Page 13 - Read Online
P. 13

Haydu et al. J Cancer Metastasis Treat 2021;7:36  https://dx.doi.org/10.20517/2394-4722.2021.39  Page 9 of 14

               months, the median duration of response was not reached, and the median PFS was 18 months. Safety
               profiles were similar to what has been seen previously with liso-cel. Interestingly, of the 20 patients
               evaluable for minimal residual disease (MRD), 75% (15/20) had undetectable MRD in the blood and 87%
               (13/15) also had undetectable MRD in the bone marrow, raising the possibility of CLL/SLL disease
               eradication with CAR T approaches.

               Bridging patients to CAR T-cell therapies
               Bridging patients to CAR T therapy is an area of unmet clinical need. Bridging was permitted in the
                      [11]
                                        [9]
               JULIET  and TRANSCEND  trials of relapsed/refectory large B-cell lymphomas, with the majority of
               patients receiving bridging therapy (92% and 72%, respectively). ZUMA-1  prohibited bridging but most
                                                                              [10]
               patients receiving axi-cel in the real world context have also required bridging therapy . Importantly,
                                                                                            [44]
               bridging therapy has been associated with lower response rates and durability, with an ORR of 67.3% for
               bridged patients in TRANSCEND compared to 80.2% for non-bridged patients, as well as a higher incidence
               of CRS and neurologic events in bridged patients . A retrospective analysis of 298 patients with large B-cell
                                                        [9]
               lymphoma treated with commercially available axi-cel found that bridging therapy was associated with an
               inferior OS (HR = 1.7 in bridged vs. non bridged patients) . The inferior outcomes associated with
                                                                    [23]
               bridging are likely multifactorial and may largely reflect the characteristics of patients who require bridging
               therapy: patients with bulkier, more rapidly progressive, and/or symptomatic disease who are likely to have
               additional risk factors for inferior outcomes and increased toxicities, including high tumor burden.


               Current bridging approaches are not effective in many patients, a perhaps unsurprising observation given
               that patients eligible for CAR T therapy have typically had chemotherapy-refractory disease. Importantly, a
               significant subset of patients never make it to CAR T-cell infusion in part due to failure of bridging therapy
               to control disease, with 54/165 patients in JULIET ultimately not receiving CAR T-cells  and death from
                                                                                          [26]
               disease progression in 33/344 patients in TRANSCEND . In the brexu-cel relapsed/refractory MCL trial,
                                                               [9]
               the majority of the bridged patients actually had an increase in median tumor burden by radiographic
               imaging  over  the  bridging  period , further  reflecting  the  challenging  nature  of  controlling
                                                [12]
               relapsed/refractory disease with current bridging techniques and underscoring the need for novel
               approaches that maximize efficacy and limit systemic toxicity.


               In DLBCL patients awaiting CAR T therapy, bridging has typically been attempted with steroids,
               chemoimmunotherapy, and/or radiation. Steroids are a viable bridging option with less systemic toxicity
               than chemoimmunotherapy, though responses are often short lived. Radiation is an attractive bridging
               modality for localized and/or focally symptomatic disease given the minimal systemic toxicity. Potential
               novel bridging approaches include agents which have demonstrated efficacy in relapsed/refractory DLBCL,
               including brentuximab vedotin for CD30+ disease , lenalidomide or BTKi for non-GCB disease , and the
                                                         [45]
                                                                                                 [46]
               CD79B antibody drug conjugate polatuzumab vedotin . The anti-CD19 therapy tafastimab has promising
                                                             [47]
               activity in combination with lenalidomide in relapsed/refractory DLBCL for which it is FDA approved ,
                                                                                                       [48]
               but the impact of CD19-targeted agents prior to planned anti-CD19 CAR T-cells is unknown, and should be
               avoided in the absence of data showing it does not impair the curative potential of subsequent anti-CD19
               CAR T-cells.

               THE FUTURE OF CAR T
               Expanding eligibility
               Patients with secondary CNS lymphoma were excluded from the initial axi-cel and tisa-cel trials given the
               theoretical concern for CAR T-cell related neurotoxicity but were eligible for liso-cel in TRANSCEND as
               long as there was also measurable non-CNS disease. Overall, 7/269 (3%) of patients in TRANSCEND had
   8   9   10   11   12   13   14   15   16   17   18