
Afami-Cel Shows Durable Persistence and Tumor-Killing Abilities in Synovial Sarcoma
Treatment with afami-cel was associated with long-term persistence in the periphery that led to clinical benefit in patients with synovial sarcoma.
Treatment with afamitresgene autoleucel (afami-cel; Tecelra) was associated with long-term persistence in the periphery that led to clinical benefit in patients with synovial sarcoma, according to findings from translational analyses of the phase 2 SPEARHEAD-1 trial (NCT04044768) presented at the 2024 CTOS Annual Meeting.1
SPEARHEAD-1 investigators found that afami-cel accumulated after infusion and reached peak concentration at a median of 7 days post-infusion. Furthermore, afami-cel was detected in patients at over 3 years post-infusion. The median terminal half-life of the product was 152 days; this value was 263 days in responders and 84 days in non-responders. Investigators noted that afami-cel persistence tended to plateau over time in several patients, which was consistent with the durable persistence findings. Notably, 1 patient had an afami-cel level below quantification limits at any time point.
Afami-Cel’s Mechanism of Action and FDA Approval
Afami-cel is an engineered high-affinity TCR T-cell product that expresses an exogenous T-cell receptor and targets a peptide from HLA-presented MAGE-A4—a cancer antigen that is upregulated in certain tumors. Patient eligibility for afami-cel is restricted to patients with tumors that express MAGE-A4 and have HLA molecules that will react with the afami-cel T-cell receptor.
Cohort 1 of the open-label, international SPEARHEAD-1 trial evaluated the efficacy and safety of afami-cel in 44 patients with HLA-A*02–positive, MAGE-A4–expressing, advanced or metastatic synovial sarcoma.1,2 Patients received 1 infusion of afami-cel following lymphodepleting chemotherapy.
In this trial, afami-cel elicited an overall response rate of 39% (95% CI, 24%-55%).2 The median duration of response was 11.6 months (95% CI, 4.4-18.0).1
Regarding safety, cytopenias were the most common grade 3 or higher adverse effect.2 Additionally, most patients developed cytokine release syndrome (CRS), 1 of whom had grade 3 CRS.
On August 2, 2024, the FDA
Translational Research Goals
Investigators conducted translational analyses to determine how the mechanism of action of afami-cel induced the clinical responses seen in SPEARHEAD-1.1 Blood samples from the periphery of afami-cel–treated patients were assessed, along with tumor biopsies taken before and after treatment.
The objectives of these analyses were to identify how afami-cel induces long-term responses and to collect other findings to support the product’s intended mechanism of action.
Additional Persistence Findings
At a data cutoff of March 29, 2023, additional data from the translational research showed that patients who achieved an objective response experienced greater long-term pharmacokinetic exposure to afami-cel at an area under the curve (AUC) of 0 to 12 months vs non-responders (P < .001). Greater long-term pharmacokinetic exposure also correlated with significantly longer progression-free survival (PFS). Patients with an AUC of 0 to 12 months at or above the median (n = 17) were associated with longer PFS compared with patients with an AUC of 0 to 12 months below the median (n = 16; P < .05).
After infusion, both CD4- and CD8-positive afami-cel T cells developed phenotypes of memory T cells, defined as T cells that can persist long term, which express unique phenotypic markers. Flow cytometry showed that expression of memory markers increased on afami-cel after infusion, indicating that conversion to memory phenotypes was consistent with long-term afami-cel persistence. Flow cytometry also found that afami-cel demonstrated consistently low expression of T-cell exhaustion phenotypic markers beyond 2 years post-infusion; this finding was consistent with the product’s cytolytic functionality retention and long-term persistence.
Case Study Findings
A case study showed that 1 patient who received afami-cel achieved a sustained response, including tumor cell killing, lasting longer than 3.7 years post-infusion. Two of this patient’s 3 target lesions completely resolved, and 1 lesion had a partial response (PR). Lesion 2 shrank more than 3 years post-infusion. Investigators noted that this delayed tumor shrinkage indicates that afami-cel retains a long-term ability to kill tumor cells.
Through flow-based sorting, investigators detected and isolated afami-cel from this patient and used an ex vivo killing assay, which showed that the isolated afami-cel retained its killing ability 3.3 years post-infusion.
Investigators noted that similar long-term afami-cel potency was also seen in other patients.
Afami-Cel’s Activity in the Tumor Microenvironment
Using RNA in situ hybridization, investigators detected afami-cel’s infiltration of tumoral and stromal regions in 30 out of 32 tested post-infusion biopsies. This analysis demonstrated that afami-cel can infiltrate solid tumors, which is consistent with its intended mechanism of action.
Afami-cel was also shown to activate endogenous immune responses through T-cell infiltration in 23 synovial sarcoma tumors, including 4 that had a PR, 15 that had stable disease, and 4 that had progressive disease. Investigators saw a significant increase in the densities of all CD3-positive T cells in the tumor beyond afami-cel cells (P < .001) that was attributed to an influx of afami-cel transgene–negative endogenous T cells in the tumors after treatment. Levels of Ki67-positive CD8-positive T-cells (which are indicative of proliferation) and Grb-positive CD8-positive T cells (which are indicative of killing ability) also increased following afami-cel infusion.
Investigators explained that these findings show that beyond directly killing tumor cells, afami-cel activates a patient-driven antitumor response by inflaming the tumor microenvironment and recruiting endogenous T cells that can proliferate and kill tumor cells.
Disclosures: Dr Druta reported receiving consultancy fees and advisory board fees from Aadi Bioscience, Adaptimmune, Daiichi Sankyo, and Deciphera Pharmaceuticals. All other study authors are employees of, and may hold stock/options in, Adaptimmune. The study was sponsored by Adaptimmune.
References
- Druta M, Dupont CD, Bath N, et al. Translational analyses reveal mechanisms of afami-cel’s anti-tumor activity in synovial sarcoma. Presented at: 2024 CTOS Annual Meeting. November 13-16, 2024; San Diego, California.
- D'Angelo SP, Araujo DM, Abdul Razak AR, et al. Afamitresgene autoleucel for advanced synovial sarcoma and myxoid round cell liposarcoma (SPEARHEAD-1): an international, open-label, phase 2 trial. Lancet. 2024;403(10435):1460-1471. doi:10.1016/S0140-6736(24)00319-2
- Adaptimmune receives U.S. FDA accelerated approval of Tecelra (afamitresgene autoleucel), the first approved engineered cell therapy for a solid tumor. News release. Adaptimmune Therapeutics. August 1, 2024. Accessed November 20, 2024. https://www.adaptimmune.com/investors-and-media/news-center/press-releases/detail/271/adaptimmune-receives-u-s-fda-accelerated-approval-of


































