MedPath

ALLO-501 Advanced Drug Monograph

Published:May 12, 2025

Generic Name

ALLO-501

ALLO-501: An Investigational Allogeneic Anti-CD19 CAR T-Cell Therapy - A Comprehensive Review

1. Introduction to ALLO-501

Chimeric Antigen Receptor (CAR) T-cell therapy has emerged as a transformative treatment modality for various hematologic malignancies. However, the widespread application of first-generation autologous CAR T-cell therapies, which are manufactured from a patient's own T-cells, is often constrained by complex manufacturing logistics, significant treatment delays, product variability, and challenges in obtaining sufficient and functional T-cells from heavily pretreated patients.[1] To address these limitations, the field has actively pursued the development of allogeneic, or "off-the-shelf," CAR T-cell therapies derived from healthy donor T-cells. ALLO-501 is one such investigational allogeneic CAR T-cell therapy, engineered to target the CD19 antigen, a protein broadly expressed on the surface of B-cells and B-cell malignancies.[1]

ALLO-501 was developed by Allogene Therapeutics as part of a strategic collaboration with Servier. This collaboration is founded upon an exclusive license granted by Cellectis to Servier, leveraging Cellectis' pioneering TALEN® (Transcription Activator-Like Effector Nuclease) gene-editing technology.[3] Under this agreement, Servier grants Allogene exclusive rights for the development and commercialization of ALLO-501 in the United States.[7] The core premise of ALLO-501 is to provide a readily available CAR T-cell product, thereby improving patient access, enabling timely intervention, reducing product variability, and potentially allowing for repeat dosing if clinically indicated.[1] A significant practical advantage observed in early clinical studies was the ability to initiate treatment rapidly, with a median time of just two days from patient enrollment to the commencement of the study treatment regimen.[1]

It is crucial to understand that ALLO-501 served as a foundational product in Allogene's anti-CD19 AlloCAR T™ program and is the direct predecessor to cemacabtagene ansegedleucel (cema-cel), also designated as ALLO-501A.[1] While both products share the same core CAR design targeting CD19, a key molecular distinction exists: ALLO-501 incorporates a rituximab recognition domain within its CAR construct, whereas this domain has been eliminated in cema-cel (ALLO-501A).[1] This modification was a deliberate design evolution. Rituximab, an anti-CD20 monoclonal antibody, is a standard component of treatment regimens for many B-cell lymphomas. The presence of a rituximab recognition site on ALLO-501 could lead to its premature clearance by rituximab or limit its utility in patients who have recently received rituximab-containing therapies. By removing this domain, cema-cel (ALLO-501A) was engineered for broader patient applicability and has become the lead candidate for pivotal clinical studies, particularly those exploring earlier lines of therapy.[5]

The development of ALLO-501 and its successor, cema-cel, signifies more than an incremental improvement in manufacturing convenience; it embodies a potential paradigm shift in the delivery and accessibility of cellular immunotherapies. Autologous CAR T-cell therapies, despite their proven efficacy, are beset by individualized manufacturing processes that are not only time-consuming (often taking several weeks) but are also susceptible to manufacturing failures, especially when patient-derived T-cells are compromised by prior treatments or disease burden. These logistical hurdles significantly limit the number of patients who can benefit and often restrict treatment to highly specialized academic medical centers.[1] The allogeneic nature of ALLO-501, allowing for the creation of large batches of cryopreserved, "off-the-shelf" doses from healthy donor cells [1], directly confronts these critical bottlenecks. This approach has the potential to democratize access to CAR T-cell therapy, making it available to a larger patient population more expeditiously and potentially extending its reach into community oncology settings where the majority of cancer patients receive care.[9] Although the clinical program has largely transitioned to cema-cel, the initial development and clinical data generated with ALLO-501 were instrumental in establishing the foundational proof-of-concept for this specific allogeneic platform, providing critical insights into safety, efficacy, and the optimal lymphodepletion strategy that have informed the subsequent development of cema-cel.

2. Mechanism of Action and Molecular Design

The therapeutic strategy of ALLO-501 is centered on its ability to specifically recognize and eliminate CD19-expressing B-cell malignancies through a meticulously engineered allogeneic CAR T-cell product.

Target Antigen and Cytotoxic Action:

ALLO-501 CAR T-cells are designed to target the CD19 antigen. CD19 is a transmembrane protein consistently expressed across the spectrum of B-cell development and is present on the vast majority of B-cell malignancies, including prevalent Non-Hodgkin Lymphoma (NHL) subtypes such as Diffuse Large B-cell Lymphoma (DLBCL) and Follicular Lymphoma (FL).1 The CAR construct on ALLO-501 enables these engineered T-cells to bind with high affinity to CD19 on tumor cells. This binding event activates the CAR T-cell, initiating a signaling cascade that leads to a potent cytotoxic T-lymphocyte (CTL) response, ultimately resulting in the targeted lysis and elimination of the malignant B-cells.11

Allogeneic Platform and Gene-Editing Foundation:

A cornerstone of the ALLO-501 platform is its allogeneic nature; the T-cells are sourced from healthy, screened donors rather than the patient undergoing treatment.1 This necessitates sophisticated genetic engineering to overcome the immunological barriers inherent in allogeneic cell transplantation, primarily the risks of Graft-versus-Host Disease (GvHD) and host-mediated rejection of the CAR T-cells. ALLO-501 employs the TALEN® gene-editing technology, developed by Cellectis, to make precise modifications to the donor T-cell genome.3 Two critical gene edits are performed:

  1. TCRα Constant (TRAC) Gene Disruption: The gene encoding the T-cell receptor alpha constant region (TRAC) is deliberately disrupted. The TCR is responsible for recognizing antigens presented by Major Histocompatibility Complex (MHC) molecules, and in an allogeneic setting, the donor T-cell's native TCR can recognize the recipient's tissues as foreign, leading to GvHD. By knocking out the TRAC gene, TCR expression on the surface of ALLO-501 cells is eliminated. This abrogation of native TCR function is a fundamental strategy to mitigate the risk of GvHD, a potentially severe and life-threatening complication.[1] The absence of GvHD reported in clinical trials involving ALLO-501 and cema-cel provides clinical validation for the effectiveness of this gene edit.[1] Furthermore, elimination of the native TCR may also contribute to more uniform CAR expression and potentially enhance the overall potency of the CAR T-cells.[11]
  2. CD52 Gene Disruption: The gene encoding CD52, a protein widely expressed on the surface of mature lymphocytes (including T-cells, B-cells, monocytes, and macrophages), is also knocked out in ALLO-501 cells.[1] The rationale for this modification is intrinsically linked to the lymphodepletion strategy employed prior to CAR T-cell infusion. By rendering the ALLO-501 cells CD52-negative, they become resistant to the effects of anti-CD52 monoclonal antibodies, such as ALLO-647, which is co-administered as part of the lymphodepletion regimen. ALLO-647 can then selectively deplete the patient's own CD52-positive host lymphocytes. This "selective lymphodepletion" is designed to reduce the host immune response against the allogeneic CAR T-cells (i.e., prevent rejection) and create a more favorable "space" for the infused CAR T-cells to engraft, expand, and persist, thereby maximizing their anti-tumor activity.[1] This integrated approach, where the gene-edited CAR T-cell product is designed to work in concert with a specific lymphodepleting agent, represents a sophisticated multi-component therapeutic system.

Unique Feature of ALLO-501: Rituximab Recognition Domain:

ALLO-501, in its original design, incorporated a rituximab recognition domain within its CAR construct. This feature distinguishes it from its successor, cema-cel (ALLO-501A), from which this domain was intentionally removed.1 The precise rationale for including this domain in ALLO-501 is not extensively detailed in the available information. However, its subsequent removal in ALLO-501A was a strategic decision aimed at enhancing the product's clinical utility by allowing its use in a broader patient population, specifically including those who have had recent exposure to rituximab.5 This iterative modification reflects a responsive and adaptive approach to drug development, refining the product based on anticipated clinical scenarios and potential interactions with standard-of-care therapies.

The molecular architecture of ALLO-501 thus reflects a multifaceted engineering strategy: targeting a validated tumor antigen (CD19), employing an allogeneic cell source for accessibility, and incorporating critical gene edits to ensure safety (GvHD mitigation via TRAC knockout) and efficacy (enhanced persistence via CD52 knockout coupled with ALLO-647-mediated selective lymphodepletion).

3. Manufacturing and "Off-the-Shelf" Potential

A defining characteristic of ALLO-501 and its successor, cema-cel, is their development as "off-the-shelf" allogeneic CAR T-cell therapies, which offers substantial advantages in manufacturing and clinical deployment compared to autologous products.

Allogeneic Source and Scalable Manufacturing:

ALLO-501 is manufactured using T-cells sourced from carefully screened healthy donors, rather than from the individual patient who will receive the therapy.1 This foundational difference enables a centralized and standardized manufacturing process. The process has been described as scalable, with the capability to produce approximately 100 doses of CAR T-cells from a single manufacturing run.1 This scalability contrasts sharply with autologous CAR T-cell production, which necessitates a unique, patient-specific manufacturing campaign for every individual treated. Such large-scale production from healthy donor cells is a critical step towards making CAR T-cell therapy more broadly available.

Cryopreservation and On-Demand Availability:

Once manufactured, doses of ALLO-501 (and cema-cel) are cryopreserved and stored, creating an inventory of ready-to-use therapeutic product.1 This "on-demand" availability is a cornerstone of the "off-the-shelf" concept. It allows clinicians to access CAR T-cells for their patients almost immediately, bypassing the significant delays inherent in autologous CAR T-cell therapy, which include patient scheduling for leukapheresis, shipping of cells to a manufacturing facility, the multi-week cell engineering and expansion process, quality control testing, and shipment back to the treatment center.1

Logistical and Clinical Advantages:

The "off-the-shelf" model offers several profound logistical and clinical advantages:

  • Timeliness of Treatment: Perhaps the most impactful advantage is the ability to initiate treatment rapidly. For patients with aggressive, relapsed/refractory lymphomas, who may have rapidly deteriorating clinical status, the weeks-long waiting period for autologous CAR T-cells can be untenable. The ALLO-501 platform demonstrated a median time of just 2 days from enrollment to the start of study treatment (lymphodepletion followed by CAR T-cell infusion) in the ALPHA/ALPHA2 trials.[1] This speed can be crucial for patients who cannot afford to wait.
  • Overcoming Patient-Specific Manufacturing Hurdles: Autologous CAR T-cell manufacturing can be challenging or impossible for patients who have been heavily pretreated, as their T-cells may be insufficient in number or compromised in function (poor T-cell fitness).[1] Allogeneic products derived from healthy donors circumvent this issue entirely.
  • Reduced Need for Bridging Therapy: The delay in autologous CAR T-cell manufacturing often necessitates "bridging" chemotherapy to control the disease while the cells are being produced. This bridging therapy adds to the patient's treatment burden and may not always be effective.[1] The immediate availability of ALLO-501 reduces or eliminates this need.
  • Product Consistency: Manufacturing from a common cell bank derived from a few healthy donors may lead to a more consistent and well-characterized CAR T-cell product, with less inter-patient variability compared to autologous products, where each batch is unique to the patient.[2]
  • Potential for Broader Access: The simplified logistics and potential for outpatient administration (as envisioned for cema-cel in the ALPHA3 trial [9]) could allow these therapies to be administered in community oncology centers, rather than being restricted to specialized academic institutions. This would significantly expand patient access, particularly for those who live far from major transplant and CAR T centers.[10]

The manufacturing process and "off-the-shelf" availability of ALLO-501 represent a significant step towards overcoming many of the practical limitations of autologous CAR T-cell therapy. While not explicitly detailed in the provided information, the economies of scale associated with producing multiple doses from a single donor and manufacturing run [1], combined with the reduction in patient-specific logistical complexities, could also have favorable economic implications for the healthcare system in the long term. Furthermore, the availability of a standardized, readily accessible product facilitates more complex therapeutic strategies, such as repeat dosing. Indeed, one patient in the ALPHA study who progressed after an initial partial response was successfully re-treated with a second round of lymphodepletion and ALLO-501 infusion, subsequently achieving a complete response.[2] Such a strategy is far more challenging with autologous products, which might require a new apheresis and manufacturing cycle.

4. Clinical Development Program

The clinical development of ALLO-501 and its successor, ALLO-501A (cema-cel), has been primarily pursued through the ALPHA and ALPHA2 trials, focusing on patients with relapsed/refractory (R/R) B-cell Non-Hodgkin Lymphomas (NHL).

Key Clinical Trials:

The foundational clinical investigations for the Allogene anti-CD19 AlloCAR T™ platform include:

  • ALPHA Study (NCT03939026; Primary ID: ALLO-501-201): This is a Phase 1, single-arm, open-label clinical trial designed to evaluate the safety, efficacy, cellular kinetics, and immunogenicity of ALLO-501. The study enrolled adult patients with R/R Large B-cell Lymphoma (LBCL) or Follicular Lymphoma (FL).[4] Participants in the ALPHA study received ALLO-501 following a lymphodepletion regimen consisting of fludarabine, cyclophosphamide, and the anti-CD52 monoclonal antibody ALLO-647.[4] According to available information, this trial is currently listed with a "Not Recruiting" status.[4]
  • ALPHA2 Study (NCT04416984): This Phase 1 trial was designed to evaluate ALLO-501A (cema-cel), the next-generation product candidate, in patients with R/R LBCL.[6] Data from the ALPHA and ALPHA2 studies are frequently pooled and presented together, particularly for the cohort of CAR T-cell treatment-naïve patients with R/R LBCL, to provide a more comprehensive understanding of the platform's performance.[1]

Investigated Indications and Patient Population:

The broad therapeutic area targeted is R/R NHL.3 Within this, the clinical programs have specifically focused on, and reported data for, patients with:

  • Relapsed/Refractory Diffuse Large B-cell Lymphoma (DLBCL), or more broadly, Large B-cell Lymphoma (LBCL).[3]
  • Relapsed/Refractory Follicular Lymphoma (FL).[3]

A significant cohort for efficacy and safety analysis, often highlighted in communications, comprises 33 CD19 CAR T-cell treatment-naïve patients with R/R LBCL. These patients were treated with either ALLO-501 or cema-cel (ALLO-501A) manufactured using the process selected for subsequent pivotal studies, drawing from the combined data of the ALPHA and ALPHA2 trials.[1]

The Critical Role of Lymphodepletion (LD):

A robust lymphodepletion regimen is paramount for the success of any CAR T-cell therapy, as it creates a favorable microenvironment for CAR T-cell engraftment, expansion, and persistence. For allogeneic CAR T-cells like ALLO-501, lymphodepletion is even more critical to mitigate host-versus-graft rejection.

  • Regimen Components: The LD regimen employed in the ALPHA and ALPHA2 trials typically involved a 3-day course of fludarabine (e.g., 30 mg/m² daily) and cyclophosphamide (e.g., 300 or 500 mg/m² daily), administered in conjunction with ALLO-647.[1]
  • ALLO-647 (Anti-CD52 Monoclonal Antibody): ALLO-647 is an investigational antibody that specifically targets the CD52 antigen present on host lymphocytes. Its administration leads to the depletion of these host immune cells.[1] Because ALLO-501 and cema-cel CAR T-cells are genetically engineered to lack CD52 expression (CD52 knockout), they are resistant to ALLO-647. This selective depletion of host CD52-positive lymphocytes by ALLO-647 is designed to prevent the patient's immune system from rejecting the infused allogeneic CAR T-cells, thereby allowing them to expand, persist, and exert their anti-tumor effects.[1] The trials explored escalating cumulative doses of ALLO-647 (e.g., 39 mg, 60 mg, and 90 mg) to optimize this selective lymphodepletion.[1] Clinical data suggested that higher doses of ALLO-647, particularly the 90 mg dose as part of the FCA90 regimen (Fludarabine, Cyclophosphamide, and 90 mg ALLO-647), were associated with deeper and more durable responses, likely due to more effective control of host lymphocyte rejection and better CAR T-cell expansion and persistence.[2] Consequently, the FCA90 regimen was selected as the lymphodepletion strategy for the Phase 2 development of cema-cel.[8]
  • Timing: Lymphodepletion was generally initiated promptly after enrollment, with a median time of 2 days from enrollment to the start of study treatment, followed by the infusion of ALLO-501 or cema-cel.[1] This rapid timeline is a key differentiator from autologous CAR T-cell therapies.

Dosing of ALLO-501/cema-cel:

The Phase 1 studies investigated various CAR T-cell dose levels, including 40 × 10⁶, 120 × 10⁶, and 360 × 10⁶ CAR-positive cells.2 Based on the evaluation of safety and efficacy across these dose levels, a single infusion of 120 × 10⁶ CAR+ cells, administered after the FCA90 lymphodepletion regimen, was chosen as the recommended Phase 2 dose for cema-cel.8 Some study designs also explored the concept of a double infusion of ALLO-501/501A.13

The clinical development program for ALLO-501 and its successor cema-cel underscores a data-driven, iterative approach. The optimization of the lymphodepletion regimen, particularly the dose of ALLO-647, has been a central theme, recognizing that effective management of the host immune environment is crucial for the success of an allogeneic cell therapy. The ability to rapidly initiate this complex therapy offers a significant advantage for patients with aggressive lymphomas, where treatment delays can be detrimental. The Phase 1 ALPHA and ALPHA2 studies have thus served not only to evaluate the safety and preliminary efficacy of the CAR T-cell products themselves but also to meticulously refine the entire therapeutic strategy, including lymphodepletion and CAR T-cell dosing, paving the way for later-stage pivotal trials.

5. Clinical Efficacy

The clinical efficacy of the ALLO-501 platform, encompassing both ALLO-501 and its successor cemacabtagene ansegedleucel (cema-cel or ALLO-501A), has been primarily reported from the Phase 1 ALPHA and ALPHA2 trials. The most consistently presented and mature data focus on a cohort of 33 CD19 CAR T-cell treatment-naïve patients with relapsed/refractory (R/R) Large B-cell Lymphoma (LBCL) who received cells manufactured with the process selected for pivotal studies.[1]

Key Efficacy Outcomes in CAR T-Naïve R/R LBCL (N=33):

The following table summarizes the key efficacy results for this patient cohort, as reported with a data cutoff of September 26, 2024 8:

Table 1: Summary of Efficacy Outcomes for ALLO-501/cema-cel in CD19 CAR T-Naïve Relapsed/Refractory Large B-Cell Lymphoma Patients (ALPHA/ALPHA2 Trials, N=33)

Efficacy EndpointResult (Value)95% Confidence Interval (CI)
Objective Response Rate (ORR)58% (19/33)39.2% to 74.5%
Complete Response (CR) Rate42% (14/33)25.5% to 60.8%
Median Duration of Response (Overall)11.1 months3.1 months to Not Reached
Median Duration of Response (CR pts)23.1 months11.1 months to Not Reached
Median Progression-Free Survival (PFS)3.9 months1.9 months to 11.1 months
Median PFS (CR pts)24.0 months8.1 months to Not Reached

Source: Primarily.[8] Corroborating data for ORR and CR rates also found in.[1]

These data indicate that ALLO-501/cema-cel can induce responses in a significant proportion of heavily pretreated R/R LBCL patients. Notably, the 42% CR rate is a clinically meaningful outcome in this challenging patient population. Earlier data presented at the International Conference on Malignant Lymphoma (ICML) with a cutoff of April 20, 2023, focusing on 12 patients who received the selected Phase 2 regimen (single dose of ALLO-501/501A at 120 × 10⁶ CAR+ cells and FCA90 lymphodepletion), showed a CR rate of 58% (7 out of 12 patients).[13] While this subgroup showed a higher CR rate, the larger cohort of 33 patients provides a broader view of the efficacy with the pivotal manufacturing process.

A critical aspect of the efficacy profile is the durability of responses, particularly for patients who achieve a CR. The median Duration of Response (DOR) for patients achieving a CR was 23.1 months, and their median Progression-Free Survival (PFS) was 24.0 months.[8] These figures are substantial and suggest that if a deep remission is achieved with this allogeneic CAR T-cell therapy, it can be long-lasting, offering a significant clinical benefit comparable to that observed with some autologous CAR T-cell therapies.

However, the median PFS for the overall cohort of 33 patients was 3.9 months.[8] This highlights a dichotomy in outcomes: while complete responders experience prolonged disease control, a considerable number of patients either do not respond or relapse relatively quickly. This underscores the importance of identifying predictive biomarkers to select patients most likely to achieve deep and durable responses. The strategic move to incorporate minimal residual disease (MRD) detection via the Foresight CLARITY™ assay in the ALPHA3 trial for first-line consolidation therapy aims to address this by treating patients when their disease burden is at its lowest, potentially improving outcomes.[7]

Factors Influencing Efficacy:

The intensity and effectiveness of the lymphodepletion regimen, particularly the inclusion and dosing of ALLO-647, have been identified as key factors influencing CAR T-cell expansion, persistence, and ultimately, clinical efficacy.1 Data from earlier stages of the Phase 1 trials suggested that higher doses of ALLO-647 (specifically the 90 mg dose in the FCA90 regimen) were associated with deeper responses.2 This regimen was subsequently selected for the Phase 2 development of cema-cel 8, indicating that robust host lymphocyte depletion is crucial for optimizing the activity of these allogeneic CAR T-cells. The cellular kinetics data confirmed that allogeneic CAR T-cell expansion and persistence were indeed supported by this enhanced lymphodepletion strategy.1

In summary, the clinical efficacy data for the ALLO-501/cema-cel platform in CAR T-naïve R/R LBCL patients demonstrate promising response rates, with a notable proportion of patients achieving durable complete remissions. The success of the therapy appears closely linked to the depth of lymphodepletion achieved, highlighting the importance of managing the host immune environment for allogeneic cell therapies.

6. Safety and Tolerability Profile

The safety and tolerability of ALLO-501 and its successor, cema-cel, have been evaluated in the Phase 1 ALPHA and ALPHA2 clinical trials. The safety profile is generally considered manageable, particularly concerning hallmark CAR T-cell toxicities like Cytokine Release Syndrome (CRS) and Immune Effector Cell–Associated Neurotoxicity Syndrome (ICANS).[1] Data are primarily derived from the cohort of 33 CAR T-naïve R/R LBCL patients [8] and a broader safety dataset from 87 patients with R/R LBCL and follicular lymphoma (FL) treated across both studies.[6]

Table 2: Summary of Key Treatment-Emergent Adverse Events (TEAEs) in CD19 CAR T-Naïve Relapsed/Refractory Large B-Cell Lymphoma Patients Treated with ALLO-501/cema-cel (ALPHA/ALPHA2 Trials, N=33)

Adverse Event CategorySpecific EventAny Grade (%)Grade ≥3 (%)
Cytokine Release Syndrome (CRS)CRS24% (8/33)0%
Immune Effector Cell–Associated Neurotoxicity SyndromeICANS0%0%
Graft-versus-Host DiseaseGvHD0%0%
InfectionsOverall58% (19/33)15% (5/33)
CMV Reactivation30% (10/33)12% (4/33)
Hematologic ToxicitiesNeutropeniaNot specified82% (27/33)
AnemiaNot specified46% (15/33)
ThrombocytopeniaNot specified42% (14/33)
LymphopeniaNot specified33% (11/33)

Source: Primarily.[8] Supportive data for CRS, ICANS, GvHD, and infections from.[1] Note: "Not specified" for any grade hematologic toxicities as the primary source focuses on Grade ≥3.

Key CAR T-Cell Related Toxicities:

  • Cytokine Release Syndrome (CRS): In the cohort of 33 R/R LBCL patients, the incidence of any-grade CRS was 24%, with no cases of Grade ≥3 CRS reported.[1] In the broader safety population of 87 patients from ALPHA/ALPHA2, any-grade CRS occurred in 23% of patients, with only one patient (1%) experiencing a Grade 3 event.[6] This indicates a notably low rate of severe CRS.
  • Immune Effector Cell–Associated Neurotoxicity Syndrome (ICANS): No cases of ICANS of any grade were reported in the 33-patient LBCL cohort.[1] Similarly, in the larger 87-patient cohort, no Grade ≥3 ICANS events were observed.[6] The absence of significant ICANS is a remarkable safety feature.
  • Graft-versus-Host Disease (GvHD): Consistent across all reports, no cases of GvHD were observed.[1] This is a critical safety outcome for an allogeneic T-cell product and provides strong clinical validation for the effectiveness of the TALEN®-mediated TRAC gene knockout in preventing this complication.

Infection Profile:

Infections are an important consideration in heavily lymphodepleted patients.

  • In the 33 R/R LBCL patients, any-grade infections occurred in 58%, and Grade ≥3 infections were reported in 15%.[8] No fatal infections were noted in this specific cohort.
  • In the broader 87-patient safety set, any-grade infections were seen in 58% and Grade ≥3 infections in 21%. Two fatal infectious events (one COVID-19 pneumonia, one pneumonia; 2% of total) were previously reported in this larger group.[6]
  • Cytomegalovirus (CMV) reactivation was the most common infection, occurring in 30% (any grade) and 12% (Grade ≥3) of the 33 LBCL patients.[8] Similar rates (25% any grade, 9% Grade 3) were seen in the 87-patient cohort.[6] Weekly CMV monitoring was standard practice in the trials.[6]
  • Opportunistic infections, other than CMV, were reported as uncommon.[8]

Hematologic Toxicity / Cytopenias:

Prolonged cytopenias are expected following intensive lymphodepletion and CAR T-cell therapy.

  • In the 33 R/R LBCL patients, Grade ≥3 TEAEs included neutropenia (82%), anemia (46%), thrombocytopenia (42%), and lymphopenia (33%).[8]
  • The median time to recovery for absolute neutrophil count (ANC) to Grade ≤3 was 7 days, and for absolute lymphocyte count (ALC) to Grade ≤3 was 17.5 days.[8]
  • Encouragingly, the proportion of patients experiencing ongoing Grade ≥3 cytopenias tended to decrease over time. Data from the larger 87-patient cohort showed overall Grade ≥3 cytopenias at 29% on Study Day 28, reducing to 20% by Day 56, and further to 15% by Month 4.[6]
  • Hypogammaglobulinemia, a known complication of B-cell depleting therapies, was reported in 15% of the 33 LBCL patients.[8]

ALLO-647 Related Safety:

The anti-CD52 antibody ALLO-647, used for lymphodepletion, was generally well-tolerated. Infusion-Related Reactions (IRRs) were typically low grade. In the larger cohort of 87 patients, 6% experienced Grade 3 IRRs, all of which were managed with supportive care measures.6

Other Notable Adverse Events:

In the cohort of 33 R/R LBCL patients, the most common reasons for discontinuation from the study were death (48%, often related to underlying disease progression in this advanced population) and consent withdrawal (12%).8

The safety profile of ALLO-501/cema-cel, particularly the low incidence of severe CRS and the absence of ICANS and GvHD, is highly encouraging. This compares favorably with the toxicity profiles often observed with autologous CD19 CAR T-cell therapies, where severe CRS and ICANS can necessitate intensive care and prolong hospitalization. The ability to effectively prevent GvHD through TRAC gene editing is a cornerstone of the allogeneic platform's viability. However, the rates of infection, especially CMV reactivation, and the occurrence of prolonged cytopenias, while showing trends towards recovery, highlight the need for vigilant monitoring and supportive care. These latter toxicities are largely attributable to the profound and necessary lymphodepletion required for allogeneic cell engraftment and the B-cell aplasia induced by effective anti-CD19 therapy. The favorable safety profile, especially concerning CRS and ICANS, may support outpatient administration in the future, as planned for the ALPHA3 trial [9], which could significantly improve patient convenience and reduce healthcare resource utilization.

7. Regulatory Status and Developmental Trajectory

The regulatory journey and developmental path of ALLO-501 and its successor, cemacabtagene ansegedleucel (cema-cel or ALLO-501A), reflect a strategic evolution aimed at optimizing the therapeutic potential of Allogene Therapeutics' anti-CD19 AlloCAR T™ platform.

Initial Regulatory Milestones for ALLO-501:

The clinical investigation of ALLO-501 in the United States commenced following the Food and Drug Administration (FDA) approval of an Investigational New Drug (IND) application. This IND approval, announced in January 2019, permitted the initiation of the Phase 1 ALPHA study in patients with R/R NHL.14 This was a critical first step, allowing Allogene to gather initial human safety and efficacy data for its TALEN® gene-edited allogeneic CAR T-cell product.

Evolution to ALLO-501A (cemacabtagene ansegedleucel / "cema-cel"):

Based on early clinical experience and strategic considerations, Allogene advanced a next-generation candidate, ALLO-501A, subsequently named cemacabtagene ansegedleucel or "cema-cel." This product is intended for pivotal Phase 2 development and beyond.5 A key molecular difference between ALLO-501 and cema-cel is the elimination of the rituximab recognition domains in the latter.5 This modification was designed to broaden the applicability of the therapy, particularly for NHL patients who may have had recent exposure to rituximab, a commonly used anti-CD20 antibody. Consequently, the primary clinical development focus has shifted from the original ALLO-501 construct to cema-cel.7

Regulatory Designations for Cema-cel (ALLO-501A):

Cema-cel has received significant regulatory acknowledgments from the FDA, underscoring its potential to address unmet medical needs:

  • FDA Fast Track Designation: ALLO-501A (cema-cel) was granted Fast Track Designation by the FDA for the treatment of R/R Diffuse Large B-Cell Lymphoma (DLBCL).[5] This designation is intended to facilitate development and expedite the review of therapies for serious conditions that demonstrate the potential to address an unmet medical need.
  • FDA Regenerative Medicine Advanced Therapy (RMAT) Designation: In June 2022, the FDA granted RMAT designation to cema-cel for the treatment of third-line (3L) R/R LBCL.[7] The RMAT designation is specifically for cell and gene therapies targeting serious or life-threatening conditions and provides benefits similar to Breakthrough Therapy Designation, including more intensive FDA guidance on development and eligibility for accelerated approval pathways.

Information regarding Orphan Drug Designation (ODD) from the FDA or PRIME (PRIority MEdicines) designation from the European Medicines Agency (EMA) for ALLO-501 or cema-cel for specific indications like LBCL or FL was not explicitly available in the provided materials, although the general FDA ODD process is described.[14]

Current Developmental Focus and Strategic Pivot (Cema-cel):

Allogene Therapeutics has made a significant strategic decision to focus the development of cema-cel on earlier lines of LBCL treatment.

  • ALPHA3 Trial (NCT06500273): This is a pivotal Phase 2 trial evaluating cema-cel as a first-line (1L) consolidation therapy for patients with LBCL who have Minimal Residual Disease (MRD) following standard initial chemoimmunotherapy (e.g., R-CHOP).[7] The trial employs Foresight Diagnostics' investigational PhasED-Seq™ ctDNA-MRD platform to identify these high-risk patients who are likely to relapse despite an initial response to 1L therapy.[7] The vision for ALPHA3 is to position cema-cel as a "7th cycle" of frontline treatment, administered immediately upon MRD detection, to deepen responses and potentially boost cure rates.[9]
  • De-prioritization of Later Line Trials: Concurrently with the launch of ALPHA3, Allogene announced the de-prioritization of the currently enrolling third-line (3L) ALPHA2 trial (which was evaluating cema-cel) and the EXPAND trial.[9]

This strategic pivot towards 1L consolidation is a bold move. It suggests a belief that cema-cel's optimal therapeutic window and greatest clinical impact may be achieved by treating patients earlier in their disease course, when their tumor burden is lower and their overall physiological status may be better. The favorable safety profile observed in Phase 1, particularly the low rates of severe CRS and ICANS [1], likely underpins the confidence to explore its use in less heavily pretreated patients. This approach aims to redefine the standard of care by proactively intervening to prevent relapse, rather than solely competing in the more crowded R/R CAR T-cell therapy landscape.

The FDA's granting of Fast Track and RMAT designations for cema-cel in the R/R setting [5] provided significant validation of the platform's potential. These designations are not awarded lightly and reflect the agency's assessment that the therapy could offer a meaningful advantage over existing treatments or fill a void for patients with no satisfactory alternatives. They also facilitate more frequent interactions with the FDA, potentially streamlining the path to market approval.

While ALLO-501 itself may not be the final commercial product, its clinical program was instrumental. The IND approval in 2019 [14] and the subsequent Phase 1 data were crucial for demonstrating the initial safety, feasibility, and therapeutic potential of Allogene's TALEN®-edited allogeneic CAR T-cell platform. The learnings from ALLO-501, including the important design modification leading to cema-cel (removal of the rituximab-binding domain), directly shaped the trajectory of the current anti-CD19 program and informed the design of potentially registrational trials for cema-cel. ALLO-501 effectively served as the pathfinder for Allogene's current strategic direction in allogeneic CAR T-cell therapy.

8. Comprehensive Discussion and Future Perspectives

ALLO-501 stands as a pioneering candidate in the evolution of allogeneic CAR T-cell therapies, representing a significant endeavor to overcome the limitations of autologous treatments. Its development, though largely transitioned to its successor cemacabtagene ansegedleucel (cema-cel), has provided invaluable insights into the potential and challenges of "off-the-shelf" cellular immunotherapy.

Synthesis of ALLO-501's Attributes and Clinical Performance:

ALLO-501 was conceived as an allogeneic anti-CD19 CAR T-cell therapy, distinguished by its use of healthy donor T-cells and advanced TALEN® gene-editing technology. Key design features included the knockout of the TRAC gene to mitigate the risk of Graft-versus-Host Disease (GvHD) and the knockout of the CD52 gene to enable selective lymphodepletion with the anti-CD52 antibody ALLO-647, thereby promoting CAR T-cell persistence and efficacy.1

Clinical data from the Phase 1 ALPHA and ALPHA2 trials, particularly for the combined ALLO-501/cema-cel experience in CAR T-naïve relapsed/refractory Large B-cell Lymphoma (LBCL) patients, demonstrated promising anti-tumor activity. Objective response rates of 58% and complete response (CR) rates of 42% were achieved in this heavily pretreated population.[1] Crucially, responses in patients achieving a CR were notably durable, with a median Duration of Response (DOR) of 23.1 months and a median Progression-Free Survival (PFS) of 24.0 months.[8] The safety profile was generally manageable and particularly encouraging with respect to common CAR T-cell toxicities: rates of severe Cytokine Release Syndrome (CRS) were very low (0-1% Grade ≥3), and Immune Effector Cell–Associated Neurotoxicity Syndrome (ICANS) was not observed.[1] Critically, no instances of GvHD were reported, validating the TRAC knockout strategy.[1] However, infections (especially Cytomegalovirus reactivation) and hematologic cytopenias required careful monitoring and management, consistent with profound lymphodepletion and B-cell aplasia.[6]

Advantages of the Allogeneic Approach Validated by ALLO-501:

The ALLO-501 program has provided tangible evidence for several key advantages of the allogeneic CAR T-cell approach:

  • Accessibility and Timeliness: The "off-the-shelf" nature allows for rapid treatment initiation (median 2 days from enrollment) [1], a critical factor for patients with aggressive disease.
  • Scalable Manufacturing: The potential to produce multiple doses from a single donor run offers scalability and potentially greater product consistency compared to individualized autologous manufacturing.[1]
  • GvHD Mitigation: The successful prevention of GvHD through genetic engineering is a fundamental achievement for the safety of allogeneic T-cell therapy.[1]

Limitations and Ongoing Challenges:

Despite these advances, challenges remain. The overall median PFS of 3.9 months in the broader R/R LBCL cohort in Phase 1 8 indicates that while some patients achieve deep, durable remissions, many others do not, highlighting the need for better patient selection strategies or combination approaches. The requirement for a robust lymphodepletion regimen, including an additional antibody (ALLO-647), adds a layer of complexity and potential for immunosuppression-related side effects, though ALLO-647 itself appeared manageable with primarily low-grade infusion-related reactions.6 The long-term persistence of allogeneic CAR T-cells and the potential for eventual host immune rejection, even with advanced lymphodepletion strategies, remain areas of active investigation across the allogeneic field.

Role of ALLO-501 in Paving the Way for Cema-cel:

ALLO-501's clinical program was instrumental in providing proof-of-concept for Allogene's gene-editing platform and the accompanying lymphodepletion strategy. The evolution from ALLO-501 to cema-cel, characterized by the removal of the rituximab binding domain to enhance applicability 1, exemplifies an iterative and responsive drug development process, learning from early clinical data and anticipating real-world treatment scenarios.

Future Perspectives for the Allogene Platform via Cema-cel:

The developmental trajectory has now firmly shifted towards cema-cel, with a bold strategic focus on earlier lines of therapy. The pivotal Phase 2 ALPHA3 trial, evaluating cema-cel as a first-line consolidation treatment for LBCL patients with Minimal Residual Disease (MRD) after initial chemoimmunotherapy 7, has the potential to redefine the LBCL treatment paradigm. This approach aims to intercept relapse by treating high-risk patients when their disease burden is minimal, potentially increasing cure rates significantly. The integration of an ultra-sensitive MRD detection assay (Foresight CLARITY™) to identify these patients 7 showcases an innovative synergy between therapeutic and diagnostic advancements. If successful, this could shift CAR T-cell therapy from a later-line salvage option to a proactive, curative-intent intervention in the first-line setting.

Furthermore, the favorable safety profile of cema-cel, particularly the low rates of severe CRS and ICANS, opens the possibility of outpatient administration.[9] This, combined with its "off-the-shelf" availability, could dramatically expand patient access, allowing treatment in community cancer centers where the majority of LBCL patients are managed. The long-term implications for healthcare systems could be profound, potentially reducing the infrastructural and economic burdens associated with current autologous CAR T-cell therapies if cema-cel proves to be effective, safe, and more easily deployable in earlier treatment lines.

9. Conclusions

ALLO-501, while largely succeeded in clinical development by its refined successor cemacabtagene ansegedleucel (cema-cel), has played a crucial and foundational role in validating Allogene Therapeutics' allogeneic anti-CD19 CAR T-cell platform. As an early clinical candidate, ALLO-501 demonstrated the feasibility of using TALEN® gene-editing technology to create an "off-the-shelf" CAR T-cell product capable of inducing clinically meaningful responses in patients with relapsed/refractory B-cell malignancies.

The key innovations embedded in ALLO-501, notably the TRAC gene knockout for GvHD prevention and the CD52 gene knockout to enable selective lymphodepletion with ALLO-647, were successfully translated into the clinic. The Phase 1 ALPHA study provided critical proof-of-concept regarding the safety and preliminary efficacy of this approach, demonstrating a manageable safety profile characterized by low rates of severe Cytokine Release Syndrome and Immune Effector Cell–Associated Neurotoxicity Syndrome, and a complete absence of Graft-versus-Host Disease. Furthermore, the study yielded encouraging efficacy signals, particularly the durable complete responses observed in a subset of patients with Large B-cell Lymphoma.

The insights gained from the ALLO-501 program, including the strategic decision to remove the rituximab recognition domain, directly informed the development of cema-cel (ALLO-501A). Cema-cel now carries the mantle of this innovative platform forward, with regulatory designations such as FDA Fast Track and RMAT underscoring its potential. The current developmental trajectory, focused on the ALPHA3 trial evaluating cema-cel as a first-line consolidation therapy for MRD-positive LBCL patients, signifies a bold ambition to shift CAR T-cell therapy to earlier stages of treatment.

In conclusion, ALLO-501 has been a vital stepping stone, providing the essential clinical data and experience that have paved the way for cema-cel. The Allogene platform, as advanced through these candidates, holds the potential to address significant unmet medical needs in B-cell malignancies by not only improving upon the logistical challenges of autologous CAR T-cell therapies but also by potentially redefining treatment paradigms through earlier intervention and broader patient accessibility. The continued development and successful implementation of this allogeneic strategy could mark a new era in cellular immunotherapy.

Works cited

  1. Allogeneic Chimeric Antigen Receptor T-Cell Products Cemacabtagene Ansegedleucel/ALLO-501 in Relapsed/Refractory Large B-Cell Lymphoma: Phase I Experience From the ALPHA2/ALPHA Clinical Studies | Journal of Clinical Oncology - ASCO Publications, accessed May 12, 2025, https://ascopubs.org/doi/10.1200/JCO-24-01933
  2. First Allogeneic CAR T‐Cell Therapy Impresses in Relapsed/Refractory Lymphoma - PMC, accessed May 12, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC7330918/
  3. ALLO-501 by Allogene Therapeutics for Non-Hodgkin Lymphoma: Likelihood of Approval, accessed May 12, 2025, https://www.pharmaceutical-technology.com/data-insights/allo-501-allogene-therapeutics-non-hodgkin-lymphoma-likelihood-of-approval/?utm_source=lgp5-loa&utm_medium=24-270444&utm_campaign=recommended-articles-pi
  4. Safety and Efficacy of ALLO-501 Anti-CD19 Allogeneic CAR T Cells ..., accessed May 12, 2025, https://clinicaltrials.stanford.edu/trials/s/NCT03939026.html
  5. Allogene Therapeutics to Present Data from its Anti-CD19 AlloCAR T™ Therapy Program in Relapsed/Refractory Non-Hodgkin Lymphoma at the American Society of Clinical Oncology, accessed May 12, 2025, https://ir.allogene.com/news-releases/news-release-details/allogene-therapeutics-present-data-its-anti-cd19-allocar-ttm/
  6. ALLO-647 for Lymphodepletion in the Allogeneic CAR T Setting ..., accessed May 12, 2025, https://ashpublications.org/blood/article/142/Supplement%201/2095/501915/ALLO-647-for-Lymphodepletion-in-the-Allogeneic-CAR
  7. Allogene Therapeutics and Foresight Diagnostics Announce Partnership to Develop MRD-based In-Vitro Diagnostic for Use in ALPHA3, the First Pivotal Trial for Frontline Consolidation in Large B-Cell Lymphoma, accessed May 12, 2025, https://ir.allogene.com/news-releases/news-release-details/allogene-therapeutics-and-foresight-diagnostics-announce/
  8. Allogeneic Chimeric Antigen Receptor T-Cell Products ..., accessed May 12, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC12058369/
  9. Allogene Therapeutics Announces 2024 Platform Vision to Redefine the Future of CAR T Led by ALPHA3, the Industry's First Pivotal Trial for Frontline Consolidation in Large B-Cell Lymphoma, accessed May 12, 2025, https://ir.allogene.com/news-releases/news-release-details/allogene-therapeutics-announces-2024-platform-vision-redefine/
  10. ALPHA3 - Allogene, accessed May 12, 2025, https://allogene.com/alpha3/
  11. Definition of Allogeneic CRISPR-edited Anti-CD19 CAR T Cells PBLTT52CAR19 - NCI Drug Dictionary, accessed May 12, 2025, https://www.cancer.gov/publications/dictionaries/cancer-drug/def/allogeneic-crispr-edited-anti-cd19-car-t-cells-pbltt52car19
  12. Safety and Efficacy of ALLO-501 Anti-CD19 Allogeneic CAR T Cells in Adults With Relapsed/Refractory Large B Cell or Follicular Lymphoma - NCI, accessed May 12, 2025, https://www.cancer.gov/research/participate/clinical-trials-search/v?id=NCI-2019-03206
  13. Allogene to ramp up enrolment in Phase II CAR-T trials - Clinical ..., accessed May 12, 2025, https://www.clinicaltrialsarena.com/news/allogene-ramp-up-enrolment-phase-2-car-t-trials/
  14. FORM 20-F CELLECTIS S.A., accessed May 12, 2025, https://www.cellectis.com/uploads/files/CLSS_20-F_YE2024_USD_%282%29.pdf
  15. A randomized, open-label study evaluating the efficacy and safety of cemacabtagene ansegedleucel in participants with minimal residual disease after response to first line therapy for large B-cell lymphoma (ALPHA3) - MD Anderson Cancer Center, accessed May 12, 2025, https://www.mdanderson.org/patients-family/diagnosis-treatment/clinical-trials/clinical-trials-index/clinical-trials-detail.ID2024-0431.html?PageSpeed=noscript

Published at: May 12, 2025

This report is continuously updated as new research emerges.

© Copyright 2025. All Rights Reserved by MedPath