Off-the-shelf CAR T-cell therapy effective for advanced T-cell acute lymphoblastic leukemia
TruUCAR GC027 — a gene edited, investigational allogeneic chimeric antigen receptor T-cell therapy — showed efficacy for treatment of relapsed or refractory T-cell acute lymphoblastic leukemia, according to preliminary results of a first-in-human study presented at the virtual American Association for Cancer Research Annual Meeting.
There were some safety concerns regarding the investigational CAR T-cell therapy, as all five patients treated experienced grade 3 or higher cytokine release syndrome (CRS). However, no patients developed acute graft-versus-host disease after infusion, which was the aim of the genetic editing engineered into the TruUCAR GC027 (Gracell Biotechnologies) product.
“Despite the high unmet medical need of treatment for T-cell acute lymphoblastic leukemia, the development of novel immunotherapies has been lacking,” Xinxin Wang, PhD, of Gracell, said during a presentation.
One obstacle to developing effective therapies is that T-ALL cells share the same antigens as normal T cells, Wang said. That means targeted T-ALL therapies also will target normal, healthy T cells.
There were some safety concerns regarding the investigational CAR T-cell therapy, as all five patients treated experienced grade 3 or higher cytokine release syndrome (CRS).
Another issue is potential lymphoblast contamination in autologous CAR T-cell products, which Wang said can be avoided using some of the novel engineering baked into TruUCAR GC027.
TruUCAR GC027 is a second-generation anti-CD7 CAR that uses cells from HLA-mismatched healthy donors. These T cells are genetically edited using CRISPR/Cas9 technology to knock out T-cell receptor-alpha to prevent GVHD and CD7 to avoid “cell fratricide” after infusion into the patient.
The single-arm, open-label, multicenter prospective study aimed to assess the safety, clinical activity and cell expansion of TruUCAR GC027 for adults with relapsed or refractory T-ALL.
Five patients (median age, 24 years; range, 19-38) enrolled in this study have received TruUCAR GC027 as of Feb. 6 at one of three dose levels — 0.6 × 107 cells/kg (n = 1), 1 × 107 cells/kg (n = 3) or 1.5 × 107 cells/kg (n = 1). Study participants had a marrow tumor load range of 4% to 80.2% and a median five previous lines of therapy. No patients underwent previous hematopoietic stem cell transplantation.
Participants received 6 days of lymphodepletion chemotherapy followed by a single infusion of TruUCAR GC027. The investigators evaluated adverse events, disease response and cell expansion kinetics.
Four patients had a minimal residual disease (MRD)-negative complete response to therapy at the day 28 follow-up evaluation. Three of these patients remained MRD-negative at subsequent follow-up evaluations (up to 161 days), and none of them needed subsequent HSCT.
PAGE BREAK
One patient had an MRD-negative complete response at day 14 but experienced disease relapse by day 29.
All four patients who had MRD-negative complete responses also had peak cell expansion in their peripheral blood observed between weeks 1 and 2 after infusion. One patient with central nervous system disease had TruUCAR GC027 in samples from his bone marrow and cerebrospinal fluid.
Four patients had grade 3 CRS as determined by consensus grading established by the American Society for Transplantation and Cellular Therapy. The remaining patient experienced grade 4 CRS, along with elevated levels of interleukin-6, interferon-gamma and tumor necrosis factor-alpha. CRS symptoms were manageable and resolved with supportive care for all patients.
None of the study patients developed neurotoxicity or GVHD. One patient had prolonged cytopenia because of a fungal infection that required subsequent antifungal therapy.– by Drew Amorosi
Reference:
Wang X, et al. Abstract CT052. Presented at: AACR Annual Meeting; April 27-28, 2020 (virtual meeting).
Disclosures: Wang is an employee of Gracell Biotechnologies. Please see the abstract for all other researchers’ relevant financial disclosures.
Perspective
Back to Top
Joseph Alvarnas, MD
Although CAR T cells have proven to be an extraordinary tool for treating patients with relapsed and refractory B-cell acute lymphoblastic leukemia and diffuse large B-cell lymphoma, there have been a number of limiting challenges associated with delivering these therapeutics. Currently, each CAR T-cell product must be created ad hoc for each patient. This creates a built-in logistical time delay for candidate patients who have relapsed, progressive and refractory ALL and DLCBL. During this time, patients may develop progressive disease that makes them ineligible to receive CAR T cells.
Additionally, because CAR T cells to date have to be manufactured on a per-patient basis, these products are costly to deliver because each is individually manufactured; thus, they cannot be efficiently delivered on a basis that can benefit from the economies of scale that can be achieved for other pharmaceutics that are manufactured in large lots and that can be administered to multiple patients.
Finally, logistically, these cells can’t be successfully produced for every patient; there is a failure rate in cell production. All of these factors are challenges under our current CAR T model that may limit the utility and effectiveness of these cells. This study points toward a different direction — this idea of a universal CAR T cell therapy that may, in fact, help to overcome each of the three limitations that I described.
The results of the TruUCAR GC027 study are encouraging because they demonstrate the potential effectiveness of a “universal” CAR T-cell product. Creating an off-the-shelf product that can be repurposed for many patients allows us to expand the potential utility of CAR T cells. The fact that they can be delivered with a speed that is impossible with our existing portfolio of commercially available products really points to this technology as being one that is extraordinarily promising and vastly intriguing from a clinical perspective. I would, however, caution in overinterpreting the outcomes for a patient experience based upon a relatively small population. Although this study provides encouraging results for the antitumor effectiveness of these cells in both animal models and in the human subjects of this trial, these results should be validated in a larger population of patients with T-cell ALL so that we can weigh the full importance and the clinical relevance of this set of discoveries.
Moreover, we need longer-term results to see whether these cellular products can produce long-term, disease-free remissions for patients. The other issue here is when you look at the expansion kinetics of the peripheral blood of patients, you see an initial peak followed by a drop-off. One of the things that seems to be correlated with more successful CAR T-cell treatments is expansion and persistence of these cells in the blood of patients. So, I would like to see more data related to the persistence of the engineered cell population — that is, the therapeutic cells — in the blood of those patients who continue to survive. That will be a very important data point in understanding how this performs biologically over time.
Joseph Alvarnas, MD
City of Hope
Disclosures: Alvarnas reports no relevant financial disclosures.
Perspective
Back to Top
Yvonne Y. Chen, PhD
The trial conducted by Wang and colleagues specifically targets CD7 with its allogeneic CAR construct. This introduces additional challenges because CD7 is expressed on the surface of T cells. So, to prevent fratricide — or CAR T cells killing each other — the strategy is to knock out CD7 with gene editing.
Despite this concern, Wang and colleagues were able to achieve complete responses with a single dose of CAR T cells and they did so without observable GVHD or neurotoxicity.
However, it is important to note the high rate of grade 3 or higher toxicities in this small study group, with all five patients experiencing at least grade 3 CRS. This may have something to do with the fairly high dosing levels used in this trial.
Perhaps the most interesting findings for those of us in the field are yet to come and will apply to the durability of responses that will follow this relatively short 7-month monitoring period.
We will start to see more genetic features, such as knock-ins and knock-outs, as well as manufacturing protocol optimizations geared toward promoting T-cell persistence and function.
The debate will continue over whether its inherently better to do things off the shelf or to use autologous cell therapies. In the end, the correct answer may depend on the type of disease and the number of previous therapies the patient has received and, therefore, the impact on T-cell quality that can be obtained from the patient.
As we develop more sophisticated ways of engineering T cells, we need to be cognizant of the potential trade-off in T-cell function. The more genetic features we put in, the more difficult it will be to ensure uniformity and genetic stability of the T-cell product. There is a balancing act we must consider as we continue to further engineer and manipulate these T cells.
Yvonne Y. Chen, PhD
University of California, Los Angeles
Parker Institute for Cancer Immunotherapy
Disclosures: Chen is co-founder of Kalthera Therapeutics and serves as a scientific advisor to Gritstone Oncology and Notch Therapeutics.
Published by:
Sources/DisclosuresCollapse
Source:
Wang X, et al. Abstract CT052. Presented at: AACR Annual Meeting; April 27-28, 2020 (virtual meeting).
Disclosures:
Wang is an employee of Gracell Biotechnologies. Please see the abstract for all other researchers’ relevant financial disclosures.