Thursday, December 19, 2019

Unintended Consequences

With the many new cancer therapeutics coming almost daily it is being discovered that some of these check certain deficiencies and evoke others. In a recent Science piece the authors note:

Russo et al. found that human CRC cell lines that were treated with EGFR or BRAF inhibitors down-regulated the expression of high-fidelity DNA repair proteins and increased that of error-prone DNA repair proteins, which may both increase mutation rates. Using reporter assays, they further showed that the fidelity of DNA mismatch repair (MMR) and homologous recombination (HR) repair systems were impaired and that DNA damage increased during drug treatment. Genetic analysis of cell lines that had been exposed to these inhibitors revealed subclonal mutations in dinucleotide repeats, which are characteristic of defective MMR. In contrast to other cancer mutational processes—such as genetically encoded HR or MMR defects that lead to persistent mutation acquisition or overexpression of the APOBEC (apolipoprotein B mRNA editing enzyme, catalytic polypeptide-like) DNA cytidine deaminases, which generates mutational bursts—the mutagenesis program identified by Russo et al. was tightly coupled to drug exposure and ceased after drug removal. This study demonstrates that nongenotoxic targeted oncogene pathway inhibitors can promote a temporally restricted increase in mutability by switching from high-fidelity to error-prone DNA repair. Adaptive mutagenesis is a mechanism described in bacteria that increases the mutation rate in response to cell stress. This is triggered by a cell-stress signaling pathway that activates error-prone DNA double-strand break repair and it is accompanied by suppression of MMR. Adaptive mutagenesis increases the probability of generating mutations that enable evolutionary adaptation of unicellular organisms to new environments. On the basis of the pronounced similarities of drug-induced mutagenesis in CRC and adaptive mutagenesis in bacteria, Russo et al. explored whether the mammalian target of rapamycin (mTOR) pathway, a major stress signaling pathway in humans, controls drug-induced mutagenesis in cancer cells. mTOR signaling was indeed inactivated by drug treatment, but inhibiting the mTOR pathway alone did not phenocopy the changes in DNA repair protein expression. The trigger of drug-induced mutagenesis in CRC cells is therefore either more complex or different from that in bacteria.

 This is not at all unexpected. Indeed this is a clear suggestion that any single genetic control can and is adaptively bypassed. Initial control may be effected but it will be essential to treat many cancers as chronic diseases as they move from genetic change to genetic change. 

The challenge going forward will be to detect and address these changes in some dynamic basis. One suspects that non-invasive liquid biopsies may be effective.