All content on this site is intended for healthcare professionals only. By acknowledging this message and accessing the information on this website you are confirming that you are a Healthcare Professional. If you are a patient or carer, please visit Know ALL.

The ALL Hub uses cookies on this website. They help us give you the best online experience. By continuing to use our website without changing your cookie settings, you agree to our use of cookies in accordance with our updated Cookie Policy

Introducing

Now you can personalise
your ALL Hub experience!

Bookmark content to read later

Select your specific areas of interest

View content recommended for you

Find out more
  TRANSLATE

The ALL Hub website uses a third-party service provided by Google that dynamically translates web content. Translations are machine generated, so may not be an exact or complete translation, and the ALL Hub cannot guarantee the accuracy of translated content. The ALL Hub and its employees will not be liable for any direct, indirect, or consequential damages (even if foreseeable) resulting from use of the Google Translate feature. For further support with Google Translate, visit Google Translate Help.

Steering CommitteeAbout UsNewsletterContact
LOADING
You're logged in! Click here any time to manage your account or log out.
LOADING
You're logged in! Click here any time to manage your account or log out.
2024-04-03T09:42:04.000Z

Optimizing CAR T-cell therapy for adult patients with R/R B-ALL

Apr 3, 2024
Share:
Learning objective: After reading this article, learners will be able to cite a new clinical development in ALL.

Bookmark this article

Chimeric antigen receptor (CAR) T-cell therapies can significantly improve outcomes for patients with relapsed/refractory (R/R) B-cell acute lymphoblastic leukemia (B-ALL).1 Two CAR T-cell therapies, tisagenleucel and brexucabragene autoleucel, are approved by the U.S. Food and Drug Administration (FDA) for the treatment of this patient population1; however, while CAR T-cell therapy is associated with high response rates in B-ALL, remission durability can be limited.1 The burden of CAR T-cell toxicity can further limit the use of these therapies.1

Here we summarize a review published by Agrawal et al.1 in European Journal of Hematology discussing factors associated with CAR T-cell therapy outcomes in adult patients with R/R B-ALL and strategies to optimize the use of this therapy.

Clinical data1

Several clinical studies have demonstrated the efficacy of CD19-directed CAR T-cell therapies in patients with R/R B-ALL, including the ELIANA, ZUMA-3, and FELIX trials.

Factors influencing CAR T-cell therapy response1

There are several patient and disease factors influencing CAR T-cell activity that can be used to inform strategies for optimization.

  • The type of co-stimulatory signal domain used may impact response duration.
    • CD28 co-stimulation is associated with rapid expansion but a lack of persistence vs 4-1BB co-stimulation.
    • 4-1BB co-stimulation may be preferred in patients when no allogeneic hematopoietic stem cell transplantation (allo-HSCT) consolidation is planned.
  • Clinical studies have indicated that T-cell immunophenotype may impact CAR T-cell persistence, expansion, and long-term activity; further investigation could improve CAR T-cell outcomes.
  • Prior CD19-directed salvage therapy may affect outcomes after subsequent CD19-directed CAR T-cell therapy, and response to prior therapy could identify patients who are resistant to CD19-directed therapies.
    • Immediate blinatumomab salvage therapy is not recommended for patients proceeding to CAR-T cell therapy.
  • Several studies have suggested that high disease burden at lymphodepletion reduces CAR T-cell response and remission durability.
  • While the impact of high-risk genetics on CAR T-cell therapy response overall is unclear, KMT2A rearrangements are associated with a higher incidence of myeloid lineage switch, patients with TP53 mutations have lower complete remission rates and inferior outcomes, and genetic alterations affecting IKZF1 are associated with poor response.

Consolidation1

  • Early loss of B-cell aplasia and measurable residual disease detected by next-generation sequencing can identify patients with increased risk of relapse following CAR T-cell therapy.
  • Compared with pediatric patients, the durability of response to CAR T-cell therapy for adult patients is poor, particularly when not consolidated with allo-HSCT.
    • Patients with a high risk of relapse should be considered for allo-HSCT.
  • CAR-T cell therapy is typically the preferred salvage option for patients experiencing relapse after allo-HSCT.
    • Effective bridging to CAR T-cell therapy and vigilant post-CAR T-cell surveillance is necessary to achieve durable responses; second allo-HSCT can be considered for fit patients with a high risk of relapse.

Toxicity1

CAR T-cell toxicities include off-tumor effects, such as B-cell aplasia and hypogammaglobulinemia; myelosuppression; and immune-mediated effects, such as cytokine release syndrome and immune effector cell-associated neurotoxicity syndrome.

  • Disease burden at lymphodepletion is predictive of the risk of severe cytokine release syndrome and immune effector cell-associated neurotoxicity syndrome.
  • Studies are ongoing to determine the potential of tocilizumab used earlier in treatment,  steroids to manage immune-mediated effects, and whether fractioned dosing of CAR T-cells can reduce toxicity while maintaining response rates.
  • The CD19-directed antibody’s affinity may also affect the toxicity profile.

Relapse post CAR T-cell therapy1

  • Relapse following CD19-direct CAR T-cell therapy can be categorized as either CD19-positive relapse (7080% of cases) or CD19-negative relapse (2030% of cases).
  • CD19-positive relapse is associated with a loss of CAR T-cell persistence and decreased CD19 antigen density.
    • Several studies have investigated strategies to optimize the CAR T-cell construct to reduce the risk of CD19-positive relapse.
  • CD19-negative relapse occurs due to leukemic cells evading CAR T-cell therapies and is associated with a worse prognosis vs CD19-positive relapse.
    • Other leukemic targets, such as the CD22 antigen, and the targeting of multiple antigens have been investigated to overcome relapse.

Challenges associated with access to CAR T-cell therapies1

  • Barriers to access, such as insurance authorization and transferring to CAR-specialized centers, can delay therapy.
  • Manufacturing times for CAR T-cell therapies (estimated 12–20 days) can result in high dropout rates due to progression and complications.
    • Off-the-shelf allogenic CAR platforms and rapid CAR manufacturing are being explored to combat dropout rates.
Key learnings
  • Risk factors found to be associated with CAR T-cell therapy outcomes include disease burden, leukemia cytogenetics, costimulatory domain, early referral to CAR T-cell center, and long manufacturing times. 
  • Several strategies are currently being investigated to improve CAR T-cell outcomes, including consolidation with allo-HSCT to prevent relapse in high-risk patients, multitargeted CARs to overcome immune escape, and the development of new CAR designs to improve persistence.

  1. Agrawal V, Murphy L, Pourhassan H, et al. Optimizing CAR-T cell therapy in adults with B-cell acute lymphoblastic leukemia. Eur J Hematol. 2024 112(2):236-247. DOI:1111/ejh.14109

Related articles

Newsletter

Subscribe to get the best content related to ALL delivered to your inbox