Aneuploidy is a hallmark of human cancer, yet the molecular mechanisms to cope with aneuploidy-induced cellular stresses remain largely unknown. Here, we induced chromosome mis-segregation in non-transformed RPE1-hTERT cells and derived multiple stable clones with various degrees of aneuploidy. We performed an unbiased genomic, transcriptomic and proteomic profiling of 6 isogenic clones, using whole-exome DNA, mRNA and miRNA sequencing, as well as proteomics. Concomitantly, we functionally interrogated their cellular vulnerabilities, using genome-wide CRISPR/Cas9 and large-scale drug screens. Aneuploid clones activated the DNA damage response, and were consequently more resistant to further DNA damage induction. Aneuploid cells also exhibited elevated RAF/MEK/ERK pathway activity, and were more sensitive to clinically-relevant drugs targeting this pathway, and in particular to CRAF inhibition. Importantly, CRAF and MEK inhibition sensitized aneuploid cells to DNA damage-inducing chemotherapies and to PARP inhibitors. These results were validated in human cancer cell lines. Overall, our study provides a comprehensive resource for genetically-matched karyotypically-stable cells of various aneuploidy states, revealing a novel therapeutically-relevant cellular dependency of aneuploid cells. This submission contains the raw files, the library used for the analysis, and the corresponding DIA-NN report and associated files.