0

Acute lymphocytic leukemia (full) treatment: a scientific overview

Acute lymphocytic leukemia (ALL) is a rapidly developing malignant tumor of the hematologic system characterized by the proliferation of unprepared immature lymphoid progenitor cells in the bone marrow and peripheral blood, called lymphoblasts. All of these can come from B-cell or T-cell lineages, whereas B-all lineages are major subtypes. This malignant tumor represents the most common cancer in children, but also affects adults, and the results are relatively poor in adults. Over the past few decades, the therapeutic landscape for all has developed significantly over the past few decades, especially in pediatric populations, especially survival rates.

This article introduces all current therapeutic paradigms, molecularly targeted therapies, and emerging immunotherapeutic strategies, especially bispecific antibodies, aimed at the field of comprehensive hematology of scientists, clinicians and researchers.


1. All pathophysiology and classifications

Everything is cloned malignant tumor caused by the malignant transformation of lymphoid cells and cannot be distinguished normally. The accumulation of lymphocytes can impair normal hematopoiesis, leading to bone marrow failure, anemia, thrombocytopenia, and immunosuppression. Molecular classification is based on lineage (B- or T cells) and genetic abnormalities (e.g.::

  • Philadelphia Chromosomes (pH+) Allthe BCR-ABL1 fusion gene generated by the NCI case table is characterized by t(9;22)(q34;q11)

  • MLL rearrangement

  • ETV6-RUNX1 Fusion

  • High diploid and low diploid

  • Like pHwith changes in kinase activation

These molecular characteristics not only guide prognosis, but also can be more and more direct personalized therapies.


2. Treatment Overview: Principles and Stages

The overall goal of all treatments is to eliminate the leukemia explosion to achieve complete remission (CR), prevent recurrence and ensure long-term disease-free survival. Due to the rapid proliferation of lymphocytes, treatment must be initiated immediately after diagnosis.

Treatment is divided into several stages:

2.1 Induction therapy

The main purpose of induction is to reduce the burden of leukemia to undetectable levels (

  • for b-cell allstandard summary usually includes:

    • Cristine: Microtubule inhibitors disrupt mitosis

    • Corticosteroids (prednisone or dexamethasone): Lymphotoxic effects and anti-inflammatory properties

    • Anthracyclines (daunorubicin or doxorubicin): Insert DNA to inhibit topoisomerase II

    • Aspartase: Depletion of extracellular asparagine is essential for leukemia explosion

  • for pH+allinduce merger Tyrosine kinase inhibitor (TKI) For example Imatinib or Dasatinib Approval for BCR-ABL1 FDA imatinib.

The patient was hospitalized due to tumor lysis syndrome, severe cytoplasm and risk of infection.

2.2 Combination/Intensive Therapy

Post-emission consolidation is designed to eliminate minimal residual disease (MRD) and prevent recurrence by managing high doses or alternative chemotherapy combinations. The duration is variable, but usually involves multiple cycles.

MRD detection by flow cytometry or PCR guidelines – Patients with detectable MRD have poor prognosis and may receive enhanced treatment or considerations for allogeneic hematopoietic stem cell transplantation (Allo-HSCT).

2.3 Maintenance Therapy

Maintenance treatment is extended for 2-3 years and is usually composed of daily oral administration 6-mercaptopurine and weekly Methotrexateperiodic pulses of vincristine and corticosteroids to maintain remission.

Maintenance is essential to prevent recurrence and is often clinic-based.

2.4 Central nervous system (CNS) prevention

The central nervous system is a common shelter for everyone. Preventive intrathecal chemotherapy (methotrexate, cell chain shore) is usually performed at all treatment stages. High doses of systemic methotrexate can also be used. Cranial radiation has now been reserved for patients with CNS participation or at high risk of recurrence to reduce neurotoxicity NCCN guidelines on central nervous system prevention.


3. Hematopoietic stem cell transplantation

Allogeneic stem cell transplantation (Allo-HSCT) was considered in high-risk patients, including:

  • Continuous MRD after merger

  • Undesirable Cell Genetics (e.g., MLL rearrangement)

  • Early or multiple recurrences

The conditioning regimen may be medulla or reduced intensity. Transplantation provides the potential for grafts to function with leukemia, but presents risks with clinical news on HSCT for treatment-related diseases and treatment-related mortality ASH.


4. All target therapies

Targeted agents have revolutionized management for everyone, especially in refractory or recurrence settings.

4.1 Tyrosine kinase inhibitor (TKI)

TKIS targeting BCR-ABL1 (e.g., imatinib, dasatinib, ponatinib) is critical for pH+, and improves survival when used in combination with chemotherapy on TKIS or NIH clinical trials.

4.2 Inotuzumab Ozogamicin

The antibody-drug conjugate targets CD22, expressed in all explosions expressed on B cells, selectively delivering the cytotoxic payload (Calicheamicin) to leukemia cells. It shows efficacy in the relapse/refractory B-ALL FDA ionotuzumab ozogamicin tag.


5. Immunotherapy: Automotive T cells and bispecific antibodies

Immunotherapy is changing all treatments by leukemia cells using the patient’s immune system.

5.1 Chimeric antigen receptor (CAR) T cell therapy

CAR T cells are genetically designed to express receptors targeting CD19 on B-all Blasts, redirecting T cells to identify and kill leukemia. FDA-approved automotive T therapy tisagenlecleucel Significant efficacy was shown in the approval of relapsed/refractory B-ALL FDA TISAGENLECLEUCEL.

5.2 Bispecific antibodies

Bispecific antibodies (BSAB) represent a novel immunotherapy category that binds two different antigens simultaneously, usually one on leukemia explosion and one on immune effector cells. This dual targeting promotes the formation of cell lytic synapses, resulting in effective tumor cell killing.

The prototype BSAB in all treatments is Blinatumomab (blincyto):

  • mechanism: Blinatumomab is a bispecific T cell participant (occlusion) antibody that binds CD19 on B cell explosions and CD3 on cytotoxic T lymphocytes (CTLS). By physically bridging T cells and tumor cells, it activates T cells independently of the major histocompatibility complex (MHC) recognition, resulting in apoptosis of target cells FDA Blinatumomab marker.

  • Clinical uses: Blinatumomab is FDA approved for MRD-positive all and relapsed/refractory B-ALL. It can be completely remission in a large percentage of patients, including those at risk of adverse conditions.

  • advantage: Blinatumomab provides a non-chemotherapy option that utilizes endogenous immune effectors and serves as a bridge to Allo-HSCT.

  • limit: Toxicities such as cytokine release syndrome (CRS) and neurotoxicity require hospitalization monitoring, especially during the initial cycle.

  • Ongoing clinical trials: Trials are investigating a combination of chemotherapy or other immunotherapy to enhance efficacy and reduce recurrent clinical practice.

Other bispecific antibodies under investigation include antibodies directed against CD22 or CD20 that can address antigen escapes seen using CD19-targeted therapy.


6. Relapse and refractory: Challenges and innovation

Despite the progress, 20-30% of adults in all cases have relapses and remains the main cause of treatment failure. All relapses are more resistant and usually require a salvage regimen:

  • Alternative chemotherapy regimen

  • blinatumomab or inotuzumab

  • Automotive T cell therapy

  • Novel drugs under clinical research (e.g., Mening inhibitors targeting KMT2A rearrangement) are clinically investigated.

Integration adaptability of MRD monitoring guidelines.


7. Support care and toxicity management

Optimal management includes vigilant supportive care to alleviate chemotherapy-induced myelosuppression, infection, tumor lysis syndrome and organ toxicity. This includes antibacterial prevention, transfusion support, growth factor and symptom management.


8. Long-term results and survivors

Now, the treatment rate for children is over 80%, which is a victory for multidisciplinary therapy. The results in adults are still not that good, but have improved through adapting to risks.

Long-term survivors need to monitor late effects, including:

Survival plans that address these issues are crucial for the leukemia and lymphoma association survival information.


9. Future perspectives of all therapies

Ongoing research aims to refine molecular diagnosis, deepen understanding of leukemia biology, and develop next-generation therapies, including:

  • Multi-antigen-targeted bispecific antibodies

  • Enhanced automotive T-cell platform, with reduced durability and toxicity

  • Small molecule inhibitors targeting epigenetic and signaling pathways

  • Immune Checkpoint Blockade Select all subtypes

  • Single-cell multimode characteristics characterize leukemia heterogeneity and drug resistance

  • Microbiome modulation and tumor microenvironment targeting

These advances are expected to further improve cure rates and quality of life.


All treatments embody a complex multi-application that integrates intensive chemotherapy, targeted therapy, targeted drugs, immunotherapy including bispecific antibodies and stem cell transplantation. Accurate drugs driven by molecular analysis and MRD evaluation can enable risk adaptation therapy for individual patients’ biology.

Bispecific antibodies, such as gibberellumab, represent milestones in immunodirected therapy, revolutionizing treatments, especially for recurrent or MRD-positive diseases. The evolving therapeutic landscape is expected to continue to improve driven by translational research and innovative clinical trials, thus bringing hope for treatment to all patient populations.


References and further reading: