M.; Wiesneth, M.; Dohner, H.; Schmitt, M. Expression of human leucocyte antigens and co-stimulatory molecules on blasts of patients with acute myeloid leukaemia. Br. J. Haematol. 2003, 120, 1000–1008. [Google Scholar] [CrossRef]
  32. Zhu, Y.M.; Wang, P.P.; Huang, J.Y.; Chen, Y.S.; Chen, B.; Dai, Y.J.; Yan, H.; Hu, Y.; Cheng, W.Y.; Ma, T.T.; et al. Gene mutational pattern and expression level in 560 acute myeloid leukemia patients and their clinical relevance. J. Transl. Med. 2017, 15, 178. [Google Scholar] [CrossRef] [Green Version]
  33. Lin, X.; Fang, Q.; Chen, S.; Zhe, N.; Chai, Q.; Yu, M.; Zhang, Y.; Wang, Z.; Wang, J. Heme oxygenase-1 suppresses the apoptosis of acute myeloid leukemia cells via the JNK/c-JUN signaling pathway. Leuk. Res. 2015, 39, 544–552. [Google Scholar] [CrossRef]
  34. Tian, C.; Yu, Y.; Jia, Y.; Zhu, L.; Zhang, Y. HES1 activation suppresses proliferation of leukemia cells in acute myeloid leukemia. Ann. Hematol. 2015, 94, 1477–1483. [Google Scholar] [CrossRef] [PubMed]
  35. Xu, L.; Xu, J.; Ma, S.; Li, X.; Zhu, M.; Chen, S.; Han, Y.; Tang, X.; Fu, Z.; Qiu, H.; et al. High Tim-3 expression on AML blasts could enhance chemotherapy sensitivity. Oncotarget 2017, 8, 102088–102096. [Google Scholar] [CrossRef]
  36. Basharat, M.; Khan, S.A.; Din, N.U.; Ahmed, D. Immunophenotypic characterisation of morphologically diagnosed cases of Acute Myeloid Leukaemia (AML). Pak. J. Med. Sci. 2019, 35, 470–476. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  37. Alanazi, B.; Munje, C.R.; Rastogi, N.; Williamson, A.J.K.; Taylor, S.; Hole, P.S.; Hodges, M.; Doyle, M.; Baker, S.; Gilkes, A.F.; et al. Integrated nuclear proteomics and transcriptomics identifies S100A4 as a therapeutic target in acute myeloid leukemia. Leukemia 2020, 34, 427–440. [Google Scholar] [CrossRef] [PubMed]
  38. Greiner, J.; Brown, E.; Bullinger, L.; Hills, R.K.; Morris, V.; Dohner, H.; Mills, K.I.; Guinn, B.A. Survivin’ Acute Myeloid Leukaemia-A Personalised Target for inv(16) Patients. Int. J. Mol. Sci. 2021, 22, 10482. [Google Scholar] [CrossRef]
  39. Shin, S.Y.; Lee, S.T.; Kim, H.J.; Cho, E.H.; Kim, J.W.; Park, S.; Jung, C.W.; Kim, S.H. Mutation profiling of 19 candidate genes in acute myeloid leukemia suggests significance of DNMT3A mutations. Oncotarget 2016, 7, 54825–54837. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  40. Dou, B.; Jiang, Z.; Chen, X.; Wang, C.; Wu, J.; An, J.; Sheng, G. Oncogenic Long Noncoding RNA DARS-AS1 in Childhood Acute Myeloid Leukemia by Binding to microRNA-425. Technol. Cancer Res. Treat. 2020, 19, 1533033820965580. [Google Scholar] [CrossRef]
  41. Padro, T.; Bieker, R.; Ruiz, S.; Steins, M.; Retzlaff, S.; Burger, H.; Buchner, T.; Kessler, T.; Herrera, F.; Kienast, J.; et al. Overexpression of vascular endothelial growth factor (VEGF) and its cellular receptor KDR (VEGFR-2) in the bone marrow of patients with acute myeloid leukemia. Leukemia 2002, 16, 1302–1310. [Google Scholar] [CrossRef]
  42. Assi, R.; Gur, H.D.; Loghavi, S.; Konoplev, S.N.; Konopleva, M.; Daver, N.; Tashakori, M.; Kadia, T.; Routbort, M.; Salem, A.; et al. P53 protein overexpression in de novo acute myeloid leukemia patients with normal diploid karyotype correlates with FLT3 internal tandem duplication and worse relapse-free survival. Am. J. Hematol. 2018, 93, 1376–1383. [Google Scholar] [CrossRef] [Green Version]
  43. Venditti, A.; Del Poeta, G.; Buccisano, F.; Tamburini, A.; Cox-Froncillo, M.C.; Aronica, G.; Bruno, A.; Del Moro, B.; Epiceno, A.M.; Battaglia, A.; et al. Prognostic relevance of the expression of Tdt and CD7 in 335 cases of acute myeloid leukemia. Leukemia 1998, 12, 1056–1063. [Google Scholar] [CrossRef] [Green Version]
  44. Djunic, I.; Virijevic, M.; Djurasinovic, V.; Novkovic, A.; Colovic, N.; Kraguljac-Kurtovic, N.; Vidovic, A.; Suvajdzic-Vukovic, N.; Tomin, D. Prognostic significance of CD56 antigen expression in patients with acute myeloid leukemia. Med. Oncol. 2012, 29, 2077–2082. [Google Scholar] [CrossRef] [PubMed]
  45. Du, W.; Lu, C.; Zhu, X.; Hu, D.; Chen, X.; Li, J.; Liu, W.; Zhu, J.; He, Y.; Yao, J. Prognostic significance of CXCR4 expression in acute myeloid leukemia. Cancer Med. 2019, 8, 6595–6603. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Aref, S.; Azmy, E.; Ibrahim, L.; Sabry, M.; Agdar, M.E. Prognostic value of CD25/CD123 pattern of expression in acute myeloid leukemia patients with normal cytogenetic. Leuk. Res. Rep. 2020, 13, 100203. [Google Scholar] [CrossRef]
  47. Plesa, C.; Chelghoum, Y.; Plesa, A.; Elhamri, M.; Tigaud, I.; Michallet, M.; Dumontet, C.; Thomas, X. Prognostic value of immunophenotyping in elderly patients with acute myeloid leukemia: A single-institution experience. Cancer 2008, 112, 572–580. [Google Scholar] [CrossRef] [PubMed]
  48. Cheng, Y.; Jia, M.; Chen, Y.; Zhao, H.; Luo, Z.; Tang, Y. Re-evaluation of various molecular targets located on CD34(+)CD38(-)Lin(-) leukemia stem cells and other cell subsets in pediatric acute myeloid leukemia. Oncol. Lett. 2016, 11, 891–897. [Google Scholar] [CrossRef] [Green Version]
  49. Lin, Y.; Huang, Y.; Liang, C.; Xie, S.; Xie, A. Silencing of circTASP1 inhibits proliferation and induces apoptosis of acute myeloid leukaemia cells through modulating miR-515-5p/HMGA2 axis. J. Cell. Mol. Med. 2021, 25, 7367–7380. [Google Scholar] [CrossRef]
  50. Zhang, H.; Wang, P.; Li, Z.; He, Y.; Gan, W.; Jiang, H. Anti-CLL1 Chimeric Antigen Receptor T-Cell Therapy in Children with Relapsed/Refractory Acute Myeloid Leukemia. Clin. Cancer Res. 2021, 27, 3549–3555. [Google Scholar] [CrossRef]
  51. Depreter, B.; Weening, K.E.; Vandepoele, K.; Essand, M.; De Moerloose, B.; Themeli, M.; Cloos, J.; Hanekamp, D.; Moors, I.; D’Hont, I.; et al. TARP is an immunotherapeutic target in acute myeloid leukemia expressed in the leukemic stem cell compartment. Haematologica 2020, 105, 1306–1316. [Google Scholar] [CrossRef]
  52. Rostami, S.; Kazemi, A.; Chahardouli, B.; Mohammadi, S.; Nikbakht, M.; Alizadeh, N.; Mousavi, A.; Alimoghaddam, K.; Ardestani, M.T. The prognostic impact of WT1 expression levels, mutations, and SNP rs16754 in AML patients: A retrospective cohort study. J. Adv. Med. Biomed. Res. 2021, 29, 109–117. [Google Scholar] [CrossRef]
  53. Steger, B.; Floro, L.; Amberger, D.C.; Kroell, T.; Tischer, J.; Kolb, H.J.; Schmetzer, H.M. WT1, PRAME, and PR3 mRNA Expression in Acute Myeloid Leukemia (AML). J. Immunother. 2020, 43, 204–215. [Google Scholar] [CrossRef]
  54. Willier, S.; Rothamel, P.; Hastreiter, M.; Wilhelm, J.; Stenger, D.; Blaeschke, F.; Rohlfs, M.; Kaeuferle, T.; Schmid, I.; Albert, M.H.; et al. CLEC12A and CD33 coexpression as a preferential target for pediatric AML combinatorial immunotherapy. Blood 2021, 137, 1037–1049. [Google Scholar] [CrossRef] [PubMed]
  55. Chashchina, A.; Marklin, M.; Hinterleitner, C.; Salih, H.R.; Heitmann, J.S.; Klimovich, B. DNAM-1/CD226 is functionally expressed on acute myeloid leukemia (AML) cells and is associated with favorable prognosis. Sci. Rep. 2021, 11, 18012. [Google Scholar] [CrossRef] [PubMed]
  56. Churchill, H.R.O.; Fuda, F.S.; Xu, J.; Deng, M.; Zhang, C.C.; An, Z.; Zhang, N.; Chen, P.; Bergstrom, C.; Kansagra, A.; et al. Leukocyte immunoglobulin-like receptor B1 and B4 (LILRB1 and LILRB4): Highly sensitive and specific markers of acute myeloid leukemia with monocytic differentiation. Cytom. B Clin. Cytom. 2021, 100, 476–487. [Google Scholar] [CrossRef] [PubMed]
  57. Wang, J.; Wang, W.; Chen, H.; Li, W.; Huang, T.; Zhang, W.; Ling, W.; Lai, P.; Wang, Y.; Geng, S.; et al. C-Type Lectin-Like Molecule-1 as a Biomarker for Diagnosis and Prognosis in Acute Myeloid Leukemia: A Preliminary Study. Biomed. Res. Int. 2021, 2021, 6643948. [Google Scholar] [CrossRef] [PubMed]
  58. Sun, Y.; Wan, J.; Song, Q.; Luo, C.; Li, X.; Luo, Y.; Huang, X.; Ding, R.; Li, H.; Hou, Y.; et al. Prognostic Significance of CD56 Antigen Expression in Patients with De Novo Non-M3 Acute Myeloid Leukemia. Biomed. Res. Int. 2021, 2021, 1929357. [Google Scholar] [CrossRef]
  59. Mi, H.; Muruganujan, A.; Ebert, D.; Huang, X.; Thomas, P.D. PANTHER version 14: More genomes, a new PANTHER GO-slim and improvements in enrichment analysis tools. Nucleic Acids Res. 2019, 47, D419–D426. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  60. Milholland, B.; Auton, A.; Suh, Y.; Vijg, J. Age-related somatic mutations in the cancer genome. Oncotarget 2015, 6, 24627–24635. [Google Scholar] [CrossRef] [Green Version]
  61. Mak, I.W.; Evaniew, N.; Ghert, M. Lost in translation: Animal models and clinical trials in cancer treatment. Am. J. Transl. Res. 2014, 6, 114–118. [Google Scholar]
  62. Boettcher, A.N.; Loving, C.L.; Cunnick, J.E.; Tuggle, C.K. Development of Severe Combined Immunodeficient (SCID) Pig Models for Translational Cancer Modeling: Future Insights on How Humanized SCID Pigs Can Improve Preclinical Cancer Research. Front. Oncol. 2018, 8, 559. [Google Scholar] [CrossRef] [Green Version]
  63. Tian, H.; Lyu, Y.; Yang, Y.G.; Hu, Z. Humanized Rodent Models for Cancer Research. Front. Oncol. 2020, 10, 1696. [Google Scholar] [CrossRef]
  64. Marsh, V.; Mwangome, N.; Jao, I.; Wright, K.; Molyneux, S.; Davies, A. Who should decide about children’s and adolescents’ participation in health research? The views of children and adults in rural Kenya. BMC Med. Ethics 2019, 20, 41. [Google Scholar] [CrossRef] [Green Version]
  65. O’Dwyer, K.; Freyer, D.R.; Horan, J.T. Treatment strategies for adolescent and young adult patients with acute myeloid leukemia. Blood 2018, 132, 362–368. [Google Scholar] [CrossRef] [Green Version]
  66. Xue, J.; Xie, X.J.; Lin, M.F. Expression and clinical significance of antiapoptotic gene (survivin) in NB4 and acute promyelocytic leukemia cells. Sci. World J. 2012, 2012, 937087. [Google Scholar] [CrossRef] [Green Version]
  67. Zareifar, S.; Ghorbani, S.; Monabbati, A.; Bordbar, M.R.; Zekavat, O.R.; Abdolkarimi, B.; Haghpanah, S. Expression of antiapoptotic proteins livin and survivin in pediatric AML patients, as prognostic markers. Pediatr. Hematol. Oncol. 2018, 35, 250–256. [Google Scholar] [CrossRef] [PubMed]
  68. Warrier, N.M.; Agarwal, P.; Kumar, P. Emerging Importance of Survivin in Stem Cells and Cancer: The Development of New Cancer Therapeutics. Stem Cell Rev. Rep. 2020, 16, 828–852. [Google Scholar] [CrossRef] [PubMed]
  69. Ling, X.; Cao, S.; Cheng, Q.; Keefe, J.T.; Rustum, Y.M.; Li, F. A novel small molecule FL118 that selectively inhibits survivin, Mcl-1, XIAP and cIAP2 in a p53-independent manner, shows superior antitumor activity. PLoS ONE 2012, 7, e45571. [Google Scholar] [CrossRef] [Green Version]
  70. Ling, X.; Wu, W.; Fan, C.; Xu, C.; Liao, J.; Rich, L.J.; Huang, R.Y.; Repasky, E.A.; Wang, X.; Li, F. An ABCG2 non-substrate anticancer agent FL118 targets drug-resistant cancer stem-like cells and overcomes treatment resistance of human pancreatic cancer. J. Exp. Clin. Cancer Res. 2018, 37, 240. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  71. Prasad, V.; Mailankody, S. Research and Development Spending to Bring a Single Cancer Drug to Market and Revenues After Approval. JAMA Intern. Med. 2017, 177, 1569–1575. [Google Scholar] [CrossRef] [PubMed]
  72. Thompson, P.A. Occupy BTK: The key to controlling CLL. Blood 2020, 136, 4–6. [Google Scholar] [CrossRef] [PubMed]
  73. Isidori, A.; Cerchione, C.; Daver, N.; DiNardo, C.; Garcia-Manero, G.; Konopleva, M.; Jabbour, E.; Ravandi, F.; Kadia, T.; Burguera, A.F.; et al. Immunotherapy in Acute Myeloid Leukemia: Where We Stand. Front. Oncol. 2021, 11, 656218. [Google Scholar] [CrossRef]
  74. Chapuis, A.G.; Egan, D.N.; Bar, M.; Schmitt, T.M.; McAfee, M.S.; Paulson, K.G.; Voillet, V.; Gottardo, R.; Ragnarsson, G.B.; Bleakley, M.; et al. T cell receptor gene therapy targeting WT1 prevents acute myeloid leukemia relapse post-transplant. Nat. Med. 2019, 25, 1064–1072. [Google Scholar] [CrossRef] [PubMed]
  75. He, X.; Feng, Z.; Ma, J.; Ling, S.; Cao, Y.; Gurung, B.; Wu, Y.; Katona, B.W.; O’Dwyer, K.P.; Siegel, D.L.; et al. Bispecific and split CAR T cells targeting CD13 and TIM3 eradicate acute myeloid leukemia. Blood 2020, 135, 713–723. [Google Scholar] [CrossRef] [PubMed]
  76. Tran, V.T.; Phan, T.T.; Mac, H.P.; Tran, T.T.; Ho, T.T.; Pho, S.P.; Nguyen, V.N.; Vo, T.M.; Nguyen, H.T.; Le, T.T.; et al. The diagnostic power of CD117, CD13, CD56, CD64, and MPO in rapid screening acute promyelocytic leukemia. BMC Res. Notes 2020, 13, 394. [Google Scholar] [CrossRef] [PubMed]
  77. Piedfer, M.; Dauzonne, D.; Tang, R.; N’Guyen, J.; Billard, C.; Bauvois, B. Aminopeptidase-N/CD13 is a potential proapoptotic target in human myeloid tumor cells. FASEB J. 2011, 25, 2831–2842. [Google Scholar] [CrossRef] [PubMed]
  78. Bauvois, B.; Duzonne, D. Aminopeptidase-N/CD13 (EC 3.4.11.2) inhibitors: Chemistry, biological evaluations, and therapeutic prospects. Med. Res. Rev. 2005, 26, 88–130. [Google Scholar] [CrossRef] [PubMed]
  79. Haouas, H. Angiogenesis and acute myeloid leukemia. Hematology 2014, 19, 311–323. [Google Scholar] [CrossRef] [PubMed]
  80. Moriya, J.; Minamino, T. Angiogenesis, Cancer, and Vascular Aging. Front. Cardiovasc. Med. 2017, 4, 65. [Google Scholar] [CrossRef]
  81. Katuri, V.; Gerber, S.; Qiu, X.; McCarty, G.; Goldstein, S.D.; Hammers, H.; Montgomery, E.; Chen, A.R.; Loeb, D.M. WT1 regulates angiogenesis in Ewing Sarcoma. Oncotarget 2014, 5, 2436–2449. [Google Scholar] [CrossRef] [Green Version]
  82. Sanhueza, C.; Wehinger, S.; Castillo Bennett, J.; Valenzuela, M.; Owen, G.I.; Quest, A.F. The twisted survivin connection to angiogenesis. Mol. Cancer 2015, 14, 198. [Google Scholar] [CrossRef] [Green Version]
  83. Zhao, Y.; Adjei, A.A. Targeting Angiogenesis in Cancer Therapy: Moving Beyond Vascular Endothelial Growth Factor. Oncologist 2015, 20, 660–673. [Google Scholar] [CrossRef] [Green Version]
  84. Dohner, H.; Weisdorf, D.J.; Bloomfield, C.D. Acute Myeloid Leukemia. N. Engl. J. Med. 2015, 373, 1136–1152. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  85. Welner, R.S.; Amabile, G.; Bararia, D.; Czibere, A.; Yang, H.; Zhang, H.; Pontes, L.L.; Ye, M.; Levantini, E.; Di Ruscio, A.; et al. Treatment of chronic myelogenous leukemia by blocking cytokine alterations found in normal stem and progenitor cells. Cancer Cell. 2015, 27, 671–681. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  86. Carey, A.; Edwards, D.K.t.; Eide, C.A.; Newell, L.; Traer, E.; Medeiros, B.C.; Pollyea, D.A.; Deininger, M.W.; Collins, R.H.; Tyner, J.W.; et al. Identification of Interleukin-1 by Functional Screening as a Key Mediator of Cellular Expansion and Disease Progression in Acute Myeloid Leukemia. Cell. Rep. 2017, 18, 3204–3218. [Google Scholar] [CrossRef] [PubMed]
  87. Li, H.; Wu, M.; Zhao, X. Role of chemokine systems in cancer and inflammatory diseases. MedComm 2022, 3, e147. [Google Scholar] [CrossRef] [PubMed]
  88. Berraondo, P.; Sanmamed, M.F.; Ochoa, M.C.; Etxeberria, I.; Aznar, M.A.; Perez-Gracia, J.L.; Rodriguez-Ruiz, M.E.; Ponz-Sarvise, M.; Castanon, E.; Melero, I. Cytokines in clinical cancer immunotherapy. Br. J. Cancer 2019, 120, 6–15. [Google Scholar] [CrossRef] [Green Version]
  89. Yao, C.J.; Du, W.; Chen, H.B.; Xiao, S.; Wang, C.H.; Fan, Z.L. Associations of IL-10 gene polymorphisms with acute myeloid leukemia in Hunan, China. Asian Pac. J. Cancer Prev. 2013, 14, 2439–2442. [Google Scholar] [CrossRef]
  90. Vaidya, A.; Kale, V.P. TGF-beta signaling and its role in the regulation of hematopoietic stem cells. Syst. Synth. Biol. 2015, 9, 1–10. [Google Scholar] [CrossRef] [Green Version]
  91. Grob, T.; Al Hinai, A.S.A.; Sanders, M.A.; Kavelaars, F.G.; Rijken, M.; Gradowska, P.L.; Biemond, B.J.; Breems, D.A.; Maertens, J.; van Marwijk Kooy, M.; et al. Molecular characterization of mutant TP53 acute myeloid leukemia and high-risk myelodysplastic syndrome. Blood 2022, 139, 2347–2354. [Google Scholar] [CrossRef]
  92. Stengel, A.; Kern, W.; Haferlach, T.; Meggendorfer, M.; Fasan, A.; Haferlach, C. The impact of TP53 mutations and TP53 deletions on survival varies between AML, ALL, MDS and CLL: An analysis of 3307 cases. Leukemia 2017, 31, 705–711. [Google Scholar] [CrossRef]
  93. Cluzeau, T.; Loschi, M.; Fenaux, P.; Komrokji, R.; Sallman, D.A. Personalized Medicine for TP53 Mutated Myelodysplastic Syndromes and Acute Myeloid Leukemia. Int. J. Mol. Sci. 2021, 22, 10105. [Google Scholar] [CrossRef]
  94. Fletcher, D.; Brown, E.; Javadala, J.; Uysal-Onganer, P.; Guinn, B.A. microRNA expression in acute myeloid leukaemia: New targets for therapy? EJHaem 2022, 3, 596–608. [Google Scholar] [CrossRef] [PubMed]
  95. Kaner, J.D.; Mencia-Trinchant, N.; Schaap, A.; Roboz, G.J.; Lee, S.; Desai, P.; Samuel, M.; Krichevsky, S.; Udoyen, I.; Has-sane, D.C.; et al. Acute Myeloid Leukemia (AML) with Somatic Mutations in PTPN11 Is Associated with Treatment Resistance and Poor Overall Survival. Blood 2018, 132, 2760. [Google Scholar] [CrossRef]
  96. Loh, M.L.; Reynolds, M.G.; Vattikuti, S.; Gerbing, R.B.; Alonzo, T.A.; Carlson, E.; Cheng, J.W.; Lee, C.M.; Lange, B.J.; Meshinchi, S.; et al. PTPN11 mutations in pediatric patients with acute myeloid leukemia: Results from the Children’s Cancer Group. Leukemia 2004, 18, 1831–1834. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  97. Fobare, S.; Kohlschmidt, J.; Ozer, H.G.; Mrozek, K.; Nicolet, D.; Mims, A.S.; Garzon, R.; Blachly, J.S.; Orwick, S.; Carroll, A.J.; et al. Molecular, clinical, and prognostic implications of PTPN11 mutations in acute myeloid leukemia. Blood Adv. 2022, 6, 1371–1380. [Google Scholar] [CrossRef]
  98. Campbell, I.L. Cytokine-mediated inflammation, tumorigenesis, and disease-associated JAK/STAT/SOCS signaling circuits in the CNS. Brain Res. Brain Res. Rev. 2005, 48, 166–177. [Google Scholar] [CrossRef] [PubMed]
  99. Liu, Q.; Ai, B.; Kong, X.; Wang, X.; Qi, Y.; Wang, Z.; Fang, Y.; Wang, J. JAK2 expression is correlated with the molecular and clinical features of breast cancer as a favorable prognostic factor. Int. Immunopharmacol. 2021, 90, 107186. [Google Scholar] [CrossRef]
  100. Vicente, C.; Vazquez, I.; Marcotegui, N.; Conchillo, A.; Carranza, C.; Rivell, G.; Bandres, E.; Cristobal, I.; Lahortiga, I.; Calasanz, M.J.; et al. JAK2-V617F activating mutation in acute myeloid leukemia: Prognostic impact and association with other molecular markers. Leukemia 2007, 21, 2386–2390. [Google Scholar] [CrossRef]
  101. Ding, W.; Li, D.; Zhuang, C.; Wei, P.; Mou, W.; Zhang, L.; Liang, H.; Liu, Y. Essential thrombocythemia during treatment of acute myeloid leukemia with JAK2 V617F mutation: A case report of a CARE-compliant article. Medicine 2018, 97, e11331. [Google Scholar] [CrossRef]
  102. Tzelepis, K.; Koike-Yusa, H.; De Braekeleer, E.; Li, Y.; Metzakopian, E.; Dovey, O.M.; Mupo, A.; Grinkevich, V.; Li, M.; Mazan, M.; et al. A CRISPR Dropout Screen Identifies Genetic Vulnerabilities and Therapeutic Targets in Acute Myeloid Leukemia. Cell. Rep. 2016, 17, 1193–1205. [Google Scholar] [CrossRef] [Green Version]
  103. Ley, T.J.; Ding, L.; Walter, M.J.; McLellan, M.D.; Lamprecht, T.; Larson, D.E.; Kandoth, C.; Payton, J.E.; Baty, J.; Welch, J.; et al. DNMT3A mutations in acute myeloid leukemia. N. Engl. J. Med. 2010, 363, 2424–2433. [Google Scholar] [CrossRef] [Green Version]
  104. Contieri, B.; Duarte, B.K.L.; Lazarini, M. Updates on DNA methylation modifiers in acute myeloid leukemia. Ann. Hematol. 2020, 99, 693–701. [Google Scholar] [CrossRef] [PubMed]
  105. Sexauer, A.N.; Tasian, S.K. Targeting FLT3 Signaling in Childhood Acute Myeloid Leukemia. Front. Pediatr. 2017, 5, 248. [Google Scholar] [CrossRef] [Green Version]
  106. Koedam, J.; Wermke, M.; Ehninger, A.; Cartellieri, M.; Ehninger, G. Chimeric antigen receptor T-cell therapy in acute myeloid leukemia. Curr. Opin. Hematol. 2022, 29, 74–83. [Google Scholar] [CrossRef]
  107. Shrestha, E.; Liang, R.; Sirochinsky, C.; Jehuda, R.B.; Sandler, V. Preclinical Development of Anti-FLT3 CAR-T Therapy for the Treatment of Acute Myeloid Leukemia. Blood 2020, 136, 4–5. [Google Scholar] [CrossRef]
  108. Ge, S.S.; Liu, S.B.; Xue, S.L. Developments and challenges of FLT3 inhibitors in acute myeloid leukemia. Front. Oncol. 2022, 12, 996438. [Google Scholar] [CrossRef] [PubMed]
  109. Traer, E.; Martinez, J.; Javidi-Sharifi, N.; Agarwal, A.; Dunlap, J.; English, I.; Kovacsovics, T.; Tyner, J.W.; Wong, M.; Druker, B.J. FGF2 from Marrow Microenvironment Promotes Resistance to FLT3 Inhibitors in Acute Myeloid Leukemia. Cancer Res. 2016, 76, 6471–6482. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  110. Zeng, Z.; Shi, Y.X.; Samudio, I.J.; Wang, R.Y.; Ling, X.; Frolova, O.; Levis, M.; Rubin, J.B.; Negrin, R.R.; Estey, E.H.; et al. Targeting the leukemia microenvironment by CXCR4 inhibition overcomes resistance to kinase inhibitors and chemotherapy in AML. Blood 2009, 113, 6215–6224. [Google Scholar] [CrossRef] [Green Version]
  111. Wakita, S.; Yamaguchi, H.; Omori, I.; Terada, K.; Ueda, T.; Manabe, E.; Kurosawa, S.; Iida, S.; Ibaraki, T.; Sato, Y.; et al. Mutations of the epigenetics-modifying gene (DNMT3a, TET2, IDH1/2) at diagnosis may induce FLT3-ITD at relapse in de novo acute myeloid leukemia. Leukemia 2013, 27, 1044–1052. [Google Scholar] [CrossRef] [Green Version]
  112. Jaini, R.; Loya, M.G.; Eng, C. Immunotherapeutic target expression on breast tumors can be amplified by hormone receptor antagonism: A novel strategy for enhancing efficacy of targeted immunotherapy. Oncotarget 2017, 8, 32536–32549. [Google Scholar] [CrossRef] [Green Version]
  113. Cummins, K.D.; Gill, S. Chimeric antigen receptor T-cell therapy for acute myeloid leukemia: How close to reality? Haematologica 2019, 104, 1302–1308. [Google Scholar] [CrossRef] [Green Version]
  114. Sommer, C.; Cheng, H.Y.; Nguyen, D.; Dettling, D.; Yeung, Y.A.; Sutton, J.; Hamze, M.; Valton, J.; Smith, J.; Djuretic, I.; et al. Allogeneic FLT3 CAR T Cells with an Off-Switch Exhibit Potent Activity against AML and Can Be Depleted to Expedite Bone Marrow Recovery. Mol. Ther. 2020, 28, 2237–2251. [Google Scholar] [CrossRef] [PubMed]
  115. Vishwasrao, P.; Li, G.; Boucher, J.C.; Smith, D.L.; Hui, S.K. Emerging CAR T Cell Strategies for the Treatment of AML. Cancers 2022, 14, 1241. [Google Scholar] [CrossRef] [PubMed]
  116. Greiner, J.; Goetz, M.; Schuler, P.J.; Bulach, C.; Hofmann, S.; Schrezenmeier, H.; Dohner, H.; Schneider, V.; Guinn, B.A. Enhanced stimulation of antigen-specific immune responses against nucleophosmin 1 mutated acute myeloid leukaemia by an anti-programmed death 1 antibody. Br. J. Haematol. 2022, 198, 866–874. [Google Scholar] [CrossRef] [PubMed]
  117. Guinn, B.A.; Schuler, P.J.; Schrezenmeier, H.; Hofmann, S.; Weiss, J.; Bulach, C.; Götz, M.; Greiner, J. A combination of the immunotherapeutic drug anti-programmed death 1 with lenalidomide enhances specific T cell immune responses against acute myeloid leukemia cells. Int. J. Med. Sci. 2023, 24, 9285. [Google Scholar] [CrossRef]
Figure 1. Prisma-P flow diagram to show the screening process that determined whether articles met the inclusion criteria at each stage; if not, they were removed. In total, 36 articles, describing 71 targets, were identified for the quality assurance step.
Figure 1. Prisma-P flow diagram to show the screening process that determined whether articles met the inclusion criteria at each stage; if not, they were removed. In total, 36 articles, describing 71 targets, were identified for the quality assurance step.
Ijms 24 09667 g001
Figure 2. Venn diagram to illustrate which of the affected genes within adult and paediatric AML overlap with one another. Those that lie within the overlapping intersection should be assessed as potential targets for unified immunotherapy treatment approaches.
Figure 2. Venn diagram to illustrate which of the affected genes within adult and paediatric AML overlap with one another. Those that lie within the overlapping intersection should be assessed as potential targets for unified immunotherapy treatment approaches.
Ijms 24 09667 g002
Figure 3. Prioritisation of the 63 potential treatment targets identified in AML by systematic review and found in articles that remained after quality control. Prioritisation was performed using the National Cancer Institute Pilot Project as a model [10], based on nine predefined, pre-weighted criteria. The antigens were scored to determine their suitability for further investigation as therapeutically effective cancer vaccines.
Figure 3. Prioritisation of the 63 potential treatment targets identified in AML by systematic review and found in articles that remained after quality control. Prioritisation was performed using the National Cancer Institute Pilot Project as a model [10], based on nine predefined, pre-weighted criteria. The antigens were scored to determine their suitability for further investigation as therapeutically effective cancer vaccines.
Ijms 24 09667 g003
Figure 4. PANTHER was used to analyse the pathways that are linked to the 20 highest scoring targets after prioritization in both adult and paediatric AML. Pathways with the highest number of genes linked to them should be investigated further as key pathways for AML development. From those tested, only one pathway differs between paediatric and adult AML, which suggests strong similarities between the two and increases the possibility of an overlapping treatment.
Figure 4. PANTHER was used to analyse the pathways that are linked to the 20 highest scoring targets after prioritization in both adult and paediatric AML. Pathways with the highest number of genes linked to them should be investigated further as key pathways for AML development. From those tested, only one pathway differs between paediatric and adult AML, which suggests strong similarities between the two and increases the possibility of an overlapping treatment.
Ijms 24 09667 g004
Figure 5. Protein–protein interaction analysis was performed via STRING with confidence levels set to 0.7 and the species set to Homo sapiens. In both figures, the top five targets (red nodes) are shown after prioritization for adult and paediatric AML. (A) STRING network of the 20 highest scoring potential therapeutic targets for adult AML identified after prioritization. TP53 had the most connections for the adult network. (B) STRING network of the 20 highest scoring potential therapeutic targets for paediatric AML identified after prioritization. PTPN11 had the most connections for the paediatric network.
Figure 5. Protein–protein interaction analysis was performed via STRING with confidence levels set to 0.7 and the species set to Homo sapiens. In both figures, the top five targets (red nodes) are shown after prioritization for adult and paediatric AML. (A) STRING network of the 20 highest scoring potential therapeutic targets for adult AML identified after prioritization. TP53 had the most connections for the adult network. (B) STRING network of the 20 highest scoring potential therapeutic targets for paediatric AML identified after prioritization. PTPN11 had the most connections for the paediatric network.
Ijms 24 09667 g005
Table 1. Inclusion criteria.
Table 1. Inclusion criteria.
CategoryIncluded
Publication dateAll
Subgroup of interestM4/M5 Acute Myeloid Leukaemia
(FAB or WHO diagnosis)
Geographic location of publicationAll
LanguageEnglish
ParticipantsAll age groups, recently diagnosed
Reported outcomesAntigen expression %
Study designOver 10 participants
Publication typeOriginal research
Table 2. Publication database search strategy (Scopus example).
Table 2. Publication database search strategy (Scopus example).
MeSH Terms and Boolean Operators Used to Search TermsFilters
“acute myeloid leukaemia” OR “acute myeloid leukemia” OR “acute myelogenous leukaemia” OR “acute myelogenous leukemia” AND antigen OR “antigen therapy” AND humanSpecies: human
Source type: journal
Language: English
Document type: article, review
Age: adult and paediatric
Table 3. Risk of bias and quality assurance using the QUADAS-2 system of article assessment.
Table 3. Risk of bias and quality assurance using the QUADAS-2 system of article assessment.
StudyRisk of BiasApplicability Concerns
Patient
Selection
InterventionReference StandardOutcomePatient
Selection
InterventionReference Standard
Dos Santos et al. [23]Ijms 24 09667 i001Ijms 24 09667 i002Ijms 24 09667 i003Ijms 24 09667 i004Ijms 24 09667 i005Ijms 24 09667 i006Ijms 24 09667 i007
Xu et al. [24]Ijms 24 09667 i008Ijms 24 09667 i009Ijms 24 09667 i010Ijms 24 09667 i011Ijms 24 09667 i012Ijms 24 09667 i013Ijms 24 09667 i014
Darwish et al. [25]Ijms 24 09667 i015Ijms 24 09667 i016Ijms 24 09667 i017Ijms 24 09667 i018Ijms 24 09667 i019Ijms 24 09667 i020Ijms 24 09667 i021
Xiao et al. [26]Ijms 24 09667 i022Ijms 24 09667 i023Ijms 24 09667 i024Ijms 24 09667 i025Ijms 24 09667 i026Ijms 24 09667 i027Ijms 24 09667 i028
Wang et al. [27]Ijms 24 09667 i029Ijms 24 09667 i030Ijms 24 09667 i031Ijms 24 09667 i032Ijms 24 09667 i033Ijms 24 09667 i034Ijms 24 09667 i035
Xu et al. [28]Ijms 24 09667 i036Ijms 24 09667 i037Ijms 24 09667 i038Ijms 24 09667 i039Ijms 24 09667 i040Ijms 24 09667 i041Ijms 24 09667 i042
Haubner et al. [29]Ijms 24 09667 i043Ijms 24 09667 i044Ijms 24 09667 i045Ijms 24 09667 i046Ijms 24 09667 i047Ijms 24 09667 i048Ijms 24 09667 i049
Abdellateif et al. [30]Ijms 24 09667 i050Ijms 24 09667 i051Ijms 24 09667 i052Ijms 24 09667 i053Ijms 24 09667 i054Ijms 24 09667 i055Ijms 24 09667 i056
Vollmer et al. [31]Ijms 24 09667 i057Ijms 24 09667 i058Ijms 24 09667 i059Ijms 24 09667 i060Ijms 24 09667 i061Ijms 24 09667 i062Ijms 24 09667 i063
Zhu et al. [32]Ijms 24 09667 i064Ijms 24 09667 i065Ijms 24 09667 i066Ijms 24 09667 i067Ijms 24 09667 i068Ijms 24 09667 i069Ijms 24 09667 i070
Lin et al. [33]Ijms 24 09667 i071Ijms 24 09667 i072Ijms 24 09667 i073Ijms 24 09667 i074Ijms 24 09667 i075Ijms 24 09667 i076Ijms 24 09667 i077
Tian et al. [34]Ijms 24 09667 i078Ijms 24 09667 i079Ijms 24 09667 i080Ijms 24 09667 i081Ijms 24 09667 i082Ijms 24 09667 i083Ijms 24 09667 i084
Xu et al. [35]Ijms 24 09667 i085Ijms 24 09667 i086Ijms 24 09667 i087Ijms 24 09667 i088Ijms 24 09667 i089Ijms 24 09667 i090Ijms 24 09667 i091
Basharat et al. [36]Ijms 24 09667 i092Ijms 24 09667 i093Ijms 24 09667 i094Ijms 24 09667 i095Ijms 24 09667 i096Ijms 24 09667 i097Ijms 24 09667 i098
Alanazi et al. [37]Ijms 24 09667 i099Ijms 24 09667 i100Ijms 24 09667 i101Ijms 24 09667 i102Ijms 24 09667 i103Ijms 24 09667 i104Ijms 24 09667 i105
Greiner et al. [38]Ijms 24 09667 i106Ijms 24 09667 i107Ijms 24 09667 i108Ijms 24 09667 i109Ijms 24 09667 i110Ijms 24 09667 i111Ijms 24 09667 i112
Shin et al. [39]Ijms 24 09667 i113Ijms 24 09667 i114Ijms 24 09667 i115Ijms 24 09667 i116Ijms 24 09667 i117Ijms 24 09667 i118Ijms 24 09667 i119
Dou et al. [40]Ijms 24 09667 i120Ijms 24 09667 i121Ijms 24 09667 i122Ijms 24 09667 i123Ijms 24 09667 i124Ijms 24 09667 i125Ijms 24 09667 i126
Padró et al. [41]Ijms 24 09667 i127Ijms 24 09667 i128Ijms 24 09667 i129Ijms 24 09667 i130Ijms 24 09667 i131Ijms 24 09667 i132Ijms 24 09667 i133
Assi et al. [42]Ijms 24 09667 i134Ijms 24 09667 i135Ijms 24 09667 i136Ijms 24 09667 i137Ijms 24 09667 i138Ijms 24 09667 i139Ijms 24 09667 i140
Venditti et al. [43]Ijms 24 09667 i141Ijms 24 09667 i142Ijms 24 09667 i143Ijms 24 09667 i144Ijms 24 09667 i145Ijms 24 09667 i146Ijms 24 09667 i147
Djunic et al. [44]Ijms 24 09667 i148Ijms 24 09667 i149Ijms 24 09667 i150Ijms 24 09667 i151Ijms 24 09667 i152Ijms 24 09667 i153Ijms 24 09667 i154
Du et al. [45]Ijms 24 09667 i155Ijms 24 09667 i156Ijms 24 09667 i157Ijms 24 09667 i158Ijms 24 09667 i159Ijms 24 09667 i160Ijms 24 09667 i161
Aref et al. [46]Ijms 24 09667 i162Ijms 24 09667 i163Ijms 24 09667 i164Ijms 24 09667 i165Ijms 24 09667 i166Ijms 24 09667 i167Ijms 24 09667 i168
Plesa et al. [47]Ijms 24 09667 i169Ijms 24 09667 i170Ijms 24 09667 i171Ijms 24 09667 i172Ijms 24 09667 i173Ijms 24 09667 i174Ijms 24 09667 i175
Cheng et al. [48]Ijms 24 09667 i176Ijms 24 09667 i177Ijms 24 09667 i178Ijms 24 09667 i179Ijms 24 09667 i180Ijms 24 09667 i181Ijms 24 09667 i182
Lin et al. [49]Ijms 24 09667 i183Ijms 24 09667 i184Ijms 24 09667 i185Ijms 24 09667 i186Ijms 24 09667 i187Ijms 24 09667 i188Ijms 24 09667 i189
Zhang et al. [50]Ijms 24 09667 i190Ijms 24 09667 i191Ijms 24 09667 i192Ijms 24 09667 i193Ijms 24 09667 i194Ijms 24 09667 i195Ijms 24 09667 i196
Depreter et al. [51]Ijms 24 09667 i197Ijms 24 09667 i198Ijms 24 09667 i199Ijms 24 09667 i200Ijms 24 09667 i201Ijms 24 09667 i202Ijms 24 09667 i203
Rostami et al. [52]Ijms 24 09667 i204Ijms 24 09667 i205Ijms 24 09667 i206Ijms 24 09667 i207Ijms 24 09667 i208Ijms 24 09667 i209Ijms 24 09667 i210
Steger et al. [53]Ijms 24 09667 i211Ijms 24 09667 i212Ijms 24 09667 i213Ijms 24 09667 i214Ijms 24 09667 i215Ijms 24 09667 i216Ijms 24 09667 i217
Willier et al. [54]Ijms 24 09667 i218Ijms 24 09667 i219Ijms 24 09667 i220Ijms 24 09667 i221Ijms 24 09667 i222Ijms 24 09667 i223Ijms 24 09667 i224
Chashchina et al. [55]Ijms 24 09667 i225Ijms 24 09667 i226Ijms 24 09667 i227Ijms 24 09667 i228Ijms 24 09667 i229Ijms 24 09667 i230Ijms 24 09667 i231
Churchill et al. [56]Ijms 24 09667 i232Ijms 24 09667 i233Ijms 24 09667 i234Ijms 24 09667 i235Ijms 24 09667 i236Ijms 24 09667 i237Ijms 24 09667 i238
Wang et al. [57]Ijms 24 09667 i239Ijms 24 09667 i240Ijms 24 09667 i241Ijms 24 09667 i242Ijms 24 09667 i243Ijms 24 09667 i244Ijms 24 09667 i245
Sun et al. [58]Ijms 24 09667 i246Ijms 24 09667 i247Ijms 24 09667 i248Ijms 24 09667 i249Ijms 24 09667 i250Ijms 24 09667 i251Ijms 24 09667 i252
Red circles indicate a risk of bias; Green circles indicate no risk of bias, was identified.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Morris, V.S.; Ghazi, H.; Fletcher, D.M.; Guinn, B.-a. A Direct Comparison, and Prioritisation, of the Immunotherapeutic Targets Expressed by Adult and Paediatric Acute Myeloid Leukaemia Cells: A Systematic Review and Meta-Analysis. Int. J. Mol. Sci. 2023, 24, 9667. https://doi.org/10.3390/ijms24119667

AMA Style

Morris VS, Ghazi H, Fletcher DM, Guinn B-a. A Direct Comparison, and Prioritisation, of the Immunotherapeutic Targets Expressed by Adult and Paediatric Acute Myeloid Leukaemia Cells: A Systematic Review and Meta-Analysis. International Journal of Molecular Sciences. 2023; 24(11):9667. https://doi.org/10.3390/ijms24119667

Chicago/Turabian Style

Morris, Vanessa S., Hanya Ghazi, Daniel M. Fletcher, and Barbara-ann Guinn. 2023. "A Direct Comparison, and Prioritisation, of the Immunotherapeutic Targets Expressed by Adult and Paediatric Acute Myeloid Leukaemia Cells: A Systematic Review and Meta-Analysis" International Journal of Molecular Sciences 24, no. 11: 9667. https://doi.org/10.3390/ijms24119667

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