Next Article in Journal
Precision Medicine in Hematologic Malignancies: Evolving Concepts and Clinical Applications
Previous Article in Journal
An Integrative Genomics Approach for the Discovery of Potential Clinically Actionable Diagnostic and Prognostic Biomarkers in Colorectal Cancer
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Advances and Challenges in Targeted Therapy and Its Combination Strategies for Leukemia

1
Pediatric Hematology Laboratory, Division of Hematology/Oncology, Department of Pediatrics, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen 518107, China
2
Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen 518107, China
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Biomedicines 2025, 13(7), 1652; https://doi.org/10.3390/biomedicines13071652
Submission received: 29 May 2025 / Revised: 28 June 2025 / Accepted: 29 June 2025 / Published: 7 July 2025

Abstract

Leukemia is a group of hematological malignancies with a complex pathogenesis and diverse clinical manifestations. Although traditional treatments such as chemotherapy, radiotherapy, and hematopoietic stem cell transplantation have improved patient outcomes, their efficacy is often limited by non-specificity, drug resistance, and relapse. In recent years, targeted therapy has emerged as a major breakthrough, offering new opportunities for precision medicine in leukemia. The development of molecularly targeted agents has significantly advanced our ability to treat specific leukemia subtypes. However, challenges such as resistance to targeted drugs, adverse effects, and tumor heterogeneity remain significant obstacles. As a result, treatment strategies are shifting from single-agent chemotherapy toward combination therapies that integrate targeted agents, aiming to enhance therapeutic efficacy and reduce the likelihood of resistance. This review summarizes the current research landscape, clinical applications, and limitations of targeted therapies in leukemia, with a focus on recent progress in combination treatment strategies and ongoing clinical trials.

1. Introduction

Leukemia is a class of malignant hematological diseases caused by the abnormal clonal proliferation of hematopoietic stem cells. Its pathogenesis is complex, involving multiple factors such as genetic mutations, environmental exposure, infection, and endocrine factors [1,2,3]. The typical pathological feature is the accumulation of a large number of leukemia cells in the bone marrow and hematopoietic tissues, inhibiting normal hematopoietic function, and it can also infiltrate other organs, leading to anemia, bleeding, infection, and multiple organ dysfunction [4,5,6]. According to the differentiation stage and pathological characteristics of leukemia cells, leukemia can be divided into types such as acute lymphoblastic leukemia (ALL), acute myeloid leukemia (AML), chronic lymphocytic leukemia (CLL), and chronic myeloid leukemia (CML). Among them, ALL is common in children and adolescents, with the peak age of onset between 2 and 5 years old. AML is the most common type in adults, while the course of chronic leukemia is relatively slow but may transform into blast crisis [7,8]. According to statistics, in 2022, the global incidence and mortality of leukemia ranked 13th (486,777 cases) and 10th (305,033 cases), respectively, among all cancers, seriously threatening human health [9].
The traditional treatment methods for leukemia are mainly chemotherapy, but chemotherapy has many serious side effects, which may lead to treatment failure due to treatment-related death or drug resistance [10]. With the emergence of new treatment methods such as targeted therapy and immunotherapy, the treatment of leukemia has shifted from cytotoxic chemotherapy to personalized medical treatment [11]. Unlike traditional chemotherapy, targeted therapy can specifically interfere with target macromolecules related to carcinogenesis, rather than acting on all rapidly dividing cells. In addition, targeted therapy can effectively enhance the therapeutic effect of leukemia by limiting the occurrence of acquired resistance during treatment, improving the efficacy of individual components in cancer treatment mixtures, and reducing related side effects [12,13]. Although targeted drugs have achieved certain success in the treatment of leukemia, single-agent use has many limitations, such as resistance and a series of adverse reactions triggered during treatment, which seriously affect the patient’s quality of life and treatment compliance, thus affecting the therapeutic effect. Therefore, the treatment of leukemia has gradually shifted from single chemotherapy to targeted drug combination therapy, significantly improving the remission rate and survival rate of various types of leukemia [14,15,16].
In this review, we discuss the therapeutic effects and limitations of single-agent use of currently approved targeted drugs for leukemia treatment, such as tyrosine kinase inhibitors (TKIs) and FLT3 inhibitors. Subsequently, we further propose combination therapies in the forms of targeted drugs combined with chemotherapy drugs, internal combination of targeted drugs, targeted drugs combined with chimeric antigen receptor T-cell immunotherapy (CAR-T), and multi-drug combination as key strategies to enhance efficacy, hopefully bringing better treatment prospects for leukemia patients.

2. Targeted Therapy

Targeted therapy improves the precision and effectiveness of leukemia treatment by identifying and attacking specific molecular targets unique to leukemia cells, such as gene mutations or abnormal signaling pathways. Based on the mechanism of action and targets, targeted drugs can be divided into TKIs, FLT3 inhibitors, B-cell signaling pathway inhibitors, anti-apoptotic inhibitors, and immunotherapy drugs, etc.

2.1. Tyrosine Kinase Inhibitors

Tyrosine kinases play a key role in intracellular signal transduction. These enzymes regulate various cellular functions, such as growth, division, migration, and survival, by catalyzing the phosphorylation of tyrosine residues on specific proteins. Their abnormal activation can trigger tumor development, thus becoming important therapeutic targets. TKIs are small-molecule oral inhibitors that block the activity of tyrosine kinases by competitively inhibiting ATP binding at the catalytic tyrosine kinase binding site, thereby inhibiting carcinogenic signal transduction [17]. Currently, various TKIs such as Imatinib, Dasatinib, and Ponatinib have been developed, which can effectively target CML and Philadelphia chromosome-positive acute lymphoblastic leukemia (Ph+ALL) patients.

2.1.1. Imatinib

Before the advent of targeted TKIs, the treatment of leukemia mainly relied on chemotherapy, radiotherapy, and hematopoietic stem cell transplantation (HSCT), but the overall effect was poor. For example, HSCT, as a potential curative method for early CML, is limited by various factors, including whether the patient meets the transplant criteria, the risk of early treatment-related death, and long-term disability caused by chronic graft-versus-host disease [10,18]. In 1960, researchers discovered that the Philadelphia chromosome was the cause of CML development. The ABL gene on chromosome 9 translocated with the BCR gene on chromosome 22, forming the BCR-ABL fusion gene. The tyrosine kinase activity encoded by this gene is uncontrolled, leading to uncontrolled cell division [19]. By the late 1990s, Nick Lydon of Novartis synthesized a compound called CGP57148, the predecessor of Imatinib, which was promoted for CML treatment by oncologist Brian Druker at the Dana-Farber Cancer Institute. The drug underwent its first clinical trial in 1998 and received FDA approval in May 2001, officially becoming a first-line treatment for CML [20,21]. The discovery and application of Imatinib was a major breakthrough in leukemia treatment, opening a new chapter in targeted therapy for leukemia.
As the first-generation TKI, Imatinib (Figure 1) effectively treats CML and Ph+ALL patients by binding to the catalytic domain of the BCR-ABL protein, preventing ATP binding, thereby inhibiting the phosphorylation of tyrosine residues on various substrates, ultimately preventing cancer cell proliferation (Figure 2) [14,22]. Research shows that the 10-year survival rate for Imatinib treatment of CML is 83.3% [22]. It can achieve durable cytogenetic and molecular remission, significantly improving the survival rate of most patients [23]. Clinical trials such as STIM and TWISTER showed that approximately 39–45% of patients with sustained deep molecular response (DMR) can maintain treatment-free remission (TFR) for 3 years or longer after receiving Imatinib treatment [24]. After 6 years of Imatinib treatment, the proportion of patients reaching TFR can reach 21.6% [25]. Moreover, drug discontinuation does not lead to acquired resistance to Imatinib, nor does it trigger further safety issues. Ultimately, nearly 10% of patients can completely stop medication [24,26].
Although Imatinib has a certain therapeutic effect on leukemia, mutations in the BCR-ABL gene, especially the T315I mutation, can trigger drug resistance, limiting its clinical application. Research shows that about 25–30% of CML patients develop resistance to Imatinib [27]. In addition, Imatinib may cause mild to moderate adverse reactions such as superficial edema, pigmentation, and rash during treatment. Keshavamurthy Vinay et al. studied 438 patients and found that these patients took Imatinib for an average of 1820 days. Among them, 53.9% developed melasma-like pigmentation; 18.5% developed periorbital edema; 16% developed oral lichenoid reactions; 9.6% developed skin hypopigmentation, and 2.7% developed bullous pemphigoid [28]. Therefore, it is necessary to develop more effective drugs to reduce drug toxicity and improve the treatment efficiency of leukemia.

2.1.2. Dasatinib

Dasatinib (Figure 1), as a second-generation TKI, is more potent against BCR-ABL activity compared to Imatinib. It can effectively address a wider range of mutation situations, such as the E255K and G250E mutations within the phosphate-binding loop of the ATP-binding pocket, playing a key role in transforming CML into a manageable disease [15]. The 10-year follow-up results of the IRIS trial showed that for CML treatment, its 5-year overall survival (OS) rate reached 96%, treatment failure-free survival reached 95%. At the same time, the 5-year cumulative major molecular response (MMR) rate was 89%, and the MR4.5 rate (i.e., the ratio of abnormal gene copies decreasing by 4.5 logarithms) reached 79%. Among patients who achieved MR4.5, those who maintained this status for ≥2 years, 11% voluntarily chose to stop medication, and the 2-year TFR rate after discontinuation was 58% [16]. This indicates that Dasatinib, as a first-line treatment drug, has good long-term safety and efficacy in newly diagnosed CML and Ph+ALL.
Although Dasatinib is effective for Imatinib-resistant/intolerant CML and Ph+ALL, it may cause adverse reactions such as cardiovascular side effects, pulmonary arterial hypertension, pleural effusion, and bleeding in clinical applications. In addition, kinase resistance mutations, including T315I, disrupt drug binding, leading to increased Dasatinib resistance [29]. Therefore, continuous research and development of new treatment methods are needed to improve the usability of the drug.

2.1.3. Ponatinib

As a third-generation TKI, Ponatinib (Figure 1) is more effective than Dasatinib for CML and Ph+ALL patients with resistance or intolerance, especially those with the T315I mutation (Figure 2) [30]. In 2012, the drug received FDA approval. In October 2013, due to serious safety concerns, Ponatinib was voluntarily withdrawn from the market by its manufacturer. In December of the same year, the drug was reintroduced under revised warnings and precautions. Its indication was limited to patients with the T315I mutation or those who are not suitable for other TKI therapies [31].
According to long-term follow-up results of Ponatinib treatment for CML patients, the 10-year OS rate is 90%, and the 2-year event-free survival (EFS) rate is 97%. After 6 months of treatment, 96% of patients achieved complete cytogenetic response, 80% achieved MMR, 61% achieved MR4 (leukemia cells decreased by 4 logarithms, residual count less than 0.01%), and 46% achieved MR4.5. At the 12-month mark, 94% of patients achieved complete cytogenetic response, 81% achieved MMR, and the MR4 rate sustained for over 5 years was 51%. More importantly, no disease progression to accelerated phase or blast crisis occurred during the treatment period. However, 28% of patients discontinued medication due to cardiovascular toxicity [32].
The main adverse reactions of Ponatinib are arterial occlusive events, hypertension, gastrointestinal reactions, etc. Due to the increased risk of arterial occlusive events and other serious toxicities potentially induced by Ponatinib, it is currently not suitable as a first-line treatment option [32,33].
In general, the application of TKIs has brought significant improvements in treatment outcomes for CML patients. For example, the development of Imatinib has reduced the annual mortality rate of CML from 10–20% to 1–2% [34]. However, the resistance issues and other adverse reactions caused by long-term use of TKIs prompt us to continuously explore new targets.

2.2. FLT3 Inhibitors

In recent years, studies have found that mutations in the FLT3 gene are relatively common in AML, accounting for about 30% of AML cases [35]. FLT3 mutations lead to abnormal activation of the FLT3 receptor, which continuously activates downstream signaling pathways such as Ras/MAPK/, STAT5, PI3K/Akt/mTOR, inhibits cell apoptosis, and promotes the proliferation and survival of leukemia cells [36]. FLT3 inhibitors are a class of targeted therapy drugs for FLT3-mutated AML that have been approved for clinical targeted therapy. According to the mechanism of action and targets, FLT3 inhibitors can be divided into first-generation and second-generation FLT3 inhibitors. These drugs inhibit the abnormal activation of the FLT3 receptor, prevent the proliferation and survival of leukemia cells, and significantly improve the therapeutic effect and survival rate of AML patients.

2.2.1. Midostaurin

Midostaurin (Figure 1) is an oral multi-kinase small-molecule inhibitor and the first-generation FLT3 inhibitor. It mainly inhibits the tyrosine kinase activity of the FLT3 receptor, blocks its downstream signaling pathways, thereby inhibiting the proliferation and survival of leukemia cells and inducing apoptosis (Figure 2). It was approved by the FDA in 2017 for the treatment of AML and is also the first approved FLT3 inhibitor in the United States and Europe [37].
Although Midostaurin has been approved for adult FLT3-mutated AML, its effect is suboptimal for relapsed or refractory pediatric acute leukemia patients. A phase 1/2 clinical trial showed that compared to ALL patients with KMT2A rearrangement, FLT3-mutated AML patients showed more significant advantages in overall response rate (55.5% vs. 23.1%) and overall survival (3.7 months vs. 1.4 months) [38]. In addition, long-term use of Midostaurin is associated with adverse reactions such as low blood cell counts (neutropenia, anemia, thrombocytopenia), severe allergic reactions, and liver function problems [39].

2.2.2. Gilteritinib

Gilteritinib (Figure 1), as a second-generation FLT3 inhibitor, is an oral small-molecule drug. It has important therapeutic significance for AML patients carrying FLT3 internal tandem duplication mutations (FLT3-ITD) and has been approved for the treatment of relapsed/refractory FLT3-mutated acute myeloid leukemia [40]. Compared with first-generation FLT3 inhibitor therapy, Gilteritinib can effectively inhibit multiple secondary resistance mutations, thereby overcoming resistance issues and more effectively improving the survival rate and response rate of AML patients. The 2-year follow-up results of the ADMIRAL trial showed that among 247 patients in the Gilteritinib group, 26 patients survived for 2 years or more without relapse. Among them, 18 patients received HSCT, and 16 patients continued to use this drug for maintenance therapy after HSCT. During the maintenance treatment phase, Gilteritinib also showed good efficacy, enabling sustained remission status. This indicates that Gilteritinib has good efficacy and safety for FLT3-mutated AML, providing a better choice for the subsequent treatment of AML patients, especially when formulating long-term treatment plans [41].
The main side effect of Gilteritinib monotherapy is the development of acquired drug resistance. This resistance is closely related to the reactivation of the PI3K/AKT and RAS/MEK pathways. The activation of these pathways promotes the survival and proliferation of tumor cells, thereby increasing drug resistance [42]. This indicates that the management of drug resistance needs to be considered in subsequent treatment.

2.3. B-Cell Signaling Pathway Inhibitors

B-cell signaling pathway inhibitors are a class of targeted therapy drugs targeting key molecules or kinases in the B-cell signaling pathway. These drugs inhibit key components of the B-cell signaling pathway, prevent the proliferation and survival of leukemia cells, and significantly improve the therapeutic effect and survival rate of CLL and certain types of ALL patients. The main B-cell signaling pathway inhibitors include Bruton’s tyrosine kinase (BTK) inhibitors and PI3Kδ inhibitors, etc. [43].

2.3.1. BTK Kinase Inhibitors

BTK is a protein that plays a key role in malignant tumor signal transduction. BTK participates in the differentiation and proliferation of B cells by binding to the B-cell receptor (BCR) [44]. This characteristic makes BTK a hot drug target for treating B-cell-related diseases. BTK inhibitors inhibit its tyrosine kinase activity by binding to the active site of BTK. This inhibition prevents the phosphorylation of downstream signaling molecules by BTK, thereby blocking the BCR signaling pathway transmission [45]. In recent years, BTK inhibitors (such as Ibrutinib, Acalabrutinib) have achieved significant success in the treatment of CLL, greatly improving the prognosis of patients.
Ibrutinib
Ibrutinib (Figure 1) is the first BTK inhibitor approved for clinical use. It was approved by the FDA in 2016 for the initial treatment of CLL and is currently approved for patients with untreated, relapsed, or refractory disease, including patients with del(17p). PLCγ2 is a key downstream target of BTK. BTK inhibitors reduce their activity by preventing the phosphorylation of PLCγ2, thereby inhibiting the release of intracellular calcium ions and the hydrolysis of cell membrane phospholipids (Figure 3).
Follow-up results of an international multicenter phase III clinical trial showed that among elderly CLL patients, especially those with TP53 mutation, 11q deletion, and IGHV mutation, compared with traditional chlorambucil treatment plan, oral Ibrutinib treatment plan can achieve an overall response rate (ORR) of up to 92%, and its progression-free survival (PFS) (70% vs. 12%) and OS rate (83% vs. 68%) were significantly higher than the chlorambucil group [46].
Although Ibrutinib shows significant survival advantages in long-term studies, its side effects, such as atrial fibrillation and hypertension, lead to a higher treatment discontinuation rate. Research shows that among the selected 43 CLL patients, the cumulative incidence of atrial fibrillation was 30.0%, and the 5-year expected atrial fibrillation rate reached 31.5% [47]. Therefore, rational selection of targeted drugs and conducting combination therapy are crucial during the treatment process.
Acalabrutinib
Acalabrutinib (Figure 1) is a second-generation BTK inhibitor developed by AstraZeneca, characterized by high selectivity and potent effects. It has a shorter half-life and can be safely administered twice daily. This dosing method results in a longer remission period [48]. Compared with Ibrutinib, Acalabrutinib has similar efficacy in treating CLL, but relatively fewer toxic side effects [49]. In a phase I/II clinical study targeting relapsed/refractory (R/R)-CLL patients, Acalabrutinib showed good safety and efficacy, with an ORR of 94%. At 45 months, the estimated PFS value was 62% [50]. A phase 3 study further confirmed that compared with traditional treatment plans (Idelalisib plus Rituximab or Bendamustine plus Rituximab), Acalabrutinib monotherapy significantly improved the PFS of R/R-CLL patients and had acceptable safety. This drug was approved by the FDA in 2019 for the treatment of CLL [43].
The side effects of Acalabrutinib treatment for leukemia patients are similar to Ibrutinib, also prone to causing cardiovascular complications and bleeding adverse reactions. Research found that during Acalabrutinib treatment for CLL/Small Lymphocytic Lymphoma, the incidence of grade 3 and above adverse events reached 66%, including neutropenia (14%), pneumonia (11%), hypertension (7%), anemia (7%), and diarrhea (5%). The incidence rates of atrial fibrillation and major bleeding events were 7% and 5%, respectively. 56% of patients are still receiving treatment, with the main reasons for discontinuation being disease progression (21%) and adverse events (11%) [50]. Therefore, it is necessary to develop new targeted drugs or new treatment methods to reduce their toxicity.

2.3.2. PI3Kδ Inhibitors

PI3Kδ inhibitors are a class of drugs that selectively target the PI3Kδ subtype. PI3Kδ is a member of the phosphatidylinositol 3-kinase (PI3K) family, mainly expressed in white blood cells, and is closely related to tumor development, immune regulation, and angiogenesis. PI3Kδ inhibitors inhibit tumor cell proliferation and survival by blocking PI3Kδ signaling, while also regulating the immune system [51].
Idelalisib (Figure 1) is a highly efficient and selective oral small-molecule PI3Kδ inhibitor. Its mechanism of action is to inhibit the PI3Kδ-dependent signaling pathway, reduce the activity of the AKT and mammalian target of rapamycin pathways, thereby inhibiting the activity of tumor cells (Figure 3) [45]. A phase I trial found that Idelalisib showed good safety and significant clinical efficacy in R/R-CLL patients. 81.5% of patients achieved lymph node response (50% reduction in the sum of the products of the perpendicular diameters (of measured lymph nodes) of the measured nodal lesions), the ORR reached 72%, and no dose-limiting toxicity was observed within the dose range. This drug received its first approval in the United States in July 2014 for the treatment of relapsed CLL [51].
Although Idelalisib has a good therapeutic effect on CLL, it can still trigger some adverse reactions during treatment, such as sepsis, pneumonia, diarrhea, and colitis, which limit its clinical application [52].

2.4. Anti-Apoptotic Inhibitors

Members of the anti-apoptotic B-cell lymphoma-2 (BCL-2) family are core regulatory factors in the apoptosis process. They are divided into three categories: anti-apoptotic proteins (such as BCL-2, BCL-XL, MCL-1), pro-apoptotic proteins (such as BAK, BAX), and pro-apoptotic proteins containing only the BH3 domain (such as BIM, BAD, NOXA). Among them, anti-apoptotic proteins inhibit apoptosis by binding to pro-apoptotic proteins and inhibiting their activity. BCL-2 inhibitors restore the natural pathway of apoptosis by mimicking the BH3 domain and binding to anti-apoptotic proteins (such as BCL-2), preventing them from interacting with other pro-apoptotic proteins [53].
Venetoclax (Figure 1), as the first selective BCL2 inhibitor, is also the first approved new anti-cancer drug (BH3 mimetic) for routine clinical treatment. Its mechanism of action is to directly target the BCL2 protein, reduce its inhibition of BAX and BAK proteins, thereby inducing CLL cell apoptosis (Figure 3) [54,55,56]. It is currently used for CLL and AML. However, long-term use may affect normal blood cells, leading to some adverse reactions, such as persistent cytopenias, clonal hematopoiesis, treatment-related myeloid neoplasms, and mutations in the BAX gene, thereby increasing resistance to Venetoclax treatment [53,57].

2.5. Immunotherapy Drugs

Immunotherapy drugs are a class of treatment methods that identify and attack leukemia cells by activating or enhancing the patient’s own immune system. According to the mechanism of action and targets, immunotherapy drugs can be divided into bispecific T-cell engagers (BiTEs) and monoclonal antibodies, etc. These drugs show significant efficacy and lower side effects in clinical practice, especially in treating relapsed or refractory leukemia patients.

2.5.1. Monoclonal Antibodies

Monoclonal antibodies are laboratory-produced immune system proteins. Their application in leukemia is an important advancement in the field of leukemia treatment. Monoclonal antibodies can specifically bind to targets highly expressed on leukemia cells but sparsely expressed on normal cells. They exert effects through multiple mechanisms, including antibody-dependent cell-mediated cytotoxicity (ADCC), complement-dependent cytotoxicity (CDC), and directly inducing apoptosis [58]. Monoclonal antibodies can be used alone or in combination with chemotherapy drugs. Currently, multiple monoclonal antibodies have been applied in the treatment of leukemia patients.
Rituximab
Rituximab (Figure 1) is an anti-CD20 immunotherapy drug, the first monoclonal antibody approved for cancer treatment, and received FDA approval in 1997 [59,60]. Research has found that Rituximab exerts effects on ALL, CLL, hairy cell leukemia (HCL), etc., through various mechanisms (Figure 4) [61,62,63]. Specific mechanisms include: inducing NK cell release of granzymes by binding to the Fc-γ receptor on NK cells, thereby leading to cell death [64,65]; binding to the CD20 molecule on the surface of B cells, activating the extrinsic apoptosis pathway to induce cell death [66]; binding to complement components (especially C1q), activating the complement system, forming the membrane attack complex, leading to cell death [67]; binding to the Fcγ receptor on macrophages, inducing immune cell phagocytosis and clearing B cells [68]. According to long-term follow-up results of Rituximab treatment for B-ALL, compared with the control group, the proportion of patients with minimal residual disease (MRD) level below 10−4 (i.e., less than 1 bone marrow blast in 10,000 normal cells) was significantly increased (91% vs. 82%); the 2-year EFS rate after a median follow-up of 30 months (65% vs. 52%), the cumulative relapse rate (18% vs. 32%), and the 4-year cumulative relapse rate (25% vs. 41%) all showed significant advantages; after induction therapy, the complete remission (CR) rate (92% vs. 90%) showed no significant difference between the two groups. In addition, the overall incidence of serious adverse events was similar between the two groups, but fewer patients in the Rituximab group experienced allergic reactions to asparaginase [61]. Overall, the results of this study indicate that Rituximab can be a new treatment option for B-ALL.
Rituximab can be used as monotherapy or in combination with other drugs for the treatment of HCL. However, when used as monotherapy, its efficacy for relapsed or refractory HCL patients is limited, rarely inducing complete remission [69]. In addition, it can cause infusion-related reactions and rarer toxicities such as cytopenia and infection [70]. Therefore, more effective drugs need to be developed to overcome these adverse reactions and improve therapeutic effects.
Obinutuzumab
Obinutuzumab (Figure 1) is a type II fully humanized anti-CD20 antibody. Compared with Rituximab, it has stronger antibody-dependent cell-mediated cytotoxicity and direct cell death effects, but weaker complement activation effects (Figure 4) [71]. It is currently approved for CLL. According to the GAUGUIN study, Obinutuzumab monotherapy is safe and effective in R/R-CLL patients, especially in patients with low baseline tumor burden. The Phase I trial showed a good dose-response relationship, with an overall response rate and best overall response rate both being 62%. In the Phase II trial, the overall response rate was 15%, and the best overall response rate was 30%. The lower response rate may be related to the higher baseline tumor burden of the patients [72].
Compared with Rituximab, although Obinutuzumab shows better efficacy in CLL, the incidence of grade 3–4 adverse events is significantly increased, especially in terms of infusion-related reactions, thrombocytopenia, and cardiac events. In addition, the rates of grade 3–4 infection and drug discontinuation also tend to increase [70].

2.5.2. Bispecific T-Cell Engager

Bispecific T-cell Engager (BiTE) is a bispecific antibody capable of simultaneously binding to tumor-associated antigens and the CD3 molecule on the surface of T cells [73]. BiTEs consist of two single-chain variable fragments (scFv) connected by a flexible linker peptide. One scFv specifically binds to tumor-associated antigens on the surface of tumor cells, such as CD19, BCMA, etc.; the other scFv binds to the CD3 molecule on the surface of T cells [74]. As a new class of immunotherapy drugs, it has shown significant application value in the treatment of relapsed/refractory R/R-ALL.
CD19/CD3 bispecific antibody is a novel immunotherapy drug mainly used for treating B-cell related hematological malignancies, such as ALL. This antibody can simultaneously specifically bind to the CD19 antigen on the surface of B cells and the CD3 antigen on the surface of T cells, mediating the killing effect of T cells on CD19-expressing malignant B cells. Currently, this type of antibody has shown good therapeutic effects and safety in clinical trials. As an FDA-approved CD19/CD3 bispecific antibody, it is currently used for the treatment of B-ALL [75,76].
Blinatumomab (Figure 1) is a novel BiTE that can simultaneously bind to CD3 on T cells and CD19 on the surface of B-ALL cells, thereby activating T cells to kill tumor cells (Figure 5) [76]. It was approved by the FDA in 2014 and approved by China’s NMPA in 2020. It is the first approved standard BiTE molecule and the only approved drug for measuring MRD in ALL treatment. It is mainly used for treating adult relapsed/refractory precursor B-ALL [77]. Blinatumomab can be used as monotherapy or in combination with chemotherapy drugs for the treatment of B-ALL. Results of a phase 3 clinical trial showed that compared with standard chemotherapy, Blinatumomab treatment for adult relapsed/refractory precursor B-ALL has significant advantages. Comparative analysis revealed that the Blinatumomab group significantly prolonged OS and DOR, with a median OS of 7.7 months (chemotherapy group 4.0 months), reducing the risk of death by 29%. In addition, the CR rate (including complete remission with full hematologic recovery) (34% vs. 16%) and CRi rate (complete remission with incomplete or partial hematologic recovery) (44% vs. 25%) in the Blinatumomab group, as well as the 6-month EFS (31% vs. 12%), were significantly superior to the chemotherapy group. Although the Blinatumomab group reported higher rates of neurological events and cytokine release syndrome, the overall incidence of adverse events was lower. This study provides new hope for the treatment of adult relapsed or refractory B-cell precursor ALL, especially in cases where traditional chemotherapy effects are limited [76].

2.6. Differentiation Inducers

Differentiation inducers are a special type of drug that inhibits the unlimited proliferation of abnormal or immature cells and may eventually lead to their death by promoting them to regain normal maturation and differentiation ability. Among them, all-trans retinoic acid (ATRA) is a representative differentiation inducer. Its application in the treatment of acute promyelocytic leukemia (APL) has effectively changed the treatment strategy of the disease, transforming this once extremely lethal cancer into a highly curable disease [78].
As a retinoic acid differentiation inducer, ATRA is one of the earliest drugs used in tumor differentiation induction therapy and is now widely recognized as the first choice for APL induction therapy. Its mechanism of action is mainly through binding to the PML-RARα fusion protein unique to APL, relieving its inhibitory effect on differentiation-related genes, thereby inducing abnormal promyelocytes to differentiate into mature neutrophils [79]. However, long-term use may cause some adverse reactions, such as increasing the risk of differentiation syndrome, inducing excessive inflammation, a higher risk of relapse, and possible hyperleukocytosis [80].

2.7. Upcoming Targeted Therapies Under Trial

Currently, many new targeted therapeutic drugs are in the clinical trial stage, and some of them have shown good efficacy. For example, Ziftomenib, an oral selective menin inhibitor developed for patients with relapsed or refractory AML with NPM1 mutations and KMT2A rearrangements. Its mechanism of action is mainly to disrupt the binding of Menin to the binding pocket in MLL1/2 and MLL1-FP, thereby reducing the binding of MLL1/2 and MLL1-FP to their targets, inhibiting HOXA9/MEIS1 activity, and inducing differentiation and loss of viability of AML with MLL1-r or mutant (mt)-NPM1 [81]. According to the results of the KOMET-001 trial, 24% of patients achieved complete remission or complete remission with partial hematological recovery, and most adverse events are currently manageable [82]. These results show that Ziftomenib, as a new oral selective menin inhibitor, has potential clinical application value in the treatment of relapsed or refractory AML.
Nemtabrutinib, an oral reversible BTK inhibitor, can effectively inhibit C481S mutant BTK and activated PLCγ2, overcoming the drug resistance after ibrutinib treatment [83]. According to the results of a phase I clinical trial, the overall remission rate for CLL patients receiving 65 mg daily treatment reached 75% [84]. This shows that Nemtabrutinib has significant potential in the treatment of CLL.

2.8. Limitations of Targeted Therapy

In recent years, the field of targeted therapy has developed rapidly, especially in the research and development of new drugs targeting specific molecular targets and the optimization of combination therapy strategies. Taking CML as an example, TKIs such as Imatinib (first-generation) and Dasatinib (second-generation) have significantly improved patient survival rates and offer the possibility of achieving “treatment-free remission” [16,22]. Although targeted therapy effects are significant, monotherapy use has limitations. First, resistance is a major problem. In CML treatment, as time progresses, some patients develop resistance to TKIs, leading to disease progression. Research shows that about 25% of patients develop resistance after Imatinib treatment [85]. In AML, resistance often occurs after FLT3 inhibitor treatment, mainly due to secondary mutations in the FLT3 kinase domain, which prevent the drug from effectively binding to the target, reducing treatment efficacy [42].
Secondly, monotherapy is difficult to completely eradicate leukemia cells. Leukemia cells exhibit high heterogeneity, with different cell subpopulations possibly relying on multiple signaling pathways for survival. A single targeted drug may only inhibit the growth of some cells and cannot completely eliminate all leukemia cells. In addition, monotherapy may not overcome the protective effect of the tumor microenvironment on leukemia cells. Cytokines, stromal cells, etc., in the tumor microenvironment can promote the survival and proliferation of leukemia cells, reducing the efficacy of monotherapy. Moreover, monotherapy may cause some adverse reactions, affecting the patient’s quality of life and treatment compliance, limiting the dosage and duration of drug use, thus affecting treatment efficacy. To address these issues, combination therapy has become a new research direction. Combination therapy can improve treatment efficacy, overcome resistance, and reduce adverse reactions through the synergistic effects of different drugs.

3. Combination Therapy

In the treatment of leukemia, the overall efficacy of monotherapy with targeted drugs is not ideal. To effectively address key issues such as drug resistance and relapse, combination therapy strategies have gradually received attention. For example, the combined application of Venetoclax and hypomethylating agents has a significantly higher overall complete remission rate than the use of Azacitidine alone [86]. Therefore, combination therapy has become a hot research direction in the field of leukemia treatment. The combination application modes of targeted drugs are diverse, including the combination of targeted drugs and chemotherapy drugs, sequential therapy between targeted drugs, synergistic application of targeted drugs and CAR-T cell therapy, and multi-drug combination schemes, etc.

3.1. Combination of Targeted Drugs and Chemotherapy Drugs

Given the certain limitations of monotherapy targeted therapy in leukemia treatment, many studies in recent years have focused on the combined application of targeted drugs and chemotherapy drugs in leukemia treatment to enhance its therapeutic effect (Table 1).
Taking Ph+ALL and AML as examples, studies have found that the combination therapy of targeted drugs and chemotherapy drugs can significantly improve the survival rate of leukemia patients and effectively improve their prognosis.
For Ph+ALL patients, relevant studies have shown that when using Imatinib combined with a low-intensity CVAD treatment plan, the patient’s 5-year OS rate reached 45.6%, and EFS was 37.1% [14]. Compared with traditional chemotherapy plans, adding Imatinib significantly improved the patient’s complete remission rate and long-term survival rate. In addition, the latest results of the Ph+ ALL CON trial show that Ponatinib combined with low-intensity chemotherapy for newly diagnosed Ph+ALL adult patients is superior to Imatinib combined with low-intensity chemotherapy. At the end of induction therapy, the MRD-negative complete remission rate in the Ponatinib group was significantly higher than in the Imatinib group (34.4% vs. 16.7%), and the safety of the two treatment plans was comparable [87].
Furthermore, the combination of targeted drugs and the Hyper-CVAD regimen has better therapeutic effects for high-risk Ph+ALL patients, but attention should be paid to drug dosage. For example, using Ponatinib combined with the Hyper-CVAD regimen to treat Ph+ALL patients, the CR reached 100%, the complete molecular remission rate was 86%, and the 6-year OS rate was 75%. In addition, most patients did not need HSCT after achieving complete remission for the first time, and after dose adjustment, the safety of this treatment strategy was significantly improved [88]. Results from another phase II clinical study showed that the Hyper-CVAD combined with a sequential Blinatumomab treatment plan has significant efficacy for Ph-negative B-cell ALL adult patients. This treatment plan significantly improved the patient’s 3-year OS rate (81%) and relapse-free survival rate (73%), and during the treatment process, the MRD negativity rate was as high as 97% [75].
For AML patients, the combined application of targeted drugs and chemotherapy drugs has also achieved significant results. Whether in AML patients of all age groups or in AML patients carrying FLT3 mutations, this combination therapy method has shown good efficacy. For example, for elderly AML patients who are untreated and unsuitable for intensive chemotherapy, compared with Azacitidine alone, the combined use of Venetoclax and Azacitidine can significantly prolong the patient’s overall survival and improve the overall complete remission rate. Although the incidence of febrile neutropenia is relatively high in the combination therapy group, overall, the safety and effectiveness of this combination therapy are superior to Azacitidine alone [86]. Currently, this combination therapy plan has been identified as the standard treatment method for elderly or medically unfit AML patients who cannot tolerate intensive chemotherapy. There are also studies combining Venetoclax with FLAG-IDA for the treatment of newly diagnosed AML or R/R-AML patients. The ORR of this combination plan is 98%, and the MRD-negative complete remission rate is 93%. The 24-month OS rate is 76%, and EFS is 64%, and the adverse events are similar to traditional chemotherapy and manageable. In addition, this study also emphasized the key role of TP53 mutation in predicting treatment outcome and survival rate, providing an important direction for future treatment of this high-risk subgroup [89].
In addition, the RATIFY study results showed that Midostaurin combined with chemotherapy drugs has significant efficacy for AML patients carrying FLT3 mutations. Compared with the placebo group, the Midostaurin group significantly prolonged OS (74.7 months vs. 25.6 months) and EFS (8.2 months vs. 3.0 months), but there was no significant difference in CR (58.9% vs. 53.5%), and the incidence of adverse events was similar to the placebo group, indicating that Midostaurin combined chemotherapy plan can significantly improve the survival prognosis of AML patients carrying FLT3 mutations [90]. This conclusion was further validated in a phase IIIb clinical trial. Midostaurin combined with chemotherapy has safe and high response rate treatment options for adult FLT3-mutated AML patients of all age groups. This drug has now been approved for use in combination with standard induction and consolidation chemotherapy for the treatment of newly diagnosed FLT3 AML adult patients [91].
In summary, combining traditional chemotherapy drugs with targeted drugs effectively improves the OS and CR rates of leukemia patients. Currently, some combination therapy plans have been used as standard treatment plans for leukemia. However, it also faces problems such as drug interactions and complex treatment plans, which need continuous optimization and adjustment in clinical practice.

3.2. Internal Combination of Targeted Drugs

3.2.1. Internal Combination of Inhibitors

Based on the significant monotherapy activity of inhibitors in leukemia treatment, and the significant improvement in treatment effects when they are used in internal combination, researching new combination strategies (Table 2) to inhibit leukemia cells through synergistic effects, thereby improving efficacy, holds important significance.
Taking CML as an example, numerous studies have explored the combined application of targeted drug inhibitors. For instance, a phase 2 clinical trial showed that alternating Nilotinib and Imatinib treatment might help improve efficacy, enhance safety, and reduce costs. Study results indicated that the 5-year PFS and OS rates for this regimen were both 89%, and its safety, especially regarding cardiovascular adverse events, was superior to Nilotinib monotherapy [99]. There are also studies combining Dasatinib with Venetoclax for the treatment of newly diagnosed chronic myeloid leukemia in chronic phase (CML-CP) patients. Research found that the combination therapy of Dasatinib and Venetoclax is safe, feasible, and effective. At 12 months of treatment, MMR was 86%, and DMR was 53%. However, compared to Dasatinib monotherapy, the combination therapy did not significantly improve MMR and DMR rates, but instead led to a higher incidence of neutropenia [100].
Furthermore, several targeted drug inhibitor combination regimens currently in the research phase have shown good effects on CML resistance and synergy. For example, Mariarita Spampinato et al. found that combining Dasatinib with Selinexor to treat human CML cell lines (LAMA84 and K562) exhibited significant inhibitory effects. Its mechanism may involve impairing mitochondrial function and downregulating the nuclear translocation of HO-1, increasing the sensitivity of leukemia stem cells (LSCs) to apoptosis, reducing their ability to maintain the disease state, and thus potentially overcoming drug resistance [101]. Additionally, studies have shown that the combination of Imatinib and Carfilzomib exhibits synergistic effects in the treatment of CML [102].
In summary, the combined application of targeted drug inhibitors shows better efficacy in leukemia treatment, especially in overcoming drug resistance and improving treatment efficiency. However, some combination regimens perform poorly in terms of safety. Therefore, further exploration of new combination therapy strategies and optimization of drug combinations are needed to provide patients with more effective treatment options.

3.2.2. Combination of Inhibitors and Immunotherapy Drugs

By combining inhibitors with immunotherapy drugs, multiple targets of leukemia cells can be targeted simultaneously, thereby enhancing therapeutic effects, reducing drug resistance, and decreasing the occurrence of side effects. Currently, various combination regimens of inhibitors and immunotherapy drugs have been developed (Table 3).
In CLL treatment, a long-term follow-up study showed that the PFS and OS rates of CLL patients treated with Obinutuzumab combined with Chlorambucil (O+Chl) were significantly better than those treated with Rituximab combined with Chlorambucil (R+Chl). Especially in terms of OS, the O+Chl group showed a clear survival advantage [103].
However, the CLL14 study found that a fixed-duration regimen of Venetoclax plus Obinutuzumab showed significantly better long-term efficacy and safety in treatment-naive CLL patients compared to Obinutuzumab plus Chlorambucil. Results showed that the 6-year OS rate of the V+O group was significantly higher than the O+Chl group (78.7% vs. 69.2%), and the V+O group had significantly longer PFS and time to next treatment, with significantly improved quality of life [104].
Similarly, another study, ELEVATE-TN, showed that Acalabrutinib combined with Obinutuzumab (A+O) demonstrated better safety and tolerability compared to the O+Chl regimen. The results of this study showed that after a median follow-up of 74.5 months, the median PFS of the A+O combination therapy was not reached, while the median PFS of O+Chl was 27.8 months. Furthermore, the 72-month survival rate of the A+O group was 83.9%, significantly better than the A group and the O+Chl group (75.5% and 74.7%) [105]. This indicates significant individual differences in patient treatment.
Additionally, studies have used Rituximab combined with Ibrutinib (IR) for CLL treatment. According to the interim analysis of the FLAIR trial, compared with the Fludarabine, Cyclophosphamide, Rituximab (FCR) combination regimen, after a median follow-up of 53 months, the median PFS of the IR group was not reached, significantly superior to the median PFS of 67 months in the FCR group. However, the 4-year OS rates (92.1% vs. 93.5%) showed no significant difference, possibly because patients received effective second-line targeted therapy after disease progression. But the IR group treatment increased the risk of cardiovascular events, especially in patients already receiving treatment for hypertension or cardiovascular disease. Therefore, patients can choose the corresponding treatment plan based on their condition. If using the IR group treatment, a comprehensive cardiac evaluation should be performed first [62].
Overall, the combined application of inhibitors and immunotherapy drugs shows significant efficacy in the treatment of leukemia, especially in improving PFS and OS rates. However, the safety and tolerability of different combination regimens vary, and individual patient differences significantly impact treatment outcomes. Therefore, during the treatment process, the most suitable treatment plan should be selected based on the specific situation of the patient, and a comprehensive evaluation should be conducted before treatment to ensure the safety and effectiveness of the treatment. At the same time, further development of new combination therapy strategies is needed to meet the needs of different patients.
Table 3. Internal Combination of Targeted Drugs.
Table 3. Internal Combination of Targeted Drugs.
Combination
Therapy Type
Combined DrugsIndicationPatient NumberTreatment OutcomeReference
Inhibitor Internal CombinationImatinib, NilotinibCML1235-year OS and PFS rates both 89%[99]
Dasatinib, VenetoclaxCML654-year EFS and OS rates 96% and 100% respectively[100]
Dasatinib, SelinexorCMLNoneProduces significant inhibitory effects on LAMA84 and K562 cell lines[101]
Imatinib, CarfilzomibCMLNoneCan significantly reduce proliferation and induce CML stem cell apoptosis[102]
Ponatinib, AsciminibCMLNoneEffectively overcomes resistance caused by BCR-ABL1 compound mutations[106]
Imatinib, VenetoclaxCLL, CML1Achieved DMR[107]
Acalabrutinib, VenetoclaxCLL86736-month OS rate 94.1%[108]
Idelalisib, TirabrutinibCLL53Objective response rate 93%, CR 7%[109]
Gilteritinib, MEN1703AMLNoneSimultaneously inhibits PIM and FLT3, significantly enhances anti-tumor activity in vivo/in vitro[110]
Gilteritinib, GSK-J4AMLNoneCombination therapy of Gilteritinib and GSK-J4 can significantly inhibit the growth of MV4-11 and MOLM-13 cells[111]
Inhibitors in combination with immunologic drugsAcalabrutinib,
Obinutuzumab
CLL53572-month OS rate 83.9%, PFS rate 78%[105]
Obinutuzumab, chlorambucilCLL358-year OS rate 61%, EFS rate 25%[103]
Obinutuzumab, VenetoclaxCLL4326-year OS rate 78.7%, for patients with del(17p) and/or TP53 mutation, 6-year OS rate 60.0%[104]
Ibrutinib, UblituximabCLL126Median follow-up 41.6 months, ORR 83%[112]
Rituximab, IdelalisibCLL110ORR 83.6%[45]
Rituximab, IbrutinibCLL19244-year OS rate 92.1%[62]
Rituximab, VenetoclaxCLL3892-year OS rate 91.9%[113]
Dasatinib, BlinatumomabPh+ ALL63Median follow-up 18 months, OS rate 95%, Disease-Free Survival 88%[114]
Rituximab, VemurafenibHCL30Median follow-up 34 months, Relapse-Free Survival rate 85%[63]

3.3. Combination of Targeted Drugs and CAR-T

CAR-T is a revolutionary cancer treatment method that modifies a patient’s own T cells to enable them to more effectively recognize and attack cancer cells in the body. The specific process involves extracting T cells from the patient’s body, genetically modifying these cells to produce chimeric antigen receptors on their surface that target specific cancer cell antigens. These modified T cells are called CAR-T cells, which are then expanded and reinfused into the patient’s body with the aim of eliminating cancer cells [115,116]. Currently, anti-CD19 CAR-T therapy targeting malignant B cells in leukemia has received FDA and EMA approval and has shown good efficacy in clinical applications [117].
Although CAR-T cell therapy has achieved significant success in B-ALL, treatment failure due to disease relapse or primary resistance, as well as serious side effects such as cytokine release syndrome and immune effector cell-associated neurotoxicity syndrome, still limit the widespread application of CAR-T cell therapy [118]. Therefore, it is necessary to reduce its toxic side effects and increase treatment efficacy through combination therapy. A prospective single-center phase II clinical trial showed that combining Ibrutinib and CAR-T cell therapy can achieve higher CR and longer PFS in CLL patients who have not achieved complete remission. At 3 months, the complete remission rate was 43.8%, at 12 months, it was 50%, and at 12 months, 72.2% of patients had undetectable MRD. At 48 months, the overall survival rate was 84%, and the progression-free survival rate was 70%. This indicates that combination therapy has high safety and effectiveness in CLL and can significantly improve patient prognosis [119].
Furthermore, the latest research shows that Dasatinib combined with CAR-T cells for the treatment of newly diagnosed Ph+AML adult patients shows a complete molecular remission rate reaching 85%, and the 2-year OS rate and leukemia-free survival rate are both 92%, with good safety and efficacy [120].
Multiple studies (Table 4) have demonstrated that the combination of targeted drugs and CAR-T has good efficacy in the treatment of leukemia. Although there are still certain limitations, new CAR-T combination methods can be continuously developed subsequently to effectively improve the cure rate of leukemia.

3.4. Multi-Drug Combination

Although combination therapy for leukemia shows significant advantages in improving efficacy, problems still exist in terms of safety and tolerability. By using multiple drugs with different mechanisms of action in combination, leukemia cells can be attacked from multiple angles, showing more obvious advantages in overcoming drug resistance, reducing side effects, improving survival rates, and prolonging disease-free survival. Currently, various multi-drug combination therapies for leukemia have been developed and applied (Table 5).
For example, in the treatment of CLL, the iFCG regimen showed high efficacy and low side effects in newly diagnosed CLL patients. Results of a phase 2 clinical trial showed that 87% of patients achieved undetectable MRD (U-MRD) after 3 cycles of chemotherapy, and 98% of patients achieved the best U-MRD response after 12 cycles. The 3-year PFS rate and OS rate were both 98% [123]. This study provides a new, time-limited chemoimmunotherapy (CIT) option for CLL patients, capable of improving U-MRD rates while reducing the burden of chemotherapy, potentially achieving longer disease-free survival and lower long-term side effect risks, especially suitable for young patients in good physical condition.
For relapsed CLL patients, the triple combination therapy of Tirabrutinib, Idelalisib, and Obinutuzumab (TIO group) showed higher efficiency and better safety compared to the combination of Tirabrutinib and Idelalisib (TI group). Results showed that compared to the TI group, at week 25, the ORR of the TIO group was (93.3% vs. 60.0%), and the 24-month PFS was also higher (80.6% vs. 60.0%). In addition, although the CR rate of the TIO group at week 25 was only 6.7%, during subsequent follow-up, the U-MRD negativity rate of the TIO group significantly increased, from 6.7% to 36.7% [124].
Furthermore, the latest research found that the triple-drug combination therapy of Zanubrutinib, Venetoclax, and Obinutuzumab, based on MRD detection, shows higher efficacy in R/R-CLL patients. After 6 cycles of induction therapy, 52.5% of patients achieved peripheral blood U-MRD, and the best U-MRD rate reached 85%. The 18-month OS rate was 96%, and the PFS rate was 96.8%. Although there were more COVID-19-related adverse events, the overall safety was good, and no new unknown resistance mutations appeared [125].
In summary, multi-drug combination therapy for leukemia shows significant advantages in improving treatment efficacy, reducing toxic side effects, prolonging survival, and improving quality of life. By optimizing treatment plans, adverse reactions during the treatment process are significantly reduced, and the patient’s quality of life is significantly improved. However, the long-term survival status is still unsatisfactory, requiring continuous development of new drug combinations and research into new treatment methods.

4. Conclusions and Outlook

Over the past several decades, substantial advancements have been achieved in the pharmacological management of leukemia. Nevertheless, chemotherapy, as the primary treatment modality, continues to exhibit certain limitations. Firstly, traditional chemotherapeutic agents operate by targeting rapidly dividing cells, yet they lack sufficient specificity. This often results in significant toxicity and an increased risk of secondary infections, hemorrhage, organ damage, and secondary tumor development [131,132]. Secondly, chemotherapeutic agents are susceptible to inducing drug resistance during their application, and certain leukemias demonstrate insensitivity to these drugs. Consequently, the efficacy of chemotherapy as a standalone treatment is often suboptimal. Even when CR is attained, residual LSCs may persist within the patient’s body. These LSCs remain in a quiescent state and exhibit insensitivity to chemotherapy, thereby constituting the underlying cause of MRD and subsequent relapse [133].
The advent of targeted therapies has significantly enhanced the prognosis for patients with leukemia, offering renewed optimism for treatment. These therapies function by selectively binding to specific targets, thereby effectively inhibiting the growth and proliferation of leukemia cells while minimizing harm to normal cells. Imatinib, the first kinase inhibitor, exemplifies this advancement by revolutionizing CML treatment, transforming it from a rapidly fatal disease into a manageable condition [134]. However, long-term monotherapy presents several challenges, including drug relapse and resistance. Consequently, the development of novel therapeutic regimens is imperative to improve OS and PFS rates in leukemia, with the ultimate goal of achieving a cure.
Consequently, combination therapies have been formulated to achieve profound and sustained remission by leveraging the synergistic effects of multiple pharmacological agents, thereby extending the duration of treatment-free intervals and markedly enhancing the therapeutic efficacy in leukemia management. For instance, the concurrent administration of Rituximab and Vemurafenib in the treatment of relapsed HCL has been shown to significantly elevate the complete remission rate and reduce the treatment duration. Additionally, this combination effectively improves the MRD clearance rate, thereby prolonging relapse-free survival and facilitating rapid, safe, and durable complete remission [63].
While combination therapy has yielded significant outcomes, it continues to encounter numerous challenges, with relapse being a predominant issue in the current treatment paradigm. For instance, in the treatment of core binding factor acute leukemia (CBFL) using Midostaurin in conjunction with intensive chemotherapy, 97% of patients attained complete remission; however, 51.51% experienced relapse within two years of achieving remission [95]. Therefore, the synergistic application of combination therapy alongside targeted agents requires further refinement to optimize leukemia treatment outcomes.
Furthermore, a substantial challenge in combination therapy lies in the variability among individual patients. Responses to combination therapy can vary significantly, as patients exhibit diverse biological behaviors and drug sensitivities across different types and stages of leukemia. Consequently, when devising a combination therapy regimen, it is imperative to thoroughly account for patient-specific variability. This involves selecting the most appropriate drug combinations, dosages, and treatment sequences through a comprehensive evaluation of each patient. Such an approach aims to enhance the efficacy and safety of combination therapy, thereby advancing the realization of precision medicine [135,136].
In the future, with the help of advanced technologies such as high-throughput sequencing, single-cell sequencing, metabolomics, etc., we will comprehensively analyze the molecular characteristics of different leukemia cells, reveal the abnormal genes, protein expression and their interaction networks, further analyze in depth the pathogenesis of leukemia and the drug resistance mechanism, and search for new therapeutic targets and research and development of new targeted drugs. For example, the development of inhibitors for PI3Kγ, a newly discovered therapeutic target, will provide a new strategy for the combination therapy of leukemia [137].
In conclusion, the continuous development of combination therapy will bring better therapeutic prospects for leukemia patients and is expected to further improve the cure rate of leukemia and the long-term survival rate of patients.

Author Contributions

Writing—original draft: Z.Z., R.Y. and Y.D.; editing: Z.D., L.L. (Lindi Li), Y.L., H.Z. and H.X.; Supervision: C.O. and L.L. (Liang Li); Conceptualization: C.C. and C.O. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by grants from the National Natural Science Foundation of China (No. 82370164 and No. 22407146), GuangDong Basic and Applied Basic Research Foundation (2021A1515111034).

Acknowledgments

We would like to thank Feiqiu Wen and Chi-kong Li for their guidance on this thesis.

Conflicts of Interest

The authors declare that they have no conflicts of interest.

Abbreviations

ALLacute lymphoblastic leukemia;
AMLacute myeloid leukemia;
CLLchronic lymphocytic leukemia;
CMLchronic myeloid leukemia;
TKIsTyrosine kinase inhibitors;
CART-Chimeric Antigen Receptor T-Cell;
Ph+ ALLPhiladelphia positive acute lymphoblastic leukemia;
HSCTHematopoietic Stem Cell Transplantation;
DMRDeep Molecular Response;
TFRTreatment-free remission;
OSOverall Survival;
MMRMajor Molecular Response;
EFSevent-free survival;
FLT3-ITDFLT3 internal tandem duplication mutations;
BTKBruton’s tyrosine kinase;
BCRB-cell receptor;
ORRObjective Response Rate;
PFSprogression-free survival;
R/R-CLLrelapsed/refractory chronic lymphocytic leukemia
BCL-2B-cell lymphoma-2;
BiTEbispecific T-cell engager;
HCLhairy cell leukemia;
MRDminimal residual disease;
CRComplete Remission;
scFvsingle-chain variable fragments;
R/R-ALLrelapsed/refractory acute lymphoblastic leukemia;
CML-CPChronic Phase CML;
LSCsleukemia stem cells;
U-MRDundetectable Minimal Residual Disease
CRiComplete Remission with Incomplete Blood Count Recovery;
ATRAall-trans retinoic acid;
APLacute promyelocytic leukemia.

References

  1. Gale, K.B.; Ford, A.M.; Repp, R.; Borkhardt, A.; Keller, C.; Eden, O.B.; Greaves, M.F. Backtracking leukemia to birth: Identification of clonotypic gene fusion sequences in neonatal blood spots. Proc. Natl. Acad. Sci. USA 1997, 94, 13950–13954. [Google Scholar] [CrossRef]
  2. Metayer, C.; Zhang, L.; Wiemels, J.L.; Bartley, K.; Schiffman, J.; Ma, X.; Aldrich, M.C.; Chang, J.S.; Selvin, S.; Fu, C.H.; et al. Tobacco Smoke Exposure and the Risk of Childhood Acute Lymphoblastic and Myeloid Leukemias by Cytogenetic Subtype. Cancer Epidemiol. Biomark. Prev. 2013, 22, 1600–1611. [Google Scholar] [CrossRef]
  3. Deng, W.; Xu, Y.; Yuan, X. Clinical features and prognosis of acute lymphoblastic leukemia in children with Epstein-Barr virus infection. Transl. Pediatr. 2022, 11, 642–650. [Google Scholar] [CrossRef]
  4. de la Serna, J.; Montesinos, P.; Vellenga, E.; Rayón, C.; Parody, R.; León, A.; Esteve, J.; Bergua, J.M.; Milone, G.; Debén, G.; et al. Causes and prognostic factors of remission induction failure in patients with acute promyelocytic leukemia treated with all-trans retinoic acid and idarubicin. Blood 2008, 111, 3395–3402. [Google Scholar] [CrossRef]
  5. Vesa, S.; Rusu, R.; Sîrbu, D.; Curşeu, D.; Năsui, B.; Sava, M.; Bojan, A.; Lisencu, C.; Popa, M. Chemotherapy-related infectious complications in patients with Hematologic malignancies. J. Res. Med. Sci. 2018, 23, 68. [Google Scholar] [CrossRef]
  6. Tada, A.; Kikuchi, Y.; Murase, K.; Takada, K.; Takasawa, A. Drastic Multiorgan Dysfunction Due to Severe Leukostasis: A Case Report. Cureus 2022, 14, e31518. [Google Scholar] [CrossRef]
  7. Tantiworawit, A.; Power, M.M.; Barnett, M.J.; Hogge, D.E.; Nantel, S.H.; Nevill, T.J.; Shepherd, J.D.; Song, K.W.; Sutherland, H.J.; Toze, C.L.; et al. Long-term follow-up of patients with chronic myeloid leukemia in chronic phase developing sudden blast phase on imatinib therapy. Leuk. Lymphoma 2012, 53, 1321–1326. [Google Scholar] [CrossRef]
  8. Dong, Y.; Shi, O.; Zeng, Q.; Lu, X.; Wang, W.; Li, Y.; Wang, Q. Leukemia incidence trends at the global, regional, and national level between 1990 and 2017. Exp. Hematol. Oncol. 2020, 9, 14. [Google Scholar] [CrossRef]
  9. Bray, F.; Laversanne, M.; Sung, H.; Ferlay, J.; Siegel, R.L.; Soerjomataram, I.; Jemal, A. Global cancer statistics 2022: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J. Clin. 2024, 74, 229–263. [Google Scholar] [CrossRef]
  10. Hao, Y.; Zhang, N.; Wei, N.; Yin, H.; Zhang, Y.; Xu, H.; Zhu, C.; Li, D. Matrine induces apoptosis in acute myeloid leukemia cells by inhibiting the PI3K/Akt/mTOR signaling pathway. Oncol. Lett. 2019, 18, 2891–2896. [Google Scholar] [CrossRef]
  11. Huang, Y.; Qin, Y.; He, Y.; Qiu, D.; Zheng, Y.; Wei, J.; Zhang, L.; Yang, D.; Li, Y. Advances in molecular targeted drugs in combination with CAR-T cell therapy for hematologic malignancies. Drug Resist. Updat. 2024, 74, 101082. [Google Scholar] [CrossRef] [PubMed]
  12. Santos, C.; Pimentel, L.; Canzian, H.; Oliveira, A.; Junior, F.; Dantas, R.; Hoelz, L.; Marinho, D.; Cunha, A.; Bastos, M.; et al. Hybrids of Imatinib with Quinoline: Synthesis, Antimyeloproliferative Activity Evaluation, and Molecular Docking. Pharmaceuticals 2022, 15, 309. [Google Scholar] [CrossRef] [PubMed]
  13. Lee, J.-E.; Thuy, N.T.T.; Lee, J.; Cho, N.; Yoo, H.M. Platyphylloside Isolated from Betula platyphylla is Antiproliferative and Induces Apoptosis in Colon Cancer and Leukemic Cells. Molecules 2021, 24, 2960. [Google Scholar] [CrossRef] [PubMed]
  14. Chalandon, Y.; Thomas, X.; Hayette, S.; Cayuela, J.-M.; Abbal, C.; Huguet, F.; Raffoux, E.; Leguay, T.; Rousselot, P.; Lepretre, S.; et al. Randomized study of reduced-intensity chemotherapy combined with imatinib in adults with Ph-positive acute lymphoblastic leukemia. Blood 2015, 125, 3711–3719. [Google Scholar] [CrossRef]
  15. Hantschel, O.; Rix, U.; Superti-Furga, G. Target spectrum of the BCR-ABL inhibitors imatinib, nilotinib and dasatinib. Leuk. Lymphoma 2008, 49, 615–619. [Google Scholar] [CrossRef]
  16. Maiti, A.; Cortes, J.E.; Patel, K.P.; Masarova, L.; Borthakur, G.; Ravandi, F.; Verstovsek, S.; Ferrajoli, A.; Estrov, Z.; Garcia-Manero, G.; et al. Long-term results of frontline dasatinib in chronic myeloid leukemia. Cancer 2020, 126, 1502–1511. [Google Scholar] [CrossRef]
  17. Gong, H.; Jin, X.; Leng, G.; Zhang, M.; Niu, S.; Cao, W.; Zhang, H.; Zeng, Y.; Li, C.; Li, Y.; et al. Licoflavone A Suppresses Gastric Cancer Growth and Metastasis by Blocking the VEGFR-2 Signaling Pathway. J. Oncol. 2022, 2022, 5497991. [Google Scholar] [CrossRef]
  18. Chang, C.-S.; Yang, Y.-H.; Hsu, C.-N.; Lin, M.-T. Trends in the treatment changes and medication persistence of chronic myeloid leukemia in Taiwan from 1997 to 2007: A longitudinal population database analysis. BMC Health Serv. Res. 2012, 12, 359. [Google Scholar] [CrossRef]
  19. Vysochinskaya, V.; Dovbysh, O.; Gorshkov, A.; Brodskaia, A.; Dubina, M.; Vasin, A.; Zabrodskaya, Y. Advancements and Future Prospects in Molecular Targeted and siRNA Therapies for Chronic Myeloid Leukemia. Biomolecules 2024, 14, 644. [Google Scholar] [CrossRef]
  20. Iqbal, N.; Iqbal, N. Imatinib: A Breakthrough of Targeted Therapy in Cancer. Chemother. Res. Pract. 2014, 2014, 357027. [Google Scholar] [CrossRef]
  21. Mohd Yacob, A.; Muhamad, N.A.; Chang, K.M.; Akmal Hisham, H.; Mat Yusoff, Y.; Ibrahim, L. Hsa-miR-181a-5p, hsa-miR-182-5p, and hsa-miR-26a-5p as potential biomarkers for BCR-ABL1 among adult chronic myeloid leukemia treated with tyrosine kinase inhibitors at the molecular response. BMC Cancer 2022, 22, 332. [Google Scholar] [CrossRef] [PubMed]
  22. Hochhaus, A.; Larson, R.A.; Guilhot, F.; Radich, J.P.; Branford, S.; Hughes, T.P.; Baccarani, M.; Deininger, M.W.; Cervantes, F.; Fujihara, S.; et al. Long-Term Outcomes of Imatinib Treatment for Chronic Myeloid Leukemia. N. Engl. J. Med. 2017, 376, 917–927. [Google Scholar] [CrossRef] [PubMed]
  23. Gambacorti-Passerini, C.; Antolini, L.; Mahon, F.-X.; Guilhot, F.; Deininger, M.; Fava, C.; Nagler, A.; Della Casa, C.M.; Morra, E.; Abruzzese, E.; et al. Multicenter Independent Assessment of Outcomes in Chronic Myeloid Leukemia Patients Treated with Imatinib. JNCI J. Natl. Cancer Inst. 2011, 103, 553–561. [Google Scholar] [CrossRef]
  24. Saußele, S.; Richter, J.; Hochhaus, A.; Mahon, F.-X. The concept of treatment-free remission in chronic myeloid leukemia. Leukemia 2016, 30, 1638–1647. [Google Scholar] [CrossRef]
  25. Hochhaus, A.; Saglio, G.; Hughes, T.P.; Larson, R.A.; Taningco, L.; Deng, W.; Menssen, H.D.; Kantarjian, H.M. Impact of Treatment with Frontline Nilotinib (NIL) vs Imatinib (IM) on Sustained Deep Molecular Response (MR) in Patients (pts) with Newly Diagnosed Chronic Myeloid Leukemia in Chronic Phase (CML-CP). Blood 2015, 126, 2781. [Google Scholar] [CrossRef]
  26. Cheng, F.; Yuan, G.; Li, Q.; Cui, Z.; Li, W. Long-term outcomes of frontline imatinib therapy for chronic myeloid leukemia in China. Front. Oncol. 2018, 13, 1172910. [Google Scholar] [CrossRef] [PubMed]
  27. Ding, L.; Chen, Q.; Chen, K.; Jiang, Y.; Li, G.; Chen, Q.; Bai, D.; Gao, D.; Deng, M.; Zhang, H.; et al. Simvastatin potentiates the cell-killing activity of imatinib in imatinib-resistant chronic myeloid leukemia cells mainly through PI3K/AKT pathway attenuation and Myc downregulation. Eur. J. Pharmacol. 2021, 913, 174633. [Google Scholar] [CrossRef]
  28. Vinay, K.; Yanamandra, U.; Dogra, S.; Handa, S.; Suri, V.; Kumari, S.; Khadwal, A.; Prakash, G.; Lad, D.; Varma, S.; et al. Long-term mucocutaneous adverse effects of imatinib in Indian chronic myeloid leukemia patients. Int. J. Dermatol. 2018, 57, 332–338. [Google Scholar] [CrossRef]
  29. Foà, R.; Vitale, A.; Vignetti, M.; Meloni, G.; Guarini, A.; De Propris, M.S.; Elia, L.; Paoloni, F.; Fazi, P.; Cimino, G.; et al. Dasatinib as first-line treatment for adult patients with Philadelphia chromosome–positive acute lymphoblastic leukemia. Blood 2011, 118, 6521–6528. [Google Scholar] [CrossRef]
  30. Iurlo, A.; Cattaneo, D.; Malato, A.; Accurso, V.; Annunziata, M.; Gozzini, A.; Scortechini, A.R.; Bucelli, C.; Scalzulli, E.; Attolico, I.; et al. Low-dose ponatinib is a good option in chronic myeloid leukemia patients intolerant to previous TKIs. Am. J. Hematol. 2020, 95, E260–E263. [Google Scholar] [CrossRef]
  31. Pulte, E.D.; Chen, H.; Price, L.S.L.; Gudi, R.; Li, H.; Okusanya, O.O.; Ma, L.; Rodriguez, L.; Vallejo, J.; Norsworthy, K.J.; et al. FDA Approval Summary: Revised Indication and Dosing Regimen for Ponatinib Based on the Results of the OPTIC Trial. Oncol. 2022, 27, 149–157. [Google Scholar] [CrossRef]
  32. Haddad, F.G.; Sasaki, K.; Nasr, L.; Short, N.J.; Kadia, T.; Dellasala, S.; Cortes, J.; Nicolini, F.E.; Issa, G.C.; Jabbour, E.; et al. Results of ponatinib as frontline therapy for chronic myeloid leukemia in chronic phase. Cancer 2024, 130, 3344–3352. [Google Scholar] [CrossRef] [PubMed]
  33. Chan, O.; Talati, C.; Isenalumhe, L.; Shams, S.; Nodzon, L.; Fradley, M.; Sweet, K.; Pinilla-Ibarz, J. Side-effects profile and outcomes of ponatinib in the treatment of chronic myeloid leukemia. Blood Adv. 2020, 4, 530–538. [Google Scholar] [CrossRef] [PubMed]
  34. Alves, R.; Gonçalves, A.C.; Rutella, S.; Almeida, A.M.; De Las Rivas, J.; Trougakos, I.P.; Sarmento Ribeiro, A.B. Resistance to Tyrosine Kinase Inhibitors in Chronic Myeloid Leukemia—From Molecular Mechanisms to Clinical Relevance. Cancers 2021, 13, 4820. [Google Scholar] [CrossRef]
  35. Yuan, S.; Zhou, Y.; Li, Y.; Chen, Z.; Xiao, W.; Jiang, D.; Zhang, P.; Zhang, Y.; Bai, F.; Deng, J.; et al. Mitoxantrone-liposome Sensitizes FLT3-ITD Acute Myeloid Leukemia to Gilteritinib Treatment. J. Cancer 2025, 16, 1905–1917. [Google Scholar] [CrossRef] [PubMed]
  36. Rong, Q.-Y.; Lu, Y.; Zhang, W.; Rao, G.-W.; Zheng, Q. Targeting FLT3 for treating diseases: FLT3 inhibitors. Drug Discov. Today 2025, 30, 104367. [Google Scholar] [CrossRef]
  37. Levis, M. Midostaurin approved for FLT3-mutated AML. Blood 2017, 129, 3403–3406. [Google Scholar] [CrossRef]
  38. Zwaan, C.M.; Söderhäll, S.; Brethon, B.; Luciani, M.; Rizzari, C.; Stam, R.W.; Besse, E.; Dutreix, C.; Fagioli, F.; Ho, P.A.; et al. A phase 1/2, open-label, dose-escalation study of midostaurin in children with relapsed or refractory acute leukaemia. Br. J. Haematol. 2019, 185, 623–627. [Google Scholar] [CrossRef]
  39. Tzogani, K.; Yu, Y.; Meulendijks, D.; Herberts, C.; Hennik, P.; Verheijen, R.; Wangen, T.; Dahlseng Håkonsen, G.; Kaasboll, T.; Dalhus, M.; et al. European Medicines Agency review of midostaurin (Rydapt) for the treatment of adult patients with acute myeloid leukaemia and systemic mastocytosis. ESMO Open 2019, 4, e000606. [Google Scholar] [CrossRef]
  40. Perl, A.E.; Hosono, N.; Montesinos, P.; Podoltsev, N.; Martinelli, G.; Panoskaltsis, N.; Recher, C.; Smith, C.C.; Levis, M.J.; Strickland, S.; et al. Clinical outcomes in patients with relapsed/refractory FLT3-mutated acute myeloid leukemia treated with gilteritinib who received prior midostaurin or sorafenib. Blood Cancer J. 2022, 12, 84. [Google Scholar] [CrossRef]
  41. Perl, A.E.; Larson, R.A.; Podoltsev, N.A.; Strickland, S.; Wang, E.S.; Atallah, E.; Schiller, G.J.; Martinelli, G.; Neubauer, A.; Sierra, J.; et al. Follow-up of patients with R/R FLT3-mutation–positive AML treated with gilteritinib in the phase 3 ADMIRAL trial. Blood 2022, 139, 3366–3375. [Google Scholar] [CrossRef] [PubMed]
  42. Brinton, L.T.; Zhang, P.; Williams, K.; Canfield, D.; Orwick, S.; Sher, S.; Wasmuth, R.; Beaver, L.; Cempre, C.; Skinner, J.; et al. Synergistic effect of BCL2 and FLT3 co-inhibition in acute myeloid leukemia. J. Hematol. Oncol. 2020, 13, 139. [Google Scholar] [CrossRef]
  43. Ghia, P.; Pluta, A.; Wach, M.; Lysak, D.; Kozak, T.; Simkovic, M.; Kaplan, P.; Kraychok, I.; Illes, A.; de la Serna, J.; et al. ASCEND: Phase III, Randomized Trial of Acalabrutinib Versus Idelalisib Plus Rituximab or Bendamustine Plus Rituximab in Relapsed or Refractory Chronic Lymphocytic Leukemia. J. Clin. Oncol. 2020, 38, 2849–2861. [Google Scholar] [CrossRef] [PubMed]
  44. Vetrie, D.; Vořechovský, I.; Sideras, P.; Holland, J.; Davies, A.; Flinter, F.; Hammarström, L.; Kinnon, C.; Levinsky, R.; Bobrow, M.; et al. The gene involved in X-linked agammaglobulinaemia is a member of the src family of protein-tyrosine kinases. Nature 1993, 361, 226–233. [Google Scholar] [CrossRef]
  45. Sharman, J.P.; Coutre, S.E.; Furman, R.R.; Cheson, B.D.; Pagel, J.M.; Hillmen, P.; Barrientos, J.C.; Zelenetz, A.D.; Kipps, T.J.; Flinn, I.W.; et al. Final Results of a Randomized, Phase III Study of Rituximab With or Without Idelalisib Followed by Open-Label Idelalisib in Patients With Relapsed Chronic Lymphocytic Leukemia. J. Clin. Oncol. 2019, 37, 1391–1402. [Google Scholar] [CrossRef]
  46. Burger, J.A.; Barr, P.M.; Robak, T.; Owen, C.; Ghia, P.; Tedeschi, A.; Bairey, O.; Hillmen, P.; Coutre, S.E.; Devereux, S.; et al. Long-term efficacy and safety of first-line ibrutinib treatment for patients with CLL/SLL: 5 years of follow-up from the phase 3 RESONATE-2 study. Leukemia 2020, 34, 787–798. [Google Scholar] [CrossRef]
  47. Mattiello, V.; Barone, A.; Giannarelli, D.; Noto, A.; Cecchi, N.; Rampi, N.; Cassin, R.; Reda, G. Predictors of ibrutinib-associated atrial fibrillation: 5-year follow-up of a prospective study. Hematol. Oncol. 2023, 41, 363–370. [Google Scholar] [CrossRef]
  48. Woyach, J.A.; Blachly, J.S.; Rogers, K.A.; Bhat, S.A.; Jianfar, M.; Lozanski, G.; Weiss, D.M.; Andersen, B.L.; Gulrajani, M.; Frigault, M.M.; et al. Acalabrutinib plus Obinutuzumab in Treatment-Naïve and Relapsed/Refractory Chronic Lymphocytic Leukemia. Cancer Discov. 2020, 10, 394–405. [Google Scholar] [CrossRef]
  49. Byrd, J.C.; Hillmen, P.; Ghia, P.; Kater, A.P.; Chanan-Khan, A.; Furman, R.R.; O’Brien, S.; Yenerel, M.N.; Illés, A.; Kay, N.; et al. Acalabrutinib Versus Ibrutinib in Previously Treated Chronic Lymphocytic Leukemia: Results of the First Randomized Phase III Trial. J. Clin. Oncol. 2021, 39, 3441–3452. [Google Scholar] [CrossRef]
  50. Byrd, J.C.; Wierda, W.G.; Schuh, A.; Devereux, S.; Chaves, J.M.; Brown, J.R.; Hillmen, P.; Martin, P.; Awan, F.T.; Stephens, D.M.; et al. Acalabrutinib monotherapy in patients with relapsed/refractory chronic lymphocytic leukemia: Updated phase 2 results. Blood 2020, 135, 1204–1213. [Google Scholar] [CrossRef]
  51. Brown, J.R.; Byrd, J.C.; Coutre, S.E.; Benson, D.M.; Flinn, I.W.; Wagner-Johnston, N.D.; Spurgeon, S.E.; Kahl, B.S.; Bello, C.; Webb, H.K.; et al. Idelalisib, an inhibitor of phosphatidylinositol 3-kinase p110δ, for relapsed/refractory chronic lymphocytic leukemia. Blood 2014, 123, 3390–3397. [Google Scholar] [CrossRef] [PubMed]
  52. Maharaj, D.; Srinivasan, G.; Abreu, M.M.; Ko, M.-W.; Jewett, A.; Gouvea, J. Molecular Remission Using Low-Dose Immunotherapy with Minimal Toxicities for Poor Prognosis IGHV—Unmutated Chronic Lymphocytic Leukemia. Cells 2020, 10, 10. [Google Scholar] [CrossRef]
  53. Zhu, R.; Li, L.; Nguyen, B.; Seo, J.; Wu, M.; Seale, T.; Levis, M.; Duffield, A.; Hu, Y.; Small, D. FLT3 tyrosine kinase inhibitors synergize with BCL-2 inhibition to eliminate FLT3/ITD acute leukemia cells through BIM activation. Signal Transduct. Target. Ther. 2021, 6, 186. [Google Scholar] [CrossRef]
  54. Souers, A.J.; Leverson, J.D.; Boghaert, E.R.; Ackler, S.L.; Catron, N.D.; Chen, J.; Dayton, B.D.; Ding, H.; Enschede, S.H.; Fairbrother, W.J.; et al. ABT-199, a potent and selective BCL-2 inhibitor, achieves antitumor activity while sparing platelets. Nat. Med. 2013, 19, 202–208. [Google Scholar] [CrossRef] [PubMed]
  55. Anderson, M.A.; Deng, J.; Seymour, J.F.; Tam, C.; Kim, S.Y.; Fein, J.; Yu, L.; Brown, J.R.; Westerman, D.; Si, E.G.; et al. The BCL2 selective inhibitor venetoclax induces rapid onset apoptosis of CLL cells in patients via a TP53-independent mechanism. Blood 2016, 127, 3215–3224. [Google Scholar] [CrossRef]
  56. Stilgenbauer, S.; Eichhorst, B.; Schetelig, J.; Coutre, S.; Seymour, J.F.; Munir, T.; Puvvada, S.D.; Wendtner, C.-M.; Roberts, A.W.; Jurczak, W.; et al. Venetoclax in relapsed or refractory chronic lymphocytic leukaemia with 17p deletion: A multicentre, open-label, phase 2 study. Lancet Oncol. 2016, 17, 768–778. [Google Scholar] [CrossRef]
  57. Blombery, P.; Lew, T.E.; Dengler, M.A.; Thompson, E.R.; Lin, V.S.; Chen, X.; Nguyen, T.; Panigrahi, A.; Handunnetti, S.M.; Carney, D.A.; et al. Clonal hematopoiesis, myeloid disorders and BAX-mutated myelopoiesis in patients receiving venetoclax for CLL. Blood 2022, 139, 1198–1207. [Google Scholar] [CrossRef]
  58. Jabbour, E.; O’Brien, S.; Ravandi, F.; Kantarjian, H. Monoclonal antibodies in acute lymphoblastic leukemia. Blood 2015, 125, 4010–4016. [Google Scholar] [CrossRef] [PubMed]
  59. Hagemeister, F. Rituximab for the Treatment of Non-Hodgkin’s Lymphoma and Chronic Lymphocytic Leukaemia. Drugs 2010, 70, 261–272. [Google Scholar] [CrossRef]
  60. Ribrag, V.; Koscielny, S.; Bosq, J.; Leguay, T.; Casasnovas, O.; Fornecker, L.-M.; Recher, C.; Ghesquieres, H.; Morschhauser, F.; Girault, S.; et al. Rituximab and dose-dense chemotherapy for adults with Burkitt’s lymphoma: A randomised, controlled, open-label, phase 3 trial. Lancet 2016, 387, 2402–2411. [Google Scholar] [CrossRef]
  61. Maury, S.; Chevret, S.; Thomas, X.; Heim, D.; Leguay, T.; Huguet, F.; Chevallier, P.; Hunault, M.; Boissel, N.; Escoffre-Barbe, M.; et al. Rituximab in B-Lineage Adult Acute Lymphoblastic Leukemia. N. Engl. J. Med. 2016, 375, 1044–1053. [Google Scholar] [CrossRef] [PubMed]
  62. Hillmen, P.; Pitchford, A.; Bloor, A.; Broom, A.; Young, M.; Kennedy, B.; Walewska, R.; Furtado, M.; Preston, G.; Neilson, J.R.; et al. Ibrutinib and rituximab versus fludarabine, cyclophosphamide, and rituximab for patients with previously untreated chronic lymphocytic leukaemia (FLAIR): Interim analysis of a multicentre, open-label, randomised, phase 3 trial. Lancet Oncol. 2023, 24, 535–552. [Google Scholar] [CrossRef]
  63. Tiacci, E.; De Carolis, L.; Simonetti, E.; Capponi, M.; Ambrosetti, A.; Lucia, E.; Antolino, A.; Pulsoni, A.; Ferrari, S.; Zinzani, P.L.; et al. Vemurafenib plus Rituximab in Refractory or Relapsed Hairy-Cell Leukemia. N. Engl. J. Med. 2021, 384, 1810–1823. [Google Scholar] [CrossRef]
  64. Veeramani, S.; Wang, S.-Y.; Dahle, C.; Blackwell, S.; Jacobus, L.; Knutson, T.; Button, A.; Link, B.K.; Weiner, G.J. Rituximab infusion induces NK activation in lymphoma patients with the high-affinity CD16 polymorphism. Blood 2011, 118, 3347–3349. [Google Scholar] [CrossRef]
  65. Urlaub, D.; Zhao, S.; Blank, N.; Bergner, R.; Claus, M.; Tretter, T.; Lorenz, H.-M.; Watzl, C.; Merkt, W. Activation of natural killer cells by rituximab in granulomatosis with polyangiitis. Arthritis Res. Ther. 2019, 21, 277. [Google Scholar] [CrossRef]
  66. Shan, D.; Ledbetter, J.A.; Press, O.W. Apoptosis of Malignant Human B Cells by Ligation of CD20 With Monoclonal Antibodies. Blood 1998, 91, 1644–1652. [Google Scholar] [CrossRef]
  67. Herlyn, D.M.; Koprowski, H. Monoclonal anticolon carcinoma antibodies in complement-dependent cytotoxicity. Int. J. Cancer 1981, 27, 769–774. [Google Scholar] [CrossRef]
  68. Kamen, L.; Myneni, S.; Langsdorf, C.; Kho, E.; Ordonia, B.; Thakurta, T.; Zheng, K.; Song, A.; Chung, S. A novel method for determining antibody-dependent cellular phagocytosis. J. Immunol. Methods 2019, 468, 55–60. [Google Scholar] [CrossRef]
  69. Nieva, J.; Bethel, K.; Saven, A. Phase 2 study of rituximab in the treatment of cladribine-failed patients with hairy cell leukemia. Blood 2003, 102, 810–813. [Google Scholar] [CrossRef] [PubMed]
  70. Amitai, I.; Gafter-Gvili, A.; Shargian-Alon, L.; Raanani, P.; Gurion, R. Obinutuzumab-related adverse events: A systematic review and meta-analysis. Hematol. Oncol. 2021, 39, 215–221. [Google Scholar] [CrossRef] [PubMed]
  71. Samineni, D.; Gibiansky, L.; Wang, B.; Vadhavkar, S.; Rajwanshi, R.; Tandon, M.; Sinha, A.; Al-Sawaf, O.; Fischer, K.; Hallek, M.; et al. Pharmacokinetics and Exposure-Response Analysis of Venetoclax + Obinutuzumab in Chronic Lymphocytic Leukemia: Phase 1b Study and Phase 3 CLL14 Trial. Adv. Ther. 2022, 39, 3635–3653. [Google Scholar] [CrossRef]
  72. Cartron, G.; de Guibert, S.; Dilhuydy, M.-S.; Morschhauser, F.; Leblond, V.; Dupuis, J.; Mahe, B.; Bouabdallah, R.; Lei, G.; Wenger, M.; et al. Obinutuzumab (GA101) in relapsed/refractory chronic lymphocytic leukemia: Final data from the phase 1/2 GAUGUIN study. Blood 2014, 124, 2196–2202. [Google Scholar] [CrossRef] [PubMed]
  73. Brischwein, K.; Parr, L.; Pflanz, S.; Volkland, J.; Lumsden, J.; Klinger, M.; Locher, M.; Hammond, S.A.; Kiener, P.; Kufer, P.; et al. Strictly Target Cell-dependent Activation of T Cells by Bispecific Single-chain Antibody Constructs of the BiTE Class. J. Immunother. 2007, 30, 798–807. [Google Scholar] [CrossRef]
  74. Yuraszeck, T.; Kasichayanula, S.; Benjamin, J. Translation and Clinical Development of Bispecific T-cell Engaging Antibodies for Cancer Treatment. Clin. Pharmacol. Ther. 2017, 101, 634–645. [Google Scholar] [CrossRef]
  75. Jabbour, E.; Short, N.J.; Jain, N.; Thompson, P.A.; Kadia, T.M.; Ferrajoli, A.; Huang, X.; Yilmaz, M.; Alvarado, Y.; Patel, K.P.; et al. Hyper-CVAD and sequential blinatumomab for newly diagnosed Philadelphia chromosome-negative B-cell acute lymphocytic leukaemia: A single-arm, single-centre, phase 2 trial. Lancet Haematol. 2022, 9, e878–e885. [Google Scholar] [CrossRef]
  76. Kantarjian, H.; Stein, A.; Gökbuget, N.; Fielding, A.K.; Schuh, A.C.; Ribera, J.-M.; Wei, A.; Dombret, H.; Foà, R.; Bassan, R.; et al. Blinatumomab versus Chemotherapy for Advanced Acute Lymphoblastic Leukemia. N. Engl. J. Med. 2017, 376, 836–847. [Google Scholar] [CrossRef] [PubMed]
  77. Gökbuget, N.; Dombret, H.; Bonifacio, M.; Reichle, A.; Graux, C.; Faul, C.; Diedrich, H.; Topp, M.S.; Brüggemann, M.; Horst, H.-A.; et al. Blinatumomab for minimal residual disease in adults with B-cell precursor acute lymphoblastic leukemia. Blood 2018, 131, 1522–1531. [Google Scholar] [CrossRef]
  78. Huang, M.; Ye, Y.; Chen, S.; Chai, J.; Lu, J.; Zhoa, L.; Gu, L.; Wang, Z. Use of all-trans retinoic acid in the treatment of acute promyelocytic leukemia. Blood 1988, 72, 567–572. [Google Scholar] [CrossRef]
  79. Liang, C.; Qiao, G.; Liu, Y.; Tian, L.; Hui, N.; Li, J.; Ma, Y.; Li, H.; Zhao, Q.; Cao, W.; et al. Overview of all-trans-retinoic acid (ATRA) and its analogues: Structures, activities, and mechanisms in acute promyelocytic leukaemia. Eur. J. Med. Chem. 2021, 220, 113451. [Google Scholar] [CrossRef] [PubMed]
  80. Sun, X.; Mu, X.; Li, F.; Wang, Y.; Yang, X.; Guo, Q. Roles of PADI4 in the expression of cytokines involved in inflammation and adhesion in differentiated NB4 cells treated with ATRA. Exp. Ther. Med. 2018, 25, 118. [Google Scholar] [CrossRef]
  81. Fiskus, W.; Daver, N.; Boettcher, S.; Mill, C.P.; Sasaki, K.; Birdwell, C.E.; Davis, J.A.; Das, K.; Takahashi, K.; Kadia, T.M.; et al. Activity of menin inhibitor ziftomenib (KO-539) as monotherapy or in combinations against AML cells with MLL1 rearrangement or mutant NPM1. Leukemia 2022, 36, 2729–2733. [Google Scholar] [CrossRef] [PubMed]
  82. Wang, E.S.; Issa, G.C.; Erba, H.P.; Altman, J.K.; Montesinos, P.; DeBotton, S.; Walter, R.B.; Pettit, K.; Savona, M.R.; Shah, M.V.; et al. Ziftomenib in relapsed or refractory acute myeloid leukaemia (KOMET-001): A multicentre, open-label, multi-cohort, phase 1 trial. Lancet Oncol. 2024, 25, 1310–1324. [Google Scholar] [CrossRef] [PubMed]
  83. Reiff, S.D.; Mantel, R.; Smith, L.L.; Greene, J.T.; Muhowski, E.M.; Fabian, C.A.; Goettl, V.M.; Tran, M.; Harrington, B.K.; Rogers, K.A.; et al. The BTK Inhibitor ARQ 531 Targets Ibrutinib-Resistant CLL and Richter Transformation. Cancer Discov. 2018, 8, 1300–1315. [Google Scholar] [CrossRef]
  84. Woyach, J.A.; Stephens, D.M.; Flinn, I.W.; Bhat, S.A.; Savage, R.E.; Chai, F.; Eathiraj, S.; Reiff, S.D.; Muhowski, E.M.; Granlund, L.; et al. First-in-Human Study of the Reversible BTK Inhibitor Nemtabrutinib in Patients with Relapsed/Refractory Chronic Lymphocytic Leukemia and B-Cell Non–Hodgkin Lymphoma. Cancer Discov. 2024, 14, 66–75. [Google Scholar] [CrossRef]
  85. Hochhaus, A.; O’Brien, S.G.; Guilhot, F.; Druker, B.J.; Branford, S.; Foroni, L.; Goldman, J.M.; Müller, M.C.; Radich, J.P.; Rudoltz, M.; et al. Six-year follow-up of patients receiving imatinib for the first-line treatment of chronic myeloid leukemia. Leukemia 2009, 23, 1054–1061. [Google Scholar] [CrossRef] [PubMed]
  86. DiNardo, C.D.; Jonas, B.A.; Pullarkat, V.; Thirman, M.J.; Garcia, J.S.; Wei, A.H.; Konopleva, M.; Döhner, H.; Letai, A.; Fenaux, P.; et al. Azacitidine and Venetoclax in Previously Untreated Acute Myeloid Leukemia. N. Engl. J. Med. 2020, 383, 617–629. [Google Scholar] [CrossRef]
  87. Jabbour, E.; Kantarjian, H.M.; Aldoss, I.; Montesinos, P.; Leonard, J.T.; Gómez-Almaguer, D.; Baer, M.R.; Gambacorti-Passerini, C.; McCloskey, J.; Minami, Y.; et al. Ponatinib vs Imatinib in Frontline Philadelphia Chromosome–Positive Acute Lymphoblastic Leukemia. JAMA 2024, 331, 1814. [Google Scholar] [CrossRef]
  88. Kantarjian, H.; Short, N.J.; Jain, N.; Sasaki, K.; Huang, X.; Haddad, F.G.; Khouri, I.; DiNardo, C.D.; Pemmaraju, N.; Wierda, W.; et al. Frontline combination of ponatinib and hyper-CVAD in Philadelphia chromosome-positive acute lymphoblastic leukemia: 80-months follow-up results. Am. J. Hematol. 2023, 98, 493–501. [Google Scholar] [CrossRef]
  89. DiNardo, C.D.; Lachowiez, C.A.; Takahashi, K.; Loghavi, S.; Kadia, T.; Daver, N.; Xiao, L.; Adeoti, M.; Short, N.J.; Sasaki, K.; et al. Venetoclax combined with FLAG-IDA induction and consolidation in newly diagnosed acute myeloid leukemia. Am. J. Hematol. 2022, 97, 1035–1043. [Google Scholar] [CrossRef]
  90. Stone, R.M.; Mandrekar, S.J.; Sanford, B.L.; Laumann, K.; Geyer, S.; Bloomfield, C.D.; Thiede, C.; Prior, T.W.; Döhner, K.; Marcucci, G.; et al. Midostaurin plus Chemotherapy for Acute Myeloid Leukemia with a FLT3 Mutation. N. Engl. J. Med. 2017, 377, 454–464. [Google Scholar] [CrossRef]
  91. Sierra, J.; Montesinos, P.; Thomas, X.; Griskevicius, L.; Cluzeau, T.; Caillot, D.; Legrand, O.; Minotti, C.; Luppi, M.; Farkas, F.; et al. Midostaurin plus daunorubicin or idarubicin for young and older adults with FLT3-mutated AML: A phase 3b trial. Blood Adv. 2023, 7, 6441–6450. [Google Scholar] [CrossRef]
  92. Morita, K.; Kantarjian, H.M.; Sasaki, K.; Issa, G.C.; Jain, N.; Konopleva, M.; Short, N.J.; Takahashi, K.; DiNardo, C.D.; Kadia, T.M.; et al. Outcome of patients with chronic myeloid leukemia in lymphoid blastic phase and Philadelphia chromosome–positive acute lymphoblastic leukemia treated with hyper-CVAD and dasatinib. Cancer 2021, 127, 2641–2647. [Google Scholar] [CrossRef]
  93. Abaza, Y.; Kantarjian, H.; Alwash, Y.; Borthakur, G.; Champlin, R.; Kadia, T.; Garcia-Manero, G.; Daver, N.; Ravandi, F.; Verstovsek, S.; et al. Phase I/II study of dasatinib in combination with decitabine in patients with accelerated or blast phase chronic myeloid leukemia. Am. J. Hematol. 2020, 95, 1288–1295. [Google Scholar] [CrossRef]
  94. Marcucci, G.; Geyer, S.; Laumann, K.; Zhao, W.; Bucci, D.; Uy, G.L.; Blum, W.; Eisfeld, A.-K.; Pardee, T.S.; Wang, E.S.; et al. Combination of dasatinib with chemotherapy in previously untreated core binding factor acute myeloid leukemia: CALGB 10801. Blood Adv. 2020, 4, 696–705. [Google Scholar] [CrossRef]
  95. Cairoli, R.; Gatti, A.; Grillo, G.; Stefanucci, M.R.; Di Camillo, B.; Fumagalli, M.; Krampera, M.; Nadali, G.; Zappasodi, P.; Borlenghi, E.; et al. Efficacy of Midostaurin Combined With Intensive Chemotherapy in Core Binding Factor Leukemia: A Phase II Clinical Trial. Am. J. Hematol. 2025, 100, 346–349. [Google Scholar] [CrossRef]
  96. Döhner, H.; Weber, D.; Krzykalla, J.; Fiedler, W.; Wulf, G.; Salih, H.; Lübbert, M.; Kühn, M.W.M.; Schroeder, T.; Salwender, H.; et al. Midostaurin plus intensive chemotherapy for younger and older patients with AML and FLT3 internal tandem duplications. Blood Adv. 2022, 6, 5345–5355. [Google Scholar] [CrossRef]
  97. Pratz, K.W.; Cherry, M.; Altman, J.K.; Cooper, B.W.; Podoltsev, N.A.; Cruz, J.C.; Lin, T.L.; Schiller, G.J.; Jurcic, J.G.; Asch, A.; et al. Gilteritinib in Combination With Induction and Consolidation Chemotherapy and as Maintenance Therapy: A Phase IB Study in Patients With Newly Diagnosed AML. J. Clin. Oncol. 2023, 41, 4236–4246. [Google Scholar] [CrossRef]
  98. Stilgenbauer, S.; Bosch, F.; Ilhan, O.; Kisro, J.; Mahé, B.; Mikuskova, E.; Osmanov, D.; Reda, G.; Robinson, S.; Tausch, E.; et al. Safety and efficacy of obinutuzumab alone or with chemotherapy in previously untreated or relapsed/refractory chronic lymphocytic leukaemia patients: Final analysis of the Phase IIIb GREEN study. Br. J. Haematol. 2021, 193, 325–338. [Google Scholar] [CrossRef]
  99. Gugliotta, G.; Castagnetti, F.; Breccia, M.; Gozzini, A.; Usala, E.; Carella, A.M.; Rege-Cambrin, G.; Martino, B.; Abruzzese, E.; Albano, F.; et al. Rotation of nilotinib and imatinib for first-line treatment of chronic phase chronic myeloid leukemia. Am. J. Hematol. 2016, 91, 617–622. [Google Scholar] [CrossRef]
  100. Jabbour, E.; Haddad, F.G.; Sasaki, K.; Carter, B.Z.; Alvarado, Y.; Nasnas, C.; Nasr, L.; Masarova, L.; Daver, N.; Pemmaraju, N.; et al. Combination of dasatinib and venetoclax in newly diagnosed chronic phase chronic myeloid leukemia. Cancer 2024, 130, 2652–2659. [Google Scholar] [CrossRef] [PubMed]
  101. Spampinato, M.; Zuppelli, T.; Dulcamare, I.; Longhitano, L.; Sambataro, D.; Santisi, A.; Alanazi, A.M.; Barbagallo, I.A.; Vicario, N.; Parenti, R.; et al. Enhanced Antitumor Activity by the Combination of Dasatinib and Selinexor in Chronic Myeloid Leukemia. Pharmaceuticals 2024, 17, 894. [Google Scholar] [CrossRef] [PubMed]
  102. Crawford, L.J.; Chan, E.T.; Aujay, M.; Holyoake, T.L.; Melo, J.V.; Jorgensen, H.G.; Suresh, S.; Walker, B.; Irvine, A.E. Synergistic effects of proteasome inhibitor carfilzomib in combination with tyrosine kinase inhibitors in imatinib-sensitive and -resistant chronic myeloid leukemia models. Oncogenesis 2024, 3, e90. [Google Scholar] [CrossRef] [PubMed]
  103. Goede, V.; Fischer, K.; Robrecht, S.; Giza, A.; Dyer, M.J.; Eckart, M.J.; Müller, L.; Smolej, L.; Kutsch, N.; Fink, A.-M.; et al. Long-term outcomes of chemoimmunotherapy with obinutuzumab/chlorambucil in chronic lymphocytic leukemia. Blood Adv. 2025, 9, 2431–2435. [Google Scholar] [CrossRef]
  104. Al-Sawaf, O.; Robrecht, S.; Zhang, C.; Olivieri, S.; Chang, Y.M.; Fink, A.M.; Tausch, E.; Schneider, C.; Ritgen, M.; Kreuzer, K.-A.; et al. Venetoclax-obinutuzumab for previously untreated chronic lymphocytic leukemia: 6-year results of the randomized phase 3 CLL14 study. Blood 2024, 144, 1924–1935. [Google Scholar] [CrossRef] [PubMed]
  105. Sharman, J.P.; Egyed, M.; Jurczak, W.; Skarbnik, A.P.; Patel, K.; Flinn, I.W.; Kamdar, M.; Munir, T.; Walewska, R.; Hughes, M.; et al. Acalabrutinib-Obinutuzumab Improves Survival vs Chemoimmunotherapy in treatment-naive CLL in the 6-year Follow-up of ELEVATE-TN. Blood J. 2025, in press. [Google Scholar] [CrossRef]
  106. Eide, C.A.; Brewer, D.; Xie, T.; Schultz, A.R.; Savage, S.L.; Muratcioglu, S.; Merz, N.; Press, R.D.; O’Hare, T.; Jacob, T.; et al. Overcoming clinical BCR-ABL1 compound mutant resistance with combined ponatinib and asciminib therapy. Cancer Cell 2024, 42, 1486–1488. [Google Scholar] [CrossRef]
  107. Przespolewski, E.R.; Baron, J.; Kashef, F.; Fu, K.; Jani Sait, S.N.; Hernandez-Ilizaliturri, F.; Thompson, J. Concomitant Venetoclax and Imatinib for Comanaging Chronic Lymphocytic Leukemia and Chronic Myeloid Leukemia: A Case Report. J. Natl. Compr. Canc. Netw. 2023, 21, 102–107. [Google Scholar] [CrossRef]
  108. Brown, J.R.; Seymour, J.F.; Jurczak, W.; Aw, A.; Wach, M.; Illes, A.; Tedeschi, A.; Owen, C.; Skarbnik, A.; Lysak, D.; et al. Fixed-Duration Acalabrutinib Combinations in Untreated Chronic Lymphocytic Leukemia. N. Engl. J. Med. 2025, 392, 748–762. [Google Scholar] [CrossRef]
  109. Danilov, A.V.; Herbaux, C.; Walter, H.S.; Hillmen, P.; Rule, S.A.; Kio, E.A.; Karlin, L.; Dyer, M.J.S.; Mitra, S.S.; Yi, P.C.; et al. Phase Ib Study of Tirabrutinib in Combination with Idelalisib or Entospletinib in Previously Treated Chronic Lymphocytic Leukemia. Clin. Cancer Res. 2020, 26, 2810–2818. [Google Scholar] [CrossRef]
  110. Zicari, S.; Merlino, G.; Paoli, A.; Fiascarelli, A.; Tunici, P.; Bisignano, D.; Belli, F.; Irrissuto, C.; Talucci, S.; Cirigliano, E.; et al. The Dual PIM/FLT3 Inhibitor MEN1703 Combines Synergistically With Gilteritinib in FLT3-ITD-Mutant Acute Myeloid Leukaemia. J. Cell. Mol. Med. 2024, 28, e70235. [Google Scholar] [CrossRef]
  111. Zhou, Q.; Guan, Y.; Zhao, P.; Chu, H.; Xi, Y. Combined anti-leukemic effect of gilteritinib and GSK-J4 in FLT3-ITD+ acute myeloid leukemia. Transl. Oncol. 2025, 52, 102271. [Google Scholar] [CrossRef]
  112. Sharman, J.P.; Brander, D.M.; Mato, A.R.; Ghosh, N.; Schuster, S.J.; Kambhampati, S.; Burke, J.M.; Lansigan, F.; Schreeder, M.T.; Lunin, S.D.; et al. Ublituximab plus ibrutinib versus ibrutinib alone for patients with relapsed or refractory high-risk chronic lymphocytic leukaemia (GENUINE): A phase 3, multicentre, open-label, randomised trial. Lancet Haematol. 2021, 8, e254–e266. [Google Scholar] [CrossRef] [PubMed]
  113. Seymour, J.F.; Kipps, T.J.; Eichhorst, B.; Hillmen, P.; D’Rozario, J.; Assouline, S.; Owen, C.; Gerecitano, J.; Robak, T.; De la Serna, J.; et al. Venetoclax–Rituximab in Relapsed or Refractory Chronic Lymphocytic Leukemia. N. Engl. J. Med. 2018, 378, 1107–1120. [Google Scholar] [CrossRef]
  114. Foà, R.; Bassan, R.; Vitale, A.; Elia, L.; Piciocchi, A.; Puzzolo, M.-C.; Canichella, M.; Viero, P.; Ferrara, F.; Lunghi, M.; et al. Dasatinib–Blinatumomab for Ph-Positive Acute Lymphoblastic Leukemia in Adults. N. Engl. J. Med. 2020, 383, 1613–1623. [Google Scholar] [CrossRef]
  115. Lee, H.H.; Kim, I.; Kim, U.K.; Choi, S.S.; Kim, T.Y.; Lee, D.; Lee, Y.; Lee, J.; Jo, J.; Lee, Y.-T.; et al. Therapeutic effiacy of T cells expressing chimeric antigen receptor derived from a mesothelin-specific scFv in orthotopic human pancreatic cancer animal models. Neoplasia 2022, 24, 98–108. [Google Scholar] [CrossRef] [PubMed]
  116. Wei, Z.; Cheng, Q.; Xu, N.; Zhao, C.; Xu, J.; Kang, L.; Lou, X.; Yu, L.; Feng, W. Investigation of CRS-associated cytokines in CAR-T therapy with meta-GNN and pathway crosstalk. BMC Bioinform. 2022, 23, 373. [Google Scholar] [CrossRef] [PubMed]
  117. Castellote-Borrell, M.; Domingo, M.; Merlina, F.; Lu, H.; Colell, S.; Bachiller, M.; Juan, M.; Guedan, S.; Faraudo, J.; Guasch, J. Lymph-Node Inspired Hydrogels Enhance CAR Expression and Proliferation of CAR T Cells. ACS Appl. Mater. Interfaces 2025, 17, 16548–16560. [Google Scholar] [CrossRef]
  118. Hill, J.A.; Li, D.; Hay, K.A.; Green, M.L.; Cherian, S.; Chen, X.; Riddell, S.R.; Maloney, D.G.; Boeckh, M.; Turtle, C.J. Infectious complications of CD19-targeted chimeric antigen receptor–modified T-cell immunotherapy. Blood 2018, 131, 121–130. [Google Scholar] [CrossRef]
  119. Gill, S.; Vides, V.; Frey, N.V.; Hexner, E.O.; Metzger, S.; O’Brien, M.; Hwang, W.-T.; Brogdon, J.L.; Davis, M.M.; Fraietta, J.A.; et al. Anti-CD19 CAR T cells in combination with ibrutinib for the treatment of chronic lymphocytic leukemia. Blood Adv. 2022, 6, 5774–5785. [Google Scholar] [CrossRef]
  120. Zhang, M.; Fu, S.; Feng, J.; Hong, R.; Wei, G.; Zhao, H.; Zhao, M.; Xu, H.; Cui, J.; Huang, S.; et al. Dasatinib and CAR T-Cell Therapy in Newly Diagnosed Philadelphia Chromosome–Positive Acute Lymphoblastic Leukemia. JAMA Oncol. 2025, 11, 625–629. [Google Scholar] [CrossRef]
  121. Li, K.; Wu, H.; Pan, W.; Guo, M.; Qiu, D.; He, Y.; Li, Y.; Yang, D.-H.; Huang, Y. A novel approach for relapsed/refractory FLT3mut+ acute myeloid leukaemia: Synergistic effect of the combination of bispecific FLT3scFv/NKG2D-CAR T cells and gilteritinib. Mol. Cancer 2022, 21, 66. [Google Scholar] [CrossRef] [PubMed]
  122. Funk, C.R.; Wang, S.; Chen, K.Z.; Waller, A.; Sharma, A.; Edgar, C.L.; Gupta, V.A.; Chandrakasan, S.; Zoine, J.T.; Fedanov, A.; et al. PI3Kδ/γ inhibition promotes human CART cell epigenetic and metabolic reprogramming to enhance antitumor cytotoxicity. Blood 2022, 139, 523–537. [Google Scholar] [CrossRef] [PubMed]
  123. Jain, N.; Thompson, P.; Burger, J.; Ferrajoli, A.; Takahashi, K.; Estrov, Z.; Borthakur, G.; Bose, P.; Kadia, T.; Pemmaraju, N.; et al. Ibrutinib, fludarabine, cyclophosphamide, and obinutuzumab (iFCG) regimen for chronic lymphocytic leukemia (CLL) with mutated IGHV and without TP53 aberrations. Leukemia 2021, 35, 3421–3429. [Google Scholar] [CrossRef]
  124. Kutsch, N.; Pallasch, C.; Decker, T.; Hebart, H.; Chow, K.U.; Graeven, U.; Kisro, J.; Kroeber, A.; Tausch, E.; Fischer, K.; et al. Efficacy and Safety of Tirabrutinib and Idelalisib With or Without Obinutuzumab in Relapsed Chronic Lymphocytic Leukemia. HemaSphere 2020, 6, e729. [Google Scholar] [CrossRef] [PubMed]
  125. Fürstenau, M.; Robrecht, S.; Schneider, C.; Tausch, E.; Giza, A.; Ritgen, M.; Bittenbring, J.; Hebart, H.; Schöttker, B.; Illert, A.L.; et al. MRD-guided zanubrutinib, venetoclax, and obinutuzumab in relapsed CLL: Primary end point analysis from the CLL2-BZAG trial. Blood 2025, 145, 1282–1292. [Google Scholar] [CrossRef]
  126. Rogers, K.A.; Huang, Y.; Ruppert, A.S.; Abruzzo, L.V.; Andersen, B.L.; Awan, F.T.; Bhat, S.A.; Dean, A.; Lucas, M.; Banks, C.; et al. Phase II Study of Combination Obinutuzumab, Ibrutinib, and Venetoclax in Treatment-Naïve and Relapsed or Refractory Chronic Lymphocytic Leukemia. J. Clin. Oncol. 2020, 38, 3626–3637. [Google Scholar] [CrossRef]
  127. Short, N.J.; Daver, N.; Dinardo, C.D.; Kadia, T.; Nasr, L.F.; Macaron, W.; Yilmaz, M.; Borthakur, G.; Montalban-Bravo, G.; Garcia-Manero, G.; et al. Azacitidine, Venetoclax, and Gilteritinib in Newly Diagnosed and Relapsed or Refractory FLT3-Mutated AML. J. Clin. Oncol. 2024, 42, 1499–1508. [Google Scholar] [CrossRef]
  128. Luskin, M.R.; Murakami, M.A.; Keating, J.; Flamand, Y.; Winer, E.S.; Garcia, J.S.; Stahl, M.; Stone, R.M.; Wadleigh, M.; Jaeckle, S.L.; et al. Asciminib plus dasatinib and prednisone for Philadelphia chromosome–positive acute leukemia. Blood 2025, 145, 577–589. [Google Scholar] [CrossRef]
  129. Advani, A.S.; Moseley, A.; O’Dwyer, K.M.; Wood, B.L.; Park, J.; Wieduwilt, M.; Jeyakumar, D.; Yaghmour, G.; Atallah, E.L.; Gerds, A.T.; et al. Dasatinib/prednisone induction followed by blinatumomab/dasatinib in Ph+ acute lymphoblastic leukemia. Blood Adv. 2023, 7, 1279–1285. [Google Scholar] [CrossRef]
  130. Short, N.J.; Nguyen, D.; Jabbour, E.; Senapati, J.; Zeng, Z.; Issa, G.C.; Abbas, H.; Nasnas, C.; Qiao, W.; Huang, X.; et al. Decitabine, venetoclax, and ponatinib for advanced phase chronic myeloid leukaemia and Philadelphia chromosome-positive acute myeloid leukaemia: A single-arm, single-centre phase 2 trial. Lancet Haematol. 2024, 11, e839–e849. [Google Scholar] [CrossRef]
  131. Moore, A.; Soosay Raj, T.; Smith, A. Vincristine sulfate liposomal injection for acute lymphoblastic leukemia. Int. J. Nanomed. 2013, 8, 4361–4369. [Google Scholar] [CrossRef]
  132. Watson, N.A.; Notley, R.G. Urological Complications of Cyclophosphamide. Br. J. Urol. 1973, 45, 606–609. [Google Scholar] [CrossRef] [PubMed]
  133. Hanekamp, D.; Denys, B.; Kaspers, G.J.L.; te Marvelde, J.G.; Schuurhuis, G.J.; De Haas, V.; De Moerloose, B.; de Bont, E.S.; Zwaan, C.M.; de Jong, A.; et al. Leukaemic stem cell load at diagnosis predicts the development of relapse in young acute myeloid leukaemia patients. Br. J. Haematol. 2018, 183, 512–516. [Google Scholar] [CrossRef] [PubMed]
  134. Hyland, K.A.; Nelson, A.M.; Eisel, S.L.; Hoogland, A.I.; Ibarz-Pinilla, J.; Sweet, K.; Jacobsen, P.B.; Knoop, H.; Jim, H.S.L. Fatigue Perpetuating Factors as Mediators of Change in a Cognitive Behavioral Intervention for Targeted Therapy-Related Fatigue in Chronic Myeloid Leukemia: A Pilot Study. Ann. Behav. Med. 2022, 56, 137–145. [Google Scholar] [CrossRef]
  135. Wang, H.; Chan, K.Y.Y.; Cheng, C.K.; Ng, M.H.L.; Lee, P.Y.; Cheng, F.W.T.; Lam, G.K.S.; Chow, T.W.; Ha, S.Y.; Chiang, A.K.S.; et al. Pharmacogenomic Profiling of Pediatric Acute Myeloid Leukemia to Identify Therapeutic Vulnerabilities and Inform Functional Precision Medicine. Blood Cancer Discov. 2022, 3, 516–535. [Google Scholar] [CrossRef] [PubMed]
  136. Marrero, R.J.; Wu, H.; Cao, X.; Parcha, P.K.; Elsayed, A.H.; Inaba, H.; Kuo, D.J.; Degar, B.A.; Heym, K.; Taub, J.W.; et al. Pharmacogenomic Score Effectively Personalizes Treatment of Acute Myeloid Leukemia. Clin. Cancer Res. 2024, 30, 4388–4396. [Google Scholar] [CrossRef]
  137. Luo, Q.; Raulston, E.G.; Prado, M.A.; Wu, X.; Gritsman, K.; Whalen, K.S.; Yan, K.; Booth, C.A.G.; Xu, R.; van Galen, P.; et al. Targetable leukaemia dependency on noncanonical PI3Kγ signalling. Nature 2024, 630, 198–205. [Google Scholar] [CrossRef]
Figure 1. Chemical structural formulae of Imatinib, Dasatinib, Ponatinib, Midostaurin, Gilteritinib, Ibrutinib, Acalabrutinib, Idelalisib, Venetoclax, Ziftomenib, Nemtabrutinib.
Figure 1. Chemical structural formulae of Imatinib, Dasatinib, Ponatinib, Midostaurin, Gilteritinib, Ibrutinib, Acalabrutinib, Idelalisib, Venetoclax, Ziftomenib, Nemtabrutinib.
Biomedicines 13 01652 g001
Figure 2. Mechanism of action diagram for Imatinib/Dasatinib/Ponatinib and Midostaurin/Gilteritinib, etc. (a) Imatinib/Dasatinib/Ponatinib, etc., selectively inhibits the ATP-binding site of the BCR-ABL fusion protein (Ponatinib can be adapted to T315I mutants that are resistant to other TKIs), blocking its tyrosine kinase activity, inhibiting signaling pathways such as RAS/MAPK, PI3K/AKT, STAT, etc., and blocking leukemia cell proliferation and survival. (b) Midostaurin/Gilteritinib reduces leukemia cell proliferation and induces apoptosis by binding highly selectively to the ATP-binding site of FLT3 mutant proteins, inhibiting their kinase activity, and blocking the phosphorylation of downstream signaling pathways (e.g., STAT5, Ras/MAPK).
Figure 2. Mechanism of action diagram for Imatinib/Dasatinib/Ponatinib and Midostaurin/Gilteritinib, etc. (a) Imatinib/Dasatinib/Ponatinib, etc., selectively inhibits the ATP-binding site of the BCR-ABL fusion protein (Ponatinib can be adapted to T315I mutants that are resistant to other TKIs), blocking its tyrosine kinase activity, inhibiting signaling pathways such as RAS/MAPK, PI3K/AKT, STAT, etc., and blocking leukemia cell proliferation and survival. (b) Midostaurin/Gilteritinib reduces leukemia cell proliferation and induces apoptosis by binding highly selectively to the ATP-binding site of FLT3 mutant proteins, inhibiting their kinase activity, and blocking the phosphorylation of downstream signaling pathways (e.g., STAT5, Ras/MAPK).
Biomedicines 13 01652 g002
Figure 3. Mechanism of action diagram for Ibrutinib/Acalabrutinib/Idelalisib and Venetoclax. (a) Ibrutinib/Acalabrutinib covalently binds to Cys-481 near the ATP-binding domain of BTK, blocking BTK activation and inhibiting the BCR signaling pathway. Inhibition of BTK leads to blockage of activation of downstream signaling molecules such as tour ERK1/2, PI3K, and NF-κB, which inhibits leukemic B cell proliferation and survival. Idelalisib binds to the ATP binding site of PI3Kδ, inhibits the activity of PI3Kδ, blocks the BCR signaling pathway, which in turn leads to the blockage of downstream signaling pathways such as AKT, MAPK, NF-κB, etc., and inhibits the proliferation and survival of leukemic B cells. (b) Venetoclax replaces and releases pro-apoptotic proteins (e.g., BAX, BAK) by directly binding to Bcl-2 proteins, which initiates an apoptotic cascade that leads to the formation of a pore in the outer mitochondrial membrane, the release of cytochromes, and the further activation of caspase-3/7, which leads to the apoptotic death of malignant cells.
Figure 3. Mechanism of action diagram for Ibrutinib/Acalabrutinib/Idelalisib and Venetoclax. (a) Ibrutinib/Acalabrutinib covalently binds to Cys-481 near the ATP-binding domain of BTK, blocking BTK activation and inhibiting the BCR signaling pathway. Inhibition of BTK leads to blockage of activation of downstream signaling molecules such as tour ERK1/2, PI3K, and NF-κB, which inhibits leukemic B cell proliferation and survival. Idelalisib binds to the ATP binding site of PI3Kδ, inhibits the activity of PI3Kδ, blocks the BCR signaling pathway, which in turn leads to the blockage of downstream signaling pathways such as AKT, MAPK, NF-κB, etc., and inhibits the proliferation and survival of leukemic B cells. (b) Venetoclax replaces and releases pro-apoptotic proteins (e.g., BAX, BAK) by directly binding to Bcl-2 proteins, which initiates an apoptotic cascade that leads to the formation of a pore in the outer mitochondrial membrane, the release of cytochromes, and the further activation of caspase-3/7, which leads to the apoptotic death of malignant cells.
Biomedicines 13 01652 g003
Figure 4. The Fab segment of Rituximab/Obinutuzumab specifically binds CD20 antigen, which leads to leukemia cell death through direct cytotoxic effects, antibody-dependent cell-mediated cytotoxicity, antibody-dependent cell phagocytosis, complement-dependent cytotoxicity, and induction of apoptosis. Among them, Obinutuzumab binds CD20 at a wider angle and has stronger direct cytotoxicity. Rituximab binds CD20 with lipid raft formation and strong CDC action, while Obinutuzumab binds CD20 without forming lipid rafts and thus has weaker CDC activity.
Figure 4. The Fab segment of Rituximab/Obinutuzumab specifically binds CD20 antigen, which leads to leukemia cell death through direct cytotoxic effects, antibody-dependent cell-mediated cytotoxicity, antibody-dependent cell phagocytosis, complement-dependent cytotoxicity, and induction of apoptosis. Among them, Obinutuzumab binds CD20 at a wider angle and has stronger direct cytotoxicity. Rituximab binds CD20 with lipid raft formation and strong CDC action, while Obinutuzumab binds CD20 without forming lipid rafts and thus has weaker CDC activity.
Biomedicines 13 01652 g004
Figure 5. Blinatumomab creates an anti-leukemic effect by targeting CD3 and CD19 to establish a link between T cells and tumor B cells, inducing T cells to release granzyme and perforin to target and lyse tumor cells.
Figure 5. Blinatumomab creates an anti-leukemic effect by targeting CD3 and CD19 to establish a link between T cells and tumor B cells, inducing T cells to release granzyme and perforin to target and lyse tumor cells.
Biomedicines 13 01652 g005
Table 1. Representative drugs for targeted therapy.
Table 1. Representative drugs for targeted therapy.
Combination
Therapy Type
Combined DrugsIndicationPatient NumberTreatment OutcomeReference
Tyrosine kinase inhibitorsImatinibPh+ALL, CML110610-year OS rate 83.3%[22]
Dasatinib1495-year OS rate 96%, treatment failure-free survival rate 95%.[16]
Ponatinib5110-year OS rate 90%, 2-year EFS 97%[32]
FLT3 InhibitorsMidostaurinAML22overall response rate was 55.5%, OS 3.7 months.[38]
Gilteritinib24726 patients survived for 2 years or longer without recurrence[41]
B-Cell Signaling Pathway InhibitorsIbrutinibCLL269ORR 92%,
PFS 70%,
OS rate 83%.
[46]
Acalabrutinib13445 months PFS 62%[50]
Idelalisib5481.5% of patients achieved lymph node response during treatment[51]
Anti-apoptotic InhibitorsVenetoclaxCLL and AML---
Immunotherapy DrugsRituximabALL, CLL, HCL2092-year EFS 65%[61]
Obinutuzumab33OS rate 62%, best overall response rate 62%[72]
Blinatumomab405Median OS 7.7 months, CR rate was 34%.[76]
Differentiation InducersATRAAPL---
up coming targeted therapies under trialZiftomenibAML8325% achieved complete remission or complete remission with partial hematologic recovery[82]
NemtabrutinibCLL48OS rate in patients with CLL was 75%.[84]
Table 2. Targeted Drugs + Chemotherapy Drugs.
Table 2. Targeted Drugs + Chemotherapy Drugs.
Combination
Therapy Type
Combined DrugsIndicationPatient NumberTreatment OutcomeReference
TKI + Chemotherapy DrugsImatinib, CVAD (Cyclophosphamide/Vincristine/Doxorubicin/Dexamethasone)Ph+ ALL2685-year OS rate 45.6%, EFS rate 37.1%[14]
Ponatinib, low-intensity chemotherapy regimenPh+ ALL245MRD 34.4%[87]
Ponatinib, Hyper-CVAD (Cyclophosphamide/Vincristine/Doxorubicin/Dexamethasone)Ph+ ALL866-year OS rate 75%[88]
Dasatinib, Hyper-CVAD (Cyclophosphamide/Vincristine/Doxorubicin/Dexamethasone)CML-LBP, Ph+ ALL85CML-LBP and Ph+ ALL 5-year OS rates were 59% and 48%, respectively[92]
Dasatinib, DecitabineCML30Median OS of 13.8 months[93]
Dasatinib, Cytarabine, DaunorubicinCBF-AML613-year Disease-Free Survival and OS 75% and 77% respectively[94]
Anti-apoptotic inhibitors + chemotherapeutic agentsVenetoclax, AzacitidineAML431Median follow-up 20.5 months, OS 14.7 months[86]
Venetoclax, FLAG-IDA (Fludarabine, Cytarabine, Granulocyte Colony-Stimulating Factor, Idarubicin)AML45ORR 98%, MRD 93%, 24-month OS rate 76%, EFS 64%[89]
B-cell signaling pathway inhibitors + chemotherapeutic agentsMidostaurin, Daunorubicin, CytarabineAML717Median OS 74.7 months[90]
Midostaurin, Daunorubicin or IdarubicinAML301CR+CRi 80.7%, of which 65.3% reached CR[91]
Midostaurin, Cytarabine, Daunorubicin or IdarubicinCore binding factor leukemia (CBFL)34Median follow-up 31.5 months, OS rate 73.52%[95]
Midostaurin, intensive chemotherapyAML4402-year OS rate 55%, EFS rate 41%, CR/CRi 74.9%[96]
Gilteritinib, MitoxantroneAMLnoneSignificantly inhibited the proliferation of MV4-11 and MOLM13 cells[35]
Gilteritinib, intensive induction and consolidation chemotherapyAML103Median overall survival time 46.1 months[97]
Immunotherapeutic drugs + chemotherapeutic drugsBlinatumomab, Hyper-CVAD (Cyclophosphamide/Vincristine/Doxorubicin/Dexamethasone)B-ALL383-year OS rate 81%[75]
Rituximab, Fludarabine, CyclophosphamideCLL19244-year OS rate 93.5%[62]
Obinutuzumab, Fludarabine, CyclophosphamideCLL6304-year OS rate in G-FC group reached 90%+[98]
Table 4. Targeted Drugs + CAR-T Combination.
Table 4. Targeted Drugs + CAR-T Combination.
Combination
Therapy Type
Combined DrugsIndicationPatient NumberTreatment Outcome
CAR-T, IbrutinibCLL20At 48 months, OS rate 84%, progression-free survival rate 70%[119]
CAR-T, DasatinibPh+ AML282-year OS and leukemia-free survival rates both 92%[120]
CAR-T, GilteritinibAMLNoneSignificantly enhanced the anti-AML effect of CAR T cells[121]
CAR-T, DuvelisibCLLNoneSignificantly improved the survival rate of CLL mice[122]
Table 5. Multi-drug combinations.
Table 5. Multi-drug combinations.
Combination Therapy TypeCombined DrugsIndicationPatient NumberTreatment Outcome
Ibrutinib, Fludarabine, Cyclophosphamide, ObinutuzumabCLL453-year OS rate 98%[123]
Idelalisib, Ttirabrutinib,
Obinutuzumab
CLL35ORR 93.3%[124]
Zanubrutinib, Venetoclax, ObinutuzumabCLL4218-month OS rate 96.8%, PFS rate 96%[125]
Ibrutinib, Venetoclax,
Obinutuzumab
CLL502 months after end of treatment, CR 28%[126]
Gilteritinib, Azacitidine, VenetoclaxAML5218-month OS rate 72%[127]
Dasatinib, Asciminib,
Prednisone
Ph+ ALL242-year OS rate 75%[128]
Dasatinib, Prednisone,
Blinatumomab
Ph+ ALL243-year OS rate 87%[129]
Ponatinib, Decitabine, VenetoclaxCML201-year and 2-year OS rates 41% and 34% respectively[130]
Acalabrutinib, Venetoclax,
Obinutuzumab
CLL86736-month OS rate 87.7%[108]
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

Zhong, Z.; Yao, R.; Duan, Y.; Ouyang, C.; Du, Z.; Li, L.; Zou, H.; Liu, Y.; Xue, H.; Li, L.; et al. Advances and Challenges in Targeted Therapy and Its Combination Strategies for Leukemia. Biomedicines 2025, 13, 1652. https://doi.org/10.3390/biomedicines13071652

AMA Style

Zhong Z, Yao R, Duan Y, Ouyang C, Du Z, Li L, Zou H, Liu Y, Xue H, Li L, et al. Advances and Challenges in Targeted Therapy and Its Combination Strategies for Leukemia. Biomedicines. 2025; 13(7):1652. https://doi.org/10.3390/biomedicines13071652

Chicago/Turabian Style

Zhong, Zhiyuan, Ran Yao, Yifei Duan, Cheng Ouyang, Zefan Du, Lindi Li, Hailin Zou, Yong Liu, Hongman Xue, Liang Li, and et al. 2025. "Advances and Challenges in Targeted Therapy and Its Combination Strategies for Leukemia" Biomedicines 13, no. 7: 1652. https://doi.org/10.3390/biomedicines13071652

APA Style

Zhong, Z., Yao, R., Duan, Y., Ouyang, C., Du, Z., Li, L., Zou, H., Liu, Y., Xue, H., Li, L., & Chen, C. (2025). Advances and Challenges in Targeted Therapy and Its Combination Strategies for Leukemia. Biomedicines, 13(7), 1652. https://doi.org/10.3390/biomedicines13071652

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