An Update on Lovotibeglogene Autotemcel (Lovo-cel) Clinical Trials for Sickle Cell Disease (SCD) and Analysis of Early Predictors of Response to Lovo-Cel
Speaker: Stacey Rifkin-Zenenberg, Hackensack University Medical Center, USA
Key Highlights:
Dr. Zenenberg highlighted Lovo-cel—a groundbreaking FDA-approved gene addition therapy for sickle cell disease. The presentation focused on pivotal clinical trials, including HGB-206, HGB-210, and long-term follow-up HGB-307.
Study Overview: As of July 2024 data, key findings from the study included:
- Patient Enrolment & Transplantation: 71 patients underwent mobilization; 58 proceeded to transplantation. Of these, 50 met hemoglobin (Hb) response evaluation, and 38 had complete resolution of vaso-occlusive events (VOEs).
- Follow-Up & Demographics: Median follow-up was 4 years, with a maximum of 6.6 years. 16 participants were under 18 years old.
- Mobilization Success: 83% of participants required just 1–2 mobilization cycles to reach the target CD34+ cell dose.
Key Findings:
Lovo-cel Demonstrates Durable Efficacy and Transformative Clinical Benefit:
- 90% of participants achieved globin response (composite endpoint: Hb levels and T87Q expression).
- 87% of participants achieved VOE complete resolution (VOE-CR).
- 95% of participants achieved severe VOE complete resolution (SVOE-CR).
- Pediatric patients had exceptional results, with 100% achieving VOE-CR.
T87Q Expression Predicts Clinical Outcomes:
- T87Q, the functional gene product from lovo-cel, showed strong correlation with VOE-CR, SVOE-CR, and globin response.
- Patients with ≥30% peripheral blood T87Q levels at 6 months post-infusion were more likely to achieve VOE-CR.
Drug Product Characteristics Strongly Predict T87Q Expression:
- A random forest model identified lentiviral vector (LVV) transduction efficiency as the strongest predictor of T87Q levels:
- LVV ≥60% in drug product corresponds to ≥30% T87Q in peripheral blood at 6 months.
Safety Profile: The safety profile was consistent with prior findings linked to sickle cell disease and myeloablative busulfan conditioning.
Durable Clinical Benefits with Exagamglogene Autotemcel for Transfusion-Dependent β-Thalassemia
Speaker: Franco Locatelli, Bambino Gesù Children's Hospital, Rome
Key Highlights:
Exa-cel is the first ex vivo CRISPR-Cas9 gene-editing therapy approved for patients aged ≥12 years with transfusion-dependent β-thalassemia (TDT) and sickle cell disease (SCD). It targets and edits the BCL11A erythroid-specific enhancer to reactivate fetal hemoglobin (HbF) synthesis, reducing disease burden.
Study Design and Population: CLIMB-111 (TDT) and CLIMB-121 (SCD) with long-term follow-up in CLIMB-131.
- Eligibility: TDT: ≥100 mL/kg/year RBC transfusions or ≥10 units annually for 2 consecutive years. Age: 12–35 years; 1/3 adults and 2/3 adolescents.
- Endpoints:
- Primary: Transfusion independence (TDT); freedom from severe VOEs (SCD).
- Secondary: Duration of TI, HbF levels, and safety parameters.
Key Efficacy Results:
- High Rate of Transfusion Independence (TI) in TDT:
- 98% (53/54) of patients achieved transfusion independence (primary endpoint).
- The remaining patient was transfusion-free, with an Hb of 10 g/dL, suggesting delayed benefit.
- Sustained HbF production reached a plateau at 9 months post-infusion and remained stable long-term, ensuring durability of clinical benefit.
- SCD Outcomes:
- In the CLIMB-121 study, Exa-cel led to 93% of patients achieving freedom from severe VOEs for ≥12 months (primary endpoint).
- Chronic pain episodes post-therapy primarily occurred in adults with a prior history of pain and were unrelated to HbF levels.
- Iron Overload Reduction:
- Significant improvements in iron overload parameters:
- Decreased serum ferritin levels and liver iron concentration over time.
- 50% of evaluable patients discontinued iron chelation therapy following sustained transfusion independence.
Safety Profile:
- Adverse events (AEs) were consistent with those seen following busulfan-based myeloablative conditioning.
- A fatal VOD event occurred in a pediatric patient, underscoring the importance of therapeutic drug monitoring (TDM) to avoid overexposure.
Conclusion
Exa-cel provides durable and transformative benefits for patients with TDT and SCD, achieving high rates of transfusion independence and VOE-free status. HbF levels and edited alleles remained stable long-term, ensuring sustained efficacy.
Initial Results from the BEACON Clinical Study: A Phase 1/2 Study Evaluating the Safety and Efficacy of a Single Dose of Autologous CD34+ Base Edited Hematopoietic Stem Cells (BEAM-101) in Patients with Sickle Cell Disease with Severe Vaso-Occlusive Crises
Speaker: Matthew Heeney, Boston Children's Cancer and Blood Disorders Centre, USA
Key Highlights:
Overview of BEAM-101
BEAM-101 uses adenine base editing to introduce a hereditary persistence of fetal hemoglobin polymorphism by targeting the BCL11A erythroid binding site. This approach reactivates HbF without causing double-strand breaks, reducing sickling, hemolysis, and vaso-occlusive crises (VOCs).
BEACON Trial Design & Initial Results:
The Phase 1/2 BEACON study evaluates the safety and efficacy of BEAM-101 in patients (ages 18–35) with severe sickle cell disease and ≥4 VOCs in the prior 24 months.
Key Findings:
- Participants: 7 patients (ages 19–27); 6 with homozygous SS disease.
- Engraftment & Mobilization: Target CD34+ dose achieved with 1–2 mobilization cycles. Neutrophil engraftment by Day 21, platelet engraftment by Day 34, with minimal neutropenic days (mean 6.3).
- HbF Levels: >60% HbF by 1 month post-treatment, sustained throughout follow-up.
- HbS Reduction: Levels dropped to <40%, with anemia resolved in all but one case.
- Safety: No treatment-related Grade ≥3 adverse events or VOCs; 1 fatality linked to busulfan, not BEAM-101.
Clinical Implications
- Safety Profile: BEAM-101 has a favourable safety outcome with rapid engraftment and sustained HbF induction.
- HbF Impact: Protective HbF levels minimized sickling risk and improved hemolysis markers.
- Efficient Mobilization: BEAM-101's base editing mechanism enables faster engraftment compared to traditional nuclease-based therapies.
Conclusion:
- Patients treated with BEAM 101 required a low number of mobilization cycles to achieve target dosing.
- Achieved rapid neutrophil and platelet engraftment with a low number of neutropenic days.
- Initial safety data align with busulfan conditioning and autologous hematopoietic stem cell transplantation, with no VOE post-engraftment.
- All patients experienced a rapid and sustained increase in total and HbF, maintaining levels above protective thresholds during follow-up.
- Sickle Hb levels decreased rapidly, and markers of haemolysis were normalized in all patients.
Prime Editing Enables Precise and Efficient Single Amino Acid Substitutions to Shield CD34+ Hematopoietic Stem Cells from Anti-CD117 Antibody-Based Conditioning
Speaker: Jack Heath, Prime Medicine
Key Highlights:
Prime Editing Technology:
Prime editing is a CRISPR-Cas9-based gene-editing tool enabling precise “search-and-replace” modifications of single DNA strands. Unlike other gene-editing methods, it does not create double-strand breaks (reducing risks of off-target effects and cell stress).
Objective:
- Develop prime editors to introduce a shielding mutation into the KIT gene encoding CD117 to protect CD34+ hematopoietic stem cells (HSCs) from targeted anti-CD117 antibody conditioning.
- Enable selective depletion of unshielded HSCs, while shielded cells are preserved and can repopulate the bone marrow.
Rationale for Selective Conditioning:
Traditional myeloablative conditioning (chemo/radiation) carries significant toxicity. CD117 antibody-based conditioning offers a targeted, less toxic approach by depleting HSCs. Shielded, prime-edited CD34+ cells resist depletion, allowing safer conditioning and potential for improved engraftment.
Applications:
- Allogeneic Transplants: Boost donor cell engraftment by selectively clearing the bone marrow niche.
- Autologous Transplants: Combine shielding edits with therapeutic edits to correct genetic diseases and selectively expand corrected cells post-infusion.
Key Results
- Ex Vivo Studies:
- Shielding edits successfully protected primitive HSC progenitors from anti-CD117 depletion.
- Prime editing maintained cell viability and functional potency.
- In Vivo Studies:
- Shielded CD34+ cells showed robust, durable engraftment in immunodeficient mice.
- 100% shielding efficiency was observed with 80% biallelic edits.
Safety: No off-target effects or chromosomal rearrangements detected.
Clinical Implications:
- Improved Safety: Selective anti-CD117 antibody conditioning reduces risks and toxicity.
- Enhanced Engraftment: Shielded cells can be enriched post-infusion, improving chimerism and long-term success.
- Multiplex Prime Editing: Combines shielding mutations with therapeutic edits, offering a dual approach to correct genetic diseases and enhance transplantation outcomes.
In Vivo HSC Gene Editing for Correction of the Sickle Cell Mutation Using RNA Gene Writers
Speaker: Lorenzo Tozzi, Associate Director, Tessera Therapeutics
Key Highlights:
RNA Gene Writing Technology:
- Inspired by mobile genetic elements, RNA gene writers use target-primed reverse transcription for efficient genomic modifications.
- Enable precise single-nucleotide substitutions without inducing double-strand breaks, reducing safety concerns linked to traditional gene-editing methods.
- Capable of small insertions, deletions, or large gene modifications.
In Vivo Gene Editing via Lipid Nanoparticles (LNPs):
- LNPs facilitate in vivo delivery of RNA gene writers, targeting hematopoietic stem cells (HSCs) directly.
- Overcomes the challenges of traditional gene therapy, such as:
- High costs.
- Limited accessibility.
- Toxic conditioning.
Successful Correction of the Sickle Cell Mutation: RNA gene writers corrected the beta-globin gene mutation in CD34+ cells from SCD patients.
- Efficient correction led to restored normal Hb levels.
- Reversal of sickling under hypoxic conditions in red blood cells.
- Edited cells expanded preferentially during erythroid differentiation.
Long-Term Stability of Gene Edits: Tested in healthy donor CD34+ cells to determine the impact on self-renewal.
- 60% biallelic editing persisted in hematopoietic progenitors 12 weeks post-transplant in immunocompromised mice.
- Edits remained stable across hematopoietic lineages.
Successful Targeting in Non-Human Primates (NHPs):
- Optimized LNP delivery achieved ~74% editing of the beta-2 microglobulin gene.
- Stable editing was maintained for 84 days post-treatment.
- Edited cells contributed to multi-lineage hematopoiesis.
- Beta-globin gene editing in NHPs showed 24% editing efficiency after two LNP doses.
- Secondary transplantation confirmed long-term engraftment and stable edits.
Clinical Implications:
- Reduced Ex Vivo Manipulation: LNPs eliminate the need for toxic conditioning or ex vivo editing, improving gene therapy's safety and accessibility.
- Durable Edits: Long-term stability in mouse and NHP models indicates sustained therapeutic effects.
- Optimized Delivery with LNPs: Promising results with proprietary LNPs show minimal off-target effects and efficient targeting.
ASH Annual Meeting and Exposition, 7-10 December 2024, San Diego, California