Page 31 - Read Online
P. 31

D'Souza et al. J Cancer Metastasis Treat 2022;8:28  https://dx.doi.org/10.20517/2394-4722.2022.51  Page 3 of 15

               6.2 months, but a slight improvement in overall survival [19,20] . The combination of nivolumab (anti-PD-1
               antibody) plus ipilimumab (anti-CTLA-4 antibody) (NCT03048474, NCT02716272, NCT02899299) resulted
               in a PFS of 4.8-6.8 months (OS 15.9-18.1) and further improved MPM response rates (RR) from 25.8 to 29%
                                                             [20]
               as compared with anti-PD1/ anti-PD-L1 monotherapy . Of note, the MPM patients who seemed to benefit
               the most were those with sarcomatoid histology . The promising results of opilimumab/nivolumab
                                                           [21]
               combination therapy led to the approval of this regime for use as first-line therapy for MPM. As second-line
               therapy for MPM, gemcitabine and/or vinorelbine or ICI therapies are used [22-25] . Figure 1 outlines the
               currently available therapeutic options for MPM, with the timeline representing the median OS.

               In a study by Kim et al., ICIs appeared to benefit patients with advanced MPM after initial platinum-based
                           [26]
               chemotherapy . These findings, however, could not be extrapolated in the PROMISE-meso phase III
               randomized clinical trial, which failed to demonstrate the survival benefit of pembrolizumab over
               gemcitabine or vinorelbine . Resistance to ICIs is multifactorial, involving the TME, the intrinsic tumoral
                                      [27]
               signaling and host genetic factors of which the primary resistance to ICIs in MPM is attributed to the
               moderate to low levels of PD-L1 expression with low tumor mutational burden (TMB) . Compared with
                                                                                          [28]
               MPM, MPeM has a slightly higher PD-L1 and TMB, and this difference in biology may unravel the ICI
               therapy . Despite the clinical, molecular and epidemiological differences, the MPeM treatment strategies
                      [29]
               are largely based on the MPM studies, that is retrospective evidence-based. The overall low response rate for
               the second-line treatment drugs warrants further research and patients' referrals for participating in clinical
               trials.


               CAR-T CELL THERAPY IN MM
               Cellular immunotherapy, or adoptive cell therapy, are treatments that utilize the body’s immune cells to
               fight cancer. Several cellular immunotherapies exist, such as engineered T cell receptor (TCR) therapy,
               tumor-infiltrating lymphocyte (TIL) therapy, and chimeric antigen receptor T (CAR-T) cell therapy, all
               demonstrating feasibility of adoptive transfer and treatment successes, especially in hematological
               malignancies .
                          [30]

               Cellular immunotherapy research in MM has been mainly focused on CAR-T cell therapy. CAR-T cells are
               patient autologous T cells that are genetically re-engineered ex-vivo to express a non-MHC-restricted
               receptor (CAR) targeting a specific tumor cell surface antigen. The CAR is composed of an antibody-
               derived single-chain variable fragment (scFv) fused to a flexible spacer/hinge region, a transmembrane part,
               and an intracellular signaling domain (CD3-ζ or Fc-γ) [31,32] . The CD3ζ intracellular signaling domain of
               CARs (or else, 1st generation CARs) was shown to provide limited therapeutic efficacy in initial trials , so
                                                                                                     [33]
               CAR technology evolved to 2nd and 3rd generation CARs. These newer generation CARs incorporated
               additional co-stimulatory molecules, such as CD28, 4-1BB, or OX40, to the CD3ζ primary signaling domain
               and exhibited enhanced cell proliferation and greater antitumor potency due to increased signaling
                      [34]
               strength . To date, all the available FDA/EMA/NICE-approved CAR-T cell therapies are either
               CD28/CD3ζ or 41BB/CD3ζ second-generation CARs.

               CAR-T cell therapies against tumor-associated antigens (TAAs) or stroma-associated proteins have been
               developed and trialed in MM. Mesothelin (MSLN) is a cell-surface glycoprotein involved in tumor invasion
               and is expressed in 85% to 90% of MMs, with its expression elsewhere limited to the mesothelial surfaces at
               low levels [35-37] . Previous research involving MSLN-knockout mice established no histologic or anatomic
               abnormalities, suggesting MSLN may be non-essential for normal mouse development or reproduction ,
                                                                                                       [38]
               making it a great therapeutic target. The majority of current CAR-T cell trials target MSLN in MPM  , few
                                                                                                    [39]
               in MPeM, and a range of solid tumors . Other CAR-T cell targets explored in preclinical studies of MM
                                                [40]
   26   27   28   29   30   31   32   33   34   35   36