Page 13 - Read Online
P. 13

Laubach et al. Cancer Drug Resist 2023;6:611-41  https://dx.doi.org/10.20517/cdr.2023.60                                         Page 615

                                                                      [30]
               leading to mitochondrial dysfunction and, ultimately, apoptosis . Genetic ablation of FIP200 in naïve T
                              +
                                       +
                                                                         [30]
               cells reduced CD8  and CD4  T cell infiltration and IFNg production . Recently, tumor-derived lactate was
                                                                   +
               also found to diminish TCA-intermediate recycling in CD8  T cells by inhibiting pyruvate carboxylase,
                                                [31]
               which shunts pyruvate to oxaloacetate . Pyruvate carboxylase is exceedingly important to maintain TCA
               cycle anaplerosis in CD8  T cells because succinate is diverted from the TCA cycle to participate in
                                      +
               autocrine signaling . In addition to tumor-derived lactate suppressing CD8  T cell function, it also drives
                               [31]
                                                                                +
               the expansion and function of immunosuppressive cells. Tregs inhibit the function of anti-tumor immune
               cells and require lactate to maintain their suppressor functions in the harsh TIME [32,33] . Moreover, lactate
               produced by cervical cancer cells supports immunosuppressive macrophages by regulating anti-
               inflammatory cytokine production and HIF1a expression . Taken together, these data highlight that
                                                                  [34]
               tumor-derived lactate not only directly inhibits effector T cell functions, but also indirectly through
               supporting immunosuppressive cell populations. As such, multiple reports have examined the feasibility of
               inhibiting tumor-intrinsic lactate metabolism in combination with anti-PD-1/PD-L1 therapy.
               Several correlative studies through bioinformatic analyses have demonstrated that targeting lactic acid
               metabolism might overcome ICB resistance and yield better patient outcomes. High LDH expression has
               been evaluated as a selection criterion for and predicting response to ICB therapy [35-39] . Similarly, other
               lactate-related genes have been correlated with the expression of immune checkpoint proteins, CD8  T cell
                                                                                                    +
               infiltration, and resistance to ICB in breast cancer . Moreover, decreased glycolytic flux in melanoma
                                                           [40]
               patients treated with anti-PD-1 therapy was associated with increased probability of progression-free
                      [41]
               survival .
               In addition to bioinformatics studies, numerous reports indicate that inhibiting tumor-intrinsic lactic acid
               metabolism in combination with anti-PD-1/PD-L1 therapies combats resistance and increases efficacy.
               MCT4 is regulated at the mRNA level by the demethylase alkB homolog 5 (ALKBH5) . Genetic or
                                                                                             [42]
               pharmacologic inhibition of ALKBH5 reduces intratumoral lactate concentration and the number of Tregs
               and MDSCs, but has no effect on the number of infiltrating cytotoxic T cells . Furthermore, utilizing a
                                                                                  [42]
               small molecular inhibitor of ALKBH5 significantly improved the efficacy of anti-PD-1 treatment in murine
                               [42]
               melanoma tumors . Consistent with the findings that lactic acid benefits immunosuppressive cells,
               researchers found that lactic acid produced by high-glycolytic tumors drove expression of PD-1 on Tregs,
                          +
                                                             [43]
               but not CD8  T cells, leading to anti-PD-1 resistance . However, inhibiting either LDHA in tumors or
               MCT1 in Tregs combined with anti-PD-1 therapy reversed these effects . In addition to inhibiting lactic
                                                                             [43]
               acid production and/or lactate import, antagonizing intracellular lactate signaling in malignant cells
               through HCAR1 also promotes anti-tumor effects . Abrogating HCAR1-mediated lactate signaling
                                                            [44]
               sensitized tumors to anti-PD-1 and metformin treatment, leading to reduced tumor volume and increased
               CD8  T cell infiltration and IFNg production .
                                                     [44]
                   +
               While a plethora of evidence supports the notion that lactic acid production by tumors and accumulation in
               T cells drives oncogenesis, a few reports contradict this idea. In mouse melanoma tumors, blocking the
               export of lactate and H  ions through MCT1 and MCT4 reduced the acidification of the TIME . While
                                   +
                                                                                                  [41]
               blocking MCT1 and 4 in T cells decreased lactate secretion and glucose uptake, it surprisingly did not
               impair IFNg production , which contrasts with other findings that accumulation of intracellular lactic acid
                                    [41]
               promotes acidification and dampens effector functions [21,22,24] . The authors found that inhibiting MCT1 and
               4 activities in T cells increased glucose flux through the TCA cycle and increased oxygen consumption, thus
                                                      +
                                                                                             [41]
               providing  an  explanation  as  to  why  CD8   T  cell  effector  functions  were  preserved . Moreover,
               pharmacologically inhibiting MCT1 and 4 in combination with anti-PD-1 treatment resulted in increased
                                                [41]
               efficacy and decreased tumor volume . The results from these findings are indeed surprising given the
   8   9   10   11   12   13   14   15   16   17   18