Characterization of RAC1 P29S oncogenic properties
To characterize the RAC1P29S effect on downstream signaling, morphology, and cell proliferation, we generated an inducible cell system of RAC1P29S expression. Melanocytes isolated from C57BL/6 mice were obtained and genetically altered via CRISPR/Cas technology to knock out Pten, since this is a commonly altered tumor suppressor in cutaneous melanoma and mouse models of combined Braf/Pten mutations have been well-studied (4, 5) (Supplemental Fig. 1). A plasmid containing a destabilization domain (DD) fused to RAC1P29S was then introduced into these C57BL/6 Pten−/− melanocytes (Fig. 1A). Under basal conditions, the DD results in translation of an unstable protein, which is immediately targeted and degraded by the proteasome. However, upon treatment with Shield1 ligand (SHLD1), the DD is stabilized, and the recombinant protein is expressed (11). This expression system was originally produced in a non-transformed C57BL/6 cell line to delineate molecular consequences specific to the RAC1P29S mutation without confounding signaling inputs from other potential oncogenes. After establishing cellular RAC1P29S expression, we examined the RAC1P29S effect on downstream MAPK and AKT signaling, both of which lead to cell proliferation and survival. RAC1P29S resulted in a slight increase in phospho-PAK, phospho-ERK and phospho-AKT T308 (Fig. 1B). Melanocytes that expressed RAC1P29S also displayed faster proliferation rates than non P29S-expressing counterparts (Fig. 1C). As RAC1 is a master regulator of actin dynamics, we explored the effect on motility, migration, and actin structure. To further examine the effects of RAC1P29S on cell motility and migration, we performed scratch and transwell assays. RAC1P29S-expressing melanocytes exhibited higher motility, as a significantly higher number of cells migrated into the scratch area (Fig. 1D). Additionally, we observed higher cell migration in a transwell assay (Fig. 1E). Phalloidin staining of actin displayed increased membrane ruffling and lamellipodia formation compared to melanocytes that did not express the P29S mutation (Fig. 1F). This data concludes that RAC1P29S is an activating mutation that leads to MAPK activation, lamellipodia formation, and increased motility and migration. However, as these effects are modest, there may be additional mechanisms that contribute to RAC1-driven oncogenicity.
Proteogenomic analysis of RAC1 P29S melanocytic expression reveals upregulation of cell cycle processes.
To determine the molecular changes induced by RAC1P29S expression, we performed RNA-sequencing (RNA-seq) coupled with multiplexed inhibitor bead-mass spectrometry (MIBs/MS). RNA-seq analysis of expressed recombinant RAC1P29S mouse-derived melanocytes resulted in 600 differentially expressed genes. E2F_TARGETS and G2M_CHECKPOINT were the top two enriched hallmark gene sets (Fig. 2A, B). Other cell cycle related processes included MYC_TARGETS_V1 and MITOTIC_SPINDLE (Fig. 2A). The top 10 enriched REACTOME gene sets were all cell cycle events, indicating RAC1P29S has a role in influencing cell cycle progression, consistent with previous studies on RAC1 (Fig. 2C) (12, 13). In addition, INFLAMMATORY_RESPONSE and IL-2_STAT5_SIGNALING hallmark gene sets were enriched, suggesting a potential function in modulating inflammation (Fig. 2A). Ingenuity Pathway Analysis (IPA) identified the top altered upstream regulators in response to RAC1P29S expression (Fig. 2D). Top activated upstream regulators included mainly transcription factors that have been reported to promote expression of cell cycle regulating genes such as E2F proteins, YAP1, CCND1, and FOXM1. Accordingly, the top inactive upstream regulators were tumor suppressors and cell cycle inhibitors including COPS5, SMARCB1, CDKN2A, and RB1 (Fig. 2D). Interestingly, the activity of several transcriptional regulators was altered as well, including MED1 (a subunit of the mediator complex which helps position RNA polymerase II on DNA promoter regions), HDAC1 (histone deacetylase), and PRDM16 (histone methyltransferase) (Fig. 2D). MIBs/MS further implicated cell cycle progression as an enriched pathway in RAC1P29S-expressing cells through identification of 7 kinases involved specifically in G2/M transition (Fig. 2E).
To further investigate the RAC1P29S influence on the cell cycle, the cell cycle profile was examined by flow cytometry. RAC1P29S-expressing melanocytes showed a lower percentage of cells in G2/M phase, indicating the RAC1P29S mutation may lead to accelerated transition through the G2/M cell cycle checkpoint (Supplemental Fig. 2A). P29S-expressing melanocytes also exhibited a higher percentage of cells in S phase, indicating increased DNA replication, consistent with gene set enrichment analysis (GSEA) (Supplemental Fig. 2A). Additionally, live H2B-GFP cell tracking resulted in less time spent in the prophase to anaphase stages of mitosis in the RAC1-mutant cells, further suggesting a role in mitotic cell cycle progression (Supplemental Fig. 2B).
RAC1 P29S -expressing melanocytes show increased sensitivity to CDK9 inhibition.
Due to the strong G2/M cell cycle phase transition signature yielded by the proteogenomic and cell cycle analysis, we next further examined the kinase signaling cascade that promotes G2 to M phase transition to investigate potential targeting vulnerabilities of RAC1P29S-expressing melanocytes. PAK1 phosphorylates Aurora kinases (AURK) and Polo-like kinase 1 (PLK1), which in turn phosphorylates CDC25, ultimately leading to CDK1 phosphorylation, activation, and G2/M phase transition (14, 15) (Fig. 3A). While there were no differential sensitivities to alisertib (AURKA inhibitor), tozasertib (pan-AURK inhibitor), or volasertib (PLK1 inhibitor), melanocytes expressing a RAC1P29S mutation were significantly more sensitive to dinaciclib (CDK1,2,5,9 inhibitor) (Fig. 3B). However, as dinaciclib is not specific to CDK1, we performed a CDK inhibitor viability panel to determine which CDK target conferred the observed sensitivity (Fig. 3C). Interestingly, the compounds that targeted CDK9 were all significantly more cytotoxic to melanocytes expressing the RAC1P29S mutation (Fig. 3C,D).
To further examine RAC1P29S-mediated sensitivity to CDK9, we generated a constitutive RAC1P29S-expressing melanoma cell line. YUMM1.7 cell lines (BrafV600E, Ptennull, Cdkn2dnull) were transfected with myc-tagged RAC1P29S (Fig. 4A). YUMM1.7 cells that expressed RAC1P29S were significantly more sensitive to both dinaciclib and the CDK9 specific inhibitor NVP2 (Fig. 4B,C). To confirm this sensitivity was indeed due to CDK9 inhibition we knocked out CDK9 using CRISPR-Cas9 (Fig. 4D). Either Cas9 plus sgCDK9 or Cas9 alone were electroporated into YUMM1.7 or YUMM1.7 plus RAC1P29S cells. 48 hours post electroporation, YUMM1.7 cells expressing RAC1P29S had a significantly higher percent of dead cells compared to those that did not express mutant RAC1 (Fig. 4E). After 120 hours, crystal violet staining showed RAC1P29S expressing YUMM1.7 melanoma cells were no longer viable, further implicating CDK9 as a vital protein in RAC1P29S-expressing cells (Fig. 4F).
CDK9 is a non-canonical CDK, which has a primary function in transcriptional regulation through phosphorylation of Serine 2 on the C terminal domain (CTD) tail of RNA polymerase II (16, 17). In cancer, aberrant activity of CDK9 promotes transcription of genes that facilitate cell survival, such as those that promote cell cycle progression (18). To confirm that sensitivity of cells harboring a RAC1P29S mutation is transferrable across multiple melanoma cell lines and genetic backgrounds, we examined CDK9 sensitivity across several human melanoma lines, including those that harbor endogenous RAC1P29S mutations. The melanoma cells bearing P29S mutations were significantly more sensitive to dinaciclib and NVP2 (Fig. 4G-H).
We also tested the effects of a BRD4 inhibitor, as active CDK9 and Cyclin T form the positive transcription elongation factor b (P-TEFb) complex, which associates with bromodomain-containing protein 4 (BRD4) to activate RNA polymerase II transcriptional elongation (19). We found that human melanoma lines harboring a RAC1P29S mutation are also more sensitive to inhibition of BRD4 with JQ-1 (Fig. 4I).
Although CDK9 does not primarily regulate the cell cycle as a classic CDK, previous studies have linked CDK9 with promotion of transcription of G2/M genes (18). Accordingly, melanocytes trend toward G2/M arrest after CDK9 inhibition with dinaciclib or NVP2 for 16 hours, with a lower dose necessary for G2/M arrest in RAC1P29S-expressing cells (Supplemental Fig. 3A-B).
These data suggest that RAC1P29S induces molecular changes that lead to increased dependency on CDK9, presenting a potential therapeutic target in this subset of melanoma. Additionally, the cell cycle and G2/M signature seen in the RNA-seq and MIBs/MS datasets may be influenced by CDK9 modulation of RNA polymerase II transcribed gene regulation.
CDK9 inhibition increase surface expression of PD-L1 and Class I MHC in RAC1 mutant melanocytes
Several recent studies have reported pharmacologic inhibition of CDKs to regulate PD-L1 expression and sensitize the tumor microenvironment to immunotherapy (20–26). Given the unique sensitivity of RAC1P29S mutant melanoma cells to CDK9 inhibition, we next explored the consequences of CDK9 inhibition on cellular immunogenicity. Dinaciclib and NVP2 both decreased phosphorylated RNA polymerase II at serine 2 (Fig. 5A,D). Following dinaciclib treatment, both surface expression of PD-L1 and major histocompatibility complex (MHC) Class I are significantly more elevated in P29S-expressing melanocytes (Fig. 5B). Cells treated with NVP2 also exhibited an increase in surface PD-L1 and MHC Class I expression (Fig. 5C). The same increase in PD-L1 and MHC Class I was also seen upon dinaciclib treatment in YUMM1.7 plus RAC1P29S melanoma cell lines (Fig. 5E) and upon NVP2 treatment (Fig. 5F).
MHC class I is found on the cell surface of all nucleated cells and plays an instrumental role in antigen presentation and T-cell activation. Upregulation of MHC Class I could lead to more antigen presentation and a more robust anti-tumor immune response. These results indicate CDK9 inhibition could increase the immunogenicity of the tumor microenvironment and potentially enhance immune checkpoint inhibition response.
Combination therapy of CDK9 inhibition and anti-PD-1 immunotherapy significantly decreases tumor growth in vivo
As our in vitro data indicated CDK9 inhibition as a potential enhancer of immune checkpoint inhibition, we next tested the efficacy of CDK9 inhibition with anti-PD-1 ICI in a syngeneic murine model. Subcutaneous (s.c.) tumors were established in immune competent C57BL/6 mice with YUMM1.7 (BrafV600E, Ptennull, Cdkn2dnull) melanoma cells, or YUMM1.7 cells constitutively expressing the RAC1P29S mutation. Once tumors reached approximately 100 mm3, mice were randomly allocated to 4 treatment groups and either treated with vehicle (20% (2-Hydroxypropyl)-β-cyclodextrin) + IgG isotype control, 30 mg/kg dinaciclib, 10 mg/kg anti-PD-1 Ab, or a combination of dinaciclib plus anti-PD-1 Ab. Dinaciclib was treated every 3 days, while anti-PD-1 Ab was administered every 4 days (Fig. 6A). YUMM1.7 tumors did not significantly respond to either monotherapies or the combination therapy (Fig. 6B). Tumor growth inhibition (TGI) of dinaciclib alone was 1%, anti-PD1 antibody alone was 0%, and combination treatment was 16%. Contrastingly, YUMM1.7 tumors that expressed RAC1P29S were significantly repressed with treatment of either monotherapies and overall inhibited by combination treatment (Fig. 6C). Dinaciclib alone had a TGI of 72%, anti-PD-1 Ab alone had a TGI of 66%, and dinaciclib and anti-PD-1 Ab combination had a TGI of 97%, with one complete responder. YUMM1.7 tumor response greatly varied to dinaciclib and anti-PD1 Ab combination, while YUMM1.7 + RAC1P29S tumors were all suppressed, with 4/5 tumors smaller than their size at the beginning of drug administration (Fig. 6F,G).
RAC1P29S expression increases tumor immune cell infiltration
To determine the effect of the treatment regimen on tumor immune cell infiltration, we performed immunohistochemistry (IHC) on tumors fixed from the mice at the experiment endpoint. Overall, there was more T cell (CD8) and myeloid (CD11c) immune cell infiltration in RAC1P29S-expressing melanomas and expression of these was further increased upon treatment with dinaciclib (Fig. 7A,B). Additionally, there was increased CD86 expression in the RAC1 mutant tumors, which is found on monocytes, activated B cells, and dendritic cells and is necessary for T cell activation (27) (Fig. 7C). In contrast, in parental YUMM 1.7 grafts, neither dinaciclib nor anti-PD-1 Ab elicited an increase in CD8, CD11c, or CD86 (Fig. 7A-C). Surprisingly, there was no significant difference in caspase-3 positivity in control vs. treated tumors, suggesting that either these cells cannot proliferate or that a non-apoptotic mode of programmed cell death is evoked by combined anti-CDK9 and anti-PD1 therapy.