Rac and Cdc42 are small GTPases that have been linked to multiple human cancers and are implicated in epithelial to mesenchymal transition, cell-cycle progression, migration/invasion, tumor growth, angiogenesis, and oncogenic transformation. With the exception of the P29S driver mutation in melanoma, Rac and Cdc42 are not generally mutated in cancer, but are overexpressed (gene amplification and mRNA upregulation) or hyperactivated. Rac and Cdc42 are hyperactivated via signaling through oncogenic cell surface receptors, such as growth factor receptors, which converge on the guanine nucleotide exchange factors that regulate their GDP/GTP exchange. Hence, targeting Rac and Cdc42 represents a promising strategy for precise cancer therapy, as well as for inhibition of bypass signaling that promotes resistance to cell surface receptor-targeted therapies. Therefore, an understanding of the regulatory mechanisms of these pivotal signaling intermediates is key for the development of effective inhibitors. In this review, we focus on the role of Rac and Cdc42 in cancer and summarize the regulatory mechanisms, inhibitory efficacy, and the anticancer potential of Rac- and Cdc42-targeting agents. Cancer Res; 78(12); 3101–11. ©2018 AACR.

The homologous Rho GTPases Rac and Cdc42 play a pivotal role in cancer malignancy via regulation of cytoskeletal and microtubule dynamics, migration/invasion, metastasis, epithelial to mesenchymal transition, transcription, cell proliferation, cell-cycle progression, cell polarity, apoptosis, phagocytosis, vesicle trafficking, angiogenesis, and cell–cell and cell–extracellular matrix adhesions. Rho GTPases act as key molecular switches by alternating between their active GTP-bound form and their inactive GDP-bound form, where the exchange of GDP to GTP is catalyzed by guanine nucleotide exchange factors (GEF), whereas GTP hydrolysis is regulated by GTPase-activating proteins (GAP; Fig. 1; refs. 1, 2). A number of GEFs have been identified as oncogenes and are activated by oncogenic cell surface receptor signaling from G-protein–coupled receptors, growth factor receptors, cytokine/janus kinase/STAT receptors, and integrins. Rho GTPase activity can be further regulated by guanine nucleotide dissociation inhibitors (GDI), which prevent GEF-mediated nucleotide exchange, thereby maintaining the GTPase in an inactive state. GDIs can also bind the GTP-bound state of the GTPase and prevent nucleotide hydrolysis. The molecular mechanisms and regulatory role of GEFs, GAPs, and GDIs in Rac and Cdc42 function have been extensively reviewed (3, 4). Therefore, this review will focus on the therapeutic potential and current inhibitors available for Rac and Cdc42 targeting in cancer.

Figure 1.

Targeting Rac and Cdc42. Currently available inhibitors target Rac and Cdc42 activation by disrupting GEF interactions, inhibiting nucleotide binding, blocking lipid modifications, and modulating Rho GDIs and proteasomal degradation, as well as by inhibiting downstream effector activity.

Figure 1.

Targeting Rac and Cdc42. Currently available inhibitors target Rac and Cdc42 activation by disrupting GEF interactions, inhibiting nucleotide binding, blocking lipid modifications, and modulating Rho GDIs and proteasomal degradation, as well as by inhibiting downstream effector activity.

Close modal

As reviewed recently in this journal, a large number of in vitro studies have implicated the Rac isoforms Rac1, Rac2 (in hematopoietic cells), and Rac3, and the homolog Cdc42 in human cancer, including an essential role in Ras-mediated transformation (1). Table 1 shows a survey of The Cancer Genome Atlas (TCGA) data using cBioPortal (5), where RAC1 is upregulated in >10% of cancers with high mortality rates, including bladder, skin, esophageal, gastric, head and neck, liver, pancreatic, prostate, and uterine carcinomas, glioblastoma, mesothelioma, and sarcomas. The distribution of Rac mutations in cancer has been described, which includes the driver mutation RAC1(P29S) (∼5% in melanomas) and a constitutively active splice variant Rac1b (1). CDC42 is not usually mutated but approximately 5% elevated in most cancers with the exception of cervical squamous carcinoma, pancreatic adenocarcinoma, and sarcoma, where CDC42 is upregulated by 12%, 21%, and 14%, respectively. Therefore, targeting Cdc42 is also considered a viable option for cancer therapy (6). Although the analysis of breast invasive carcinomas demonstrated only modest percentages of elevated RAC1 (∼5%) and CDC42 (∼1%), a more in-depth analysis reported RAC1 upregulation in approximately 50% of HER2-enriched and basal breast-invasive carcinoma, including association of high RAC1 expression with poor patient survival (7).

Table 1.

RAC and CDC42 alterations in cancer

Cancer typeRACCDC42Source
Bladder urothelial carcinoma 9% 4% TCGA, Provisional 
 12% 7% (103) 
Breast invasive carcinoma 4.5% 1.7% TCGA, Provisional 
 4% 0.1% (104) 
Cervical squamous cell carcinoma 9% 12% TCGA, Provisional 
Colorectal adenocarcinoma 5% 0% TCGA, Provisional 
Cutaneous melanoma 30% (3.6% P29 Mut) 6.3% TCGA, Provisional 
Esophageal carcinoma 15% 4% TCGA, Provisional 
Glioblastoma 24%  (105) 
Stomach adenocarcinoma 3.3% 1% TCGA, Provisional 
 17% 4% (106) 
Head and neck squamous cell carcinoma 11.7% (0.7% Mut) 4% TCGA, Provisional 
 9% (1% Mut) 5% (107) 
Hepatocellular carcinoma 10% 4% TCGA, Provisional 
Lung adenocarcinoma 6% 1% TCGA, Provisional 
Mesothelioma 14% 4% TCGA, Provisional 
Pancreatic adenocarcinoma 6% 2.7% TCGA, Provisional 
 21% 21% (108) 
Prostate adenocarcinoma 9% 4% TCGA, Provisional 
 10% 5% (109) 
Sarcoma 14.5% 14% TCGA, Provisional 
Thyroid carcinoma 5% 3% TCGA, Provisional 
Uterine carcinosarcoma 9% 17.5% TCGA, Provisional 
Cancer typeRACCDC42Source
Bladder urothelial carcinoma 9% 4% TCGA, Provisional 
 12% 7% (103) 
Breast invasive carcinoma 4.5% 1.7% TCGA, Provisional 
 4% 0.1% (104) 
Cervical squamous cell carcinoma 9% 12% TCGA, Provisional 
Colorectal adenocarcinoma 5% 0% TCGA, Provisional 
Cutaneous melanoma 30% (3.6% P29 Mut) 6.3% TCGA, Provisional 
Esophageal carcinoma 15% 4% TCGA, Provisional 
Glioblastoma 24%  (105) 
Stomach adenocarcinoma 3.3% 1% TCGA, Provisional 
 17% 4% (106) 
Head and neck squamous cell carcinoma 11.7% (0.7% Mut) 4% TCGA, Provisional 
 9% (1% Mut) 5% (107) 
Hepatocellular carcinoma 10% 4% TCGA, Provisional 
Lung adenocarcinoma 6% 1% TCGA, Provisional 
Mesothelioma 14% 4% TCGA, Provisional 
Pancreatic adenocarcinoma 6% 2.7% TCGA, Provisional 
 21% 21% (108) 
Prostate adenocarcinoma 9% 4% TCGA, Provisional 
 10% 5% (109) 
Sarcoma 14.5% 14% TCGA, Provisional 
Thyroid carcinoma 5% 3% TCGA, Provisional 
Uterine carcinosarcoma 9% 17.5% TCGA, Provisional 

NOTE: Percentage amplifications, mRNA upregulations, and driver mutations (as computed from cBioPortal; ref. 102).

The contribution of Rac and Cdc42 to cancer initiation and progression is considered to be high due to their central roles in oncogenic cell surface receptor and GEF signaling. Many members of the largest family of Rho GEFs, the Dbl homology (DH) family (∼70), have been identified as oncogenes (1). In addition, the dedicator of cytokinesis (DOCK) family of GEFs (11 members) that are structurally different from the DH domain Rac/Cdc42 GEFs has also been implicated in cancer (8). As has been extensively reviewed, deregulation of oncogenic GEFs such as Dbl, Vav, Trio, T-cell invasion and metastasis gene product (Tiam-1), epithelial cell transforming gene 2 (Ect2), and phosphatidylinositol-3,4,5-trisphosphate (PIP3)–dependent Rac exchange factor 1 (P-Rex-1) contributes to aberrant Rac and Cdc42 activity in cancer (1, 3, 4). An additional oncogenic effector of Rac and Cdc42 is PI3K. PI3K activates Rac and Cdc42 via activation of PIP3-regulated GEFs such as P-Rex, Vav, Sos, and SWAP70 (9). Therefore, the PI3K catalytic subunit (PIK3CA) mutations result in hyperactivation of Rac and Cdc42 GEFs, and hence, elevated Rac1 and Cdc42 signaling (9). In addition, Cdc42 has been shown to regulate cancer via activated Cdc42-associated tyrosine kinase (Ack1), Ras, and EGFR. The oncogene Ack1, which is activated by Cdc42 via EGFR/HER2, regulates Akt-mediated survival signaling to result in tumor recurrence and therapy resistance (10). Accordingly, inhibition of Cdc42 in Ras-transformed cells was shown to decrease oncogenic signaling via Akt, thus contributing to reduced cancer malignancy (11). Studies have also shown that Cdc42 prevents EGFR degradation by blocking receptor ubiquitination (12). Therefore, Cdc42 inhibitors have the potential to chemosensitize cancer therapies targeting Ras and EGFR.

Rac and Cdc42 may be activated by shared GEFs such as Vav and Ect2, Rac-specific GEFs (e.g., Tiam-1, p-Rex1), or Cdc42-specific GEFs [e.g., Intersectin, FYVE, RhoGEF, and PH domain containing protein1 (FGD1)] to activate a number of oncogenic signaling pathways. Direct downstream effectors of Rac and Cdc42 include p21-activated kinases (PAK), IQ motif containing GTPase-activating protein, Wiskott Aldrich Syndrome protein (WASP), WASP family verprolin-homologous protein and mammalian enabled (Mena)/vasodilator-stimulated phosphoprotein, and p67phox complexes. These effectors regulate migration/invasion via de novo actin polymerization, cell polarization, and matrix metalloproteinase secretion (13). PAK signaling via Rac and Cdc42 has been extensively studied in cancer and shown to regulate Src, focal adhesion kinase (FAK), PI3-K/Akt/mTOR, MAPKs [ERK, jun kinase (JNK), and p38 MAPK], protein kinase C, and STATs (14). Activated Rac has also been shown to affect cell proliferation via signaling to the oncogenes c-Myc and Cyclin D, as well as mTOR complex1 (mTORC1) and mTORC2 activation (15). Recent studies also suggest that nuclear Rho GTPases may have an additional role in regulating DNA damage response (16). Therefore, through these diverse downstream effectors, Rac and Cdc42 regulate tumor formation, growth, and metastasis, and are poised as new therapeutic targets for multiple aggressive cancers.

Rho GTPases have been previously considered “undruggable” due to their globular structure with limited small-molecule binding pockets, high affinity for GTP or GDP binding, and the micromolar levels of GTP available in cells. The complexity of Rac and Cdc42 downstream signaling pathways also confounds the challenge of targeting a particular cellular process. Nonetheless, several rational strategies have emerged to inhibit the activation of Rac and Cdc42 (summarized in Fig. 1). Of these current approaches, blocking interaction with GEFs (Table 2) or nucleotide binding (Table 3) has emerged as principal strategies for Rac and Cdc42 inhibition in cancer cells and mouse models.

Table 2.

GEF-interacting inhibitors of Rac and Cdc42 in cancer: compounds tested in cancer models

GEF-interacting inhibitors of Rac and Cdc42 in cancer: compounds tested in cancer models
GEF-interacting inhibitors of Rac and Cdc42 in cancer: compounds tested in cancer models
Table 3.

Nucleotide binding inhibitors of Rac and Cdc42 in cancer: compounds tested in cancer models

Nucleotide binding inhibitors of Rac and Cdc42 in cancer: compounds tested in cancer models
Nucleotide binding inhibitors of Rac and Cdc42 in cancer: compounds tested in cancer models

GEF interaction inhibitors

Rho GTPases are activated through exchange of GDP for GTP via GEFs, which not only facilitates interaction with downstream effectors, but also dictates the specificity of the signaling cascades that are activated (1, 17). Using structure-based mutagenesis studies, the Trp56 residue located on the groove formed by the switch I, switch II, and the β1/β2/β3 regions of Rac was identified as a critical determinant for GEF binding (18). The small-molecule NSC23766 was identified from a structure-based virtual screen of compounds that fitted into the surface groove of Rac1 critical for the Tiam1 and Trio GEF interaction (19). Confirming the oncogenic role of Tiam1 and Trio in cancer (3), NSC23766 reduced growth and invasion in several cancer types, including prostate, breast, gastric, chronic myelogenous leukemia, anaplastic large cell lymphoma, and glioblastoma (19–24). However, off-target effects in mouse platelets, such as receptor downregulation (25), as well as the high IC50 (∼50 μmol/L) of NSC23766 make it ineffective for pharmacologic use.

Our group used NSC23766 as a template for the design of derivatives, maintaining the central pyrimidine core of NSC23766 that binds the critical Tryp56 in Rac, and identified the Rac inhibitor EHop-016 (26). In metastatic cancer cells, EHop-016 blocks the interaction of the oncogene Vav with Rac and inhibits Rac activation with an IC50 of approximately 1 μmol/L, and Cdc42 at ≥10 μmol/L, without affecting Rho (14, 27). Moreover, EHop-016 reduced mammary tumor growth by approximately 80% in nude mice and inhibited angiogenesis and metastasis (28). The efficacy of EHop-016 as a Rac inhibitor has been subsequently validated in breast and prostate cancer cell lines, leukemia, melanoma, T lymphocytes, and fibrosarcoma (27, 29–32).

However, the relatively high effective concentrations and the moderate bioavailability (∼30% with t1/2 of 4.5 hours) of EHop-016 (33) needed improvement. From screening EHop-016 derivatives, we found that this class of compounds was most active when the carbazole fragment was connected to a central core of 1,5,-disubstituted 1,2,3-triazoles (14) and therefore, synthesized MBQ-167 to retain this carbazole core (34). Although EHop-016 adopts a U-shaped conformation into a binding pocket adjacent to the Trp56 of Rac, it is predicted to interact more closely with Asn39 and Asp38 in the switch I region of Rac (27). Similarly, in silico modeling predicted that MBQ-167 binds deeper into the putative binding pocket of Rac and Cdc42, forming H bonds with the side-chain of Asn39. As predicted by studies where substitution of Asn39 of both Rac and Cdc42 results in the loss of GEF binding (18), MBQ-167 inhibits both Rac and Cdc42 activation. MBQ-167 inhibits Rac 1/2/3 activity with an IC50 of 103 nmol/L and Cdc42 activity with an IC50 of 78 nmol/L, thus making MBQ-167 one of the most potent Rac and Cdc42 inhibitors currently described in the literature. In immunocompromised mouse models, MBQ-167 inhibited metastatic breast cancer growth by approximately 90% saturating at 1 mg/kg body weight (34). Our studies with EHop-016 and MBQ-167 have validated the hypothesis that inhibition of Rac and Cdc42 activation leads to reduced metastatic cancer cell viability, migration, tumor growth, metastasis, and angiogenesis (28, 34). Moreover, in addition to reducing tumor growth, these drugs also prevent the infiltration of tumor-associated macrophages and neutrophils as well as cytokine release (unpublished data), which signifies additional beneficial effects of Rac and Cdc42 inhibition in the tumor microenvironment.

Other NSC23766 derivatives, which have been developed to retain the central pyrimidine core, such as AZA1 and AZA197, are not as effective as EHop-016 or MBQ-167. AZA1 inhibits Cdc42 and Rac1 activation at concentrations ranging from 5 to 20 μmol/L and reduces migration and downstream signaling through PAK in prostate cancer cells and decreases tumor growth in mice (35). AZA197, a Cdc42-selective small-molecule inhibitor, is also active at an IC50 of 1 to 10 μmol/L and inhibits the Cdc42-GEF Dbs activity. Accordingly, AZA-197 treatment decreased PAK and ERK activities, cyclin D expression, and colon cancer cell invasion and proliferation, as well as tumor growth in mice (36, 37). Another molecule that inhibits Cdc42–Dbs interaction with an IC50 of 67 μmol/L is Compound 19 (38). However, this compound has not been analyzed in cell or animal models.

Ferri and colleagues screened a ZINC database for Rac inhibitors and identified a range of compounds with IC50s from 12 to 57 μmol/L. From this screening, compounds 4 and 5, ZINC08010136 (IC50, 12.2 μmol/L), and ZINC07949036 (IC50, 24.1 μmol/L) were selected for further testing and shown to interfere with the Tiam-1/Rac1 interaction (39, 40). The same research group subsequently published a series of small molecules also targeting Tiam-1/Rac activation with IC50s ranging from 2.5 to 27.9 μmol/L (41, 42). Although these compounds have yet to be tested in cancer cells, they are expected to be active in cancer models.

Another docking-based screening approach identified ZINC69391 as a structurally distinct compound from NSC23766 that still interferes with the Rac1–Tiam1 interaction with an IC50 of 61 μmol/L. This compound was tested in metastatic breast cancer cell lines and mouse models and inhibited cell proliferation, cell-cycle progression, migration, and metastasis (43). A more potent analog of ZINC69391, 1A-116, was also described by the same group and blocks the P-Rex1/Rac1 interaction with an IC50 of 4.0 μmol/L. Both ZINC69391 and 1A-116 have also been analyzed in breast cancer, glioma, and leukemia cells yielding antiproliferative and anti-invasive effects, indicating the therapeutic potential of inhibiting Rac activation in multiple aggressive cancers (43–45). The 1A-116 was also recently shown to chemosensitize tamoxifen-resistant breast cancer cells, thus demonstrating the utility of Rac inhibition to overcoming therapy resistance (46).

Although these compounds inhibited the Tiam-1/Rac interaction, the small-molecule compound ITX3 was shown to interfere selectively with the related GEF TrioN binding to Rac at concentrations ranging from 50 to 100 μmol/L. ITX3 is an analog of ITX1 (IC50 110 μmol/L), identified through a chemical library screen in yeast cells using GEF activity assays. Even though it showed higher potency than ITX1, the relatively high IC50 of ITX3 still limits its clinical applicability (47).

As expected from the structural similarity to GTP, 6-Thio-GTP, a metabolite of Azathioprine that is used as an immunosuppressive agent for the treatment of inflammatory diseases, competitively blocks Rho-GEF binding. 6-Thio-GTP has been shown to inhibit Rac1 activation by Vav in lymphocytes and breast cancer cells (48, 49).

Most current inhibitors of the GEF/Rac/Cdc42 interaction focus on preventing the Dbl family GEFs. However, the small-molecule inhibitor CPYPP interferes with the DOCK family GEF DOCK2, which is predominantly expressed in hematopoietic cells (50). A DOCK2-selective inhibitory peptide has also been shown to block B-cell migration and is considered a viable drug target in leukemia (51). In addition, the DOCK1 selective inhibitor TBOPP was shown to specifically inhibit DOCK1-regulated invasion in Ras-transformed cells and tumor growth and metastasis in mice with tumors from Ras-mutant lung carcinoma cells (52), thus illustrating the utility of DOCK-targeted anticancer drugs.

Structural studies with Cdc42 have revealed that similar to the critical Trp 56 in Rac1, Phe56 in Cdc42 is a significant determinant for GEF binding. Hence, Pirl1 was discovered through a biochemical suppression approach from cytoplasmic extracts of Xenopus laevis eggs. Pirl1 targets Cdc42 by specifically blocking PIP2-mediated GEF activity on the Cdc42/RhoGDI complex with an IC50 of 3 μmol/L (53). Cdc42 activity–specific inhibitor (CASIN), a structural derivative of pirl1, also disrupts intersectin (ArhGEF4)-mediated Cdc42 activation with a similar IC50 of 2 μmol/L. Using CASIN, Cdc42 was shown to regulate colon cancer initiation by interacting with the pivotal tumor suppressor adenomatous polyposis coli in incipient intestinal tumor cells. Accordingly, Cdc42 inhibition via CASIN treatment reduced in vivo tumorigenicity in colorectal cancer xenograft models and abrogated progression of mouse and human tumor organoids (54). Similarly, the small-molecule ZCL278, discovered through a high-throughput in silico screening, binds close to Phe56 in the surface groove of Cdc42 and disrupts the interaction between Intersectin and Cdc42. ZCL278 also inhibited migration in metastatic prostate cancer cells (55). Therefore, the screening for Cdc42-specific inhibitors using the unique Phe56 in its GEF-binding groove has led to the elucidation of Cdc42-specific functions driving cancer initiation and progression.

Nucleotide binding inhibitors

Unlike the GEF-binding inhibitors, which usually block specific classes of GEFs or individual GEFs from binding to the GEF-interacting domains of Rac and (or) Cdc42, a viable alternative is to prevent nucleotide binding. This strategy was used to develop EHT 1864, which inactivates Rac isoforms (1,2,3, and the Rac1b splice variant) by a tight interaction that displaces guanine nucleotides. Therefore, EHT 1864 inhibits upstream GEFs as well as downstream effector binding at concentrations ranging from 10 to 50 μmol/L (56). Studies have used EHT 1864 to elucidate the central role of Rac in transformation (via Tiam-1 or Ras), downregulation of estrogen receptor expression in breast tumors, migration in fibrosarcoma and melanoma cells, and breast cancer invasion and tumor growth (56–61). However, off-targets effects of this compound have been observed in mouse platelets, such as triggering of platelet apoptosis (25), thus raising concerns about its safety.

MLS000532223 was characterized using a flow cytometry bead-based multiplex assay for molecules that inhibit Rho family activation by noncompetitive inhibition of GTP binding. MLS0005322233 inhibits all Rho GTPases tested (Rho, Rac, and Cdc42), whereas the related compound MLS000573151 is specific for Cdc42 and affects the cytoskeletal dynamics of mast cells and leukemia cells. However, these compounds are active at approximately 10 μmol/L, thus limiting their pharmacologic use (62).

Other compounds that interfere with Rac1 nucleotide binding are compounds 1 and 6. These compounds disrupt the binding between Rac1 and its effector PAK1 and reduce cell proliferation and migration in various pancreatic cancer cell lines with EC50s of 8.3 and 22.4 μmol/L, respectively (63). The reported IC50s for these compounds are in the nanomolar range as determined by an in vitro assay using purified recombinant proteins. Hence, further studies are needed to confirm their efficacy in cell-based systems.

A series of isoquinolines and phenanthridine derivatives were also developed to block nucleotide binding with Rac1, Cdc42, and Rac1b. Efficient Rac1 and Cdc42 inhibition was induced by compound 4 with IC50s of 8 and 6 μmol/L for Rac1 and Cdc42, respectively (64). Intriguingly, this compound was more effective at inhibiting the constitutively active splice variant Rac1b (IC50 = 2.7 μmol/L), and thus hold utility as therapy for lung, thyroid, breast, and colorectal cancers with upregulated Rac1b (65–67).

There are several nucleotide-binding inhibitors that specifically block Cdc42 activity. The structural analogs CID2950007 (ML141) and CID44216842 inhibit Cdc42 activation with EC50s of approximately 2 and 1 μmol/L, respectively, and were discovered through a bead-based multiplex flow cytometry assay (68). CID2950007 reduced bradykinin-induced filopodia formation in fibroblasts, thus indicating specific inhibition of Cdc42 activation (69). These compounds inhibited ovarian cancer cell migration, without being cytotoxic in multiple cell lines (68). Even though CID44216842 is more potent, its low solubility limits its further development as an anticancer agent (68).

Recent studies have also demonstrated that the R-enantiomers of nonsteroidal anti-inflammatory drugs function as allosteric inhibitors of Rac1 and Cdc42. R-ketorolac and R-naproxen inhibited Rac1 and Cdc42 at micromolar IC50s (70) with R-ketorolac inhibiting Rac with an EC50 of 0.574 μmol/L and Cdc42 at 1 μmol/L, and consequently reducing ovarian cancer cell migration, invasion, and adhesion (71). Moreover, R-ketorolac reduced tumor development in a spontaneous breast cancer transgenic mouse model (72). This FDA-approved drug is the first Rac and Cdc42 inhibitor to be used in a P0 clinical trial, which demonstrated improved ovarian cancer patient survival (71).

Another FDA-approved drug is mitoxantrone (MTX), a topoisomerase II inhibitor, which is currently used as a cancer therapeutic. A GTPase activity AlphaScreen assay identified MTX to interfere with GTP binding with broad selectivity, targeting RhoA, Rac1, and Cdc42, and thus reduced F-actin reorganization and cell migration (73). Because this drug is known to have adverse side effects, inhibition of Rho GTPases may add to its overall off-target effects.

Spatial regulation inhibitors

Rho GTPases require to be localized to cell membranes for activation by receptor-regulated GEFs. Therefore, posttranslational modifications that target them to the plasma membrane, such as addition of isoprenoid moieties (prenylation) at the C-terminal “CAAX box,” are essential for their biological functions (74). Hence, similar to the strategy of inhibiting Ras activation with farnesyltransferase inhibitors, geranylgeranyl transferase inhibitors, such as PAIA6, have been used to alter Rho GTPase function and induce anticancer properties in various model systems (75, 76). Although these inhibitors are not specific to Rac and (or) Cdc42, testing of prenylation inhibitors such as GGTI-2418 in preclinical and clinical trials has shown multiple anticancer effects.

Similarly, because statins lower lipid synthesis by inhibiting HMG-CoA reductase, they can also be used to prevent isoprenoid synthesis and thus prenylation of Rho GTPases; however, this strategy is also limited by its nonselectivity. Moreover, alternate mechanisms have also been associated with statins, such as increased nuclear Rac1 degradation (77). Although statins have been shown to have antitumor properties (78–80), further studies are needed to determine the specificity of such inhibition.

RhoGDI modulators

Another potential strategy for Rac and Cdc42 inhibition is targeting of RhoGDIs. RhoGDIs sequester inactive GDP-bound Rho GTPases in the cytosol and inhibit their activation. Hence, the design of an interfacial interactor to prevent the release of a RhoGDI from a Rho GTPase is a potential inhibitory strategy (17). Thus, Secramine was developed to block membrane recruitment of prenylated Cdc42 by sequestering the Cdc42.RhoGDI complex, and thus inhibit actin polymerization (81). However, this strategy may not be selective, because RhoGDI also binds Rac and Rho. Moreover, GDIs play a complex role in cancer where both up- and downregulation of Rho GDIs have been shown to result in increased malignancy (17).

GAP modulators

Because GAPs catalyze the hydrolysis of GTP to GDP, increasing Rho GAP activity is a potential mechanism for Rho GTPase inactivation. As expected, GAPs have been shown to act as tumor suppressors; however, some GAPs are also overexpressed in certain cancers, thus confounding the development of GAP activators as anticancer agents (17). Nevertheless, a number of Rac and Cdc42 GAPs, such as β2-chimaerin, ARHGAP24, and Binder of Arl Two (BART), have been shown to exert a negative effect on cancer progression (1, 4, 82). However, so far, no mimics of GAPs have been developed for use as Rac and Cdc42 inhibitors.

Modulators of protein stability

Rac and Cdc42 stability can be regulated by SUMOYlation, which increases stability of the GTP-bound form, and ubiquitination, which targets them for proteasomal degradation (83). Therefore, inhibition of SUMOylation or increased ubiquitination is rational strategy to inhibit Rac and Cdc42. Luteolin is a flavonoid that promotes proteasomal degradation of Cdc42 and thus reduces invasion and migration of glioblastoma cells (84). Luteolin has also been tested in vitro and in vivo in various cancers, such as leukemia and prostate cancer, and was effective in decreasing cell growth and angiogenesis, and increasing apoptosis (85). Nonetheless, no further advancements have been made in the development of this compound as an anticancer therapeutic.

Downstream effector inhibitors

Much attention has been devoted to the prevention of downstream signaling from Rac and Cdc42 by targeting onco proteins such as PAKs. The status of the current PAK inhibitors, such as OSU-03012 (86), FL172 (87), and FRAX597 (88), has been extensively reviewed (89). Unfortunately, the PAK4 inhibitor PF-3758309 failed clinical trials, due to its undesirable pharmacokinetics and dose–response relationship, and a PAK inhibitor has yet to receive FDA approval.

Other downstream effector inhibitors, such as Phox-I1 and 8-hydroxy-2-deoxyguanine, which target the binding between Rac1 and its downstream effector p67phox, a structural component of the NADPH oxidase complex, have been shown to inhibit cancer progression (90). Further studies are needed to validate the clinical applicability of these compounds.

Efforts are also underway to target the interaction of Cdc42 with downstream effector N-WASP, thus inhibiting the activation of the actin-related protein (Arp)2/3 complex and ensuing actin polymerization. Potential of inhibitors, such as the 14-aminoacid cyclic peptide 187-1 and wiskostatin, as anticancer compounds remains to be recognized (91, 92).

Rac and Cdc42 GTPases are key signaling intermediates whose dysregulation has been distinctly associated with cancer initiation and progression. Rational design of small-molecule inhibitors to Rac and Cdc42 has shown promising preclinical outcomes. Nonetheless, no clinically effective drugs targeting these Rho GTPases have been approved for cancer therapy, and nor are they currently in clinical trials. Testing these inhibitors, in patient-derived orthotopic xenograft mouse models of different cancer types, is also essential for demonstrating drug efficacy in a physiologically relevant setting (93–96). Furthermore, studies regarding the toxicity and pharmacokinetic properties of these inhibitors are scarce in the literature. Additional factors such as feedback mechanisms and impact on the tumor microenvironment, as well as effects on the immune and cardiovascular systems, need to be considered in further development of Rac and Cdc42 inhibitors as anticancer agents.

The central role of Rac and Cdc42 in multiple oncogenic signaling pathways also needs to be recognized regarding the therapeutic utility of Rac and Cdc42 inhibition in cancer. Rac and Cdc42 are pivotal downstream regulators of a myriad of cell signaling receptors, as well as Ras/MAPK and PI3-K/Akt pathways, for which a number of therapies exist or are currently undergoing clinical trials. However, acquired resistance to such targeted therapies is frequently a cause of failure in cancer treatment. Therefore, inhibition of Rac and Cdc42 may be a sound strategy to potentiate receptor- and PI3-K–targeted therapies.

Our studies with the Rac/Cdc42 inhibitors EHop-016 and MBQ-167 in breast cancer models (HER2 and triple-negative breast cancer) have shown that consistent with their role in suppression of cell growth and migration/invasion, Rac and Cdc42 inhibition does not reduce the size of the primary tumor but prevents its further growth and metastasis. Therefore, as illustrated by reports where Rac1 inhibition sensitized pancreatic and breast cancer cells to radiotherapy (97, 98), Rac and Cdc42 inhibitors may be useful in combination therapy with classic chemo- and radiotherapies.

Moreover, Rac/Cdc42/PAK signaling is specifically implicated with therapy resistance of HER2-type breast cancers. Accordingly, studies by our group and others have shown that Rac/Cdc42 inhibition decreases the viability of breast cancers resistant to EGFR/HER2 therapy (unpublished data and refs. 14, 24, 99, 100). Moreover, a recent study demonstrated the utility of using EHop-016 as combined therapy with Akt/mTOR inhibitors to treat Integrin-mediated high-grade myxofibrosarcoma (30). In addition, Rac/Cdc42 inhibition as a strategy to overcome therapy resistance has been validated using 1A-116, EHT 1864, and ML141 (46, 57, 101).

In conclusion, future therapeutic strategies should focus on the utility of Rac/Cdc42 inhibitors in combination therapy with existing cancer therapeutics, as well as to diminish cancer therapy resistance. Such combination therapies with novel targeting strategies, such as immunoliposomes to target overexpressed receptors, will facilitate the administration of lower concentrations of Rac and Cdc42 inhibitors, thus circumventing potential immune and cardiovascular toxicities. Considering their enormous potential for preventing not only tumor growth and metastasis, but also the tumor-promoting effects of the immune and stromal cells in the tumor microenvironment, Rac and Cdc42 inhibitors are poised for clinical testing to assess the risks and benefits of targeting Rac/Cdc42 in human cancer.

S. Dharmawardhane has ownership interest (including patents) in US patents US 08884006 B2 and US 9278956 B1. No potential conflicts of interest were disclosed by the other author.

M.d.M. Maldonado is supported by NIGMS-RISE R25 GM061838 to UPR MSC. S. Dharmawardhane is supported by NIH/NIGMS SC3GM084824, NIH/NCI U54CA096297, NIH/NIGMS P20 GM103475, Puerto Rico Science and Technology Research Trust, and Susan Komen for the Cure.

The authors acknowledge Gabriela Rosado-Gonzalez for contributing to the literature search for this review.

1.
Kazanietz
MG
,
Caloca
MJ
. 
The Rac GTPase in cancer: from old concepts to new paradigms
.
Cancer Res
2017
;
77
:
5445
51
.
2.
Stengel
K
,
Zheng
Y
. 
Cdc42 in oncogenic transformation, invasion, and tumorigenesis
.
Cell Signal
2011
;
23
:
1415
23
.
3.
Cook
DR
,
Rossman
KL
,
Der
CJ
. 
Rho guanine nucleotide exchange factors: regulators of Rho GTPase activity in development and disease
.
Oncogene
2014
;
33
:
4021
35
.
4.
Wertheimer
E
,
Gutierrez-Uzquiza
A
,
Rosemblit
C
,
Lopez-Haber
C
,
Sosa
MS
,
Kazanietz
MG
. 
Rac signaling in breast cancer: a tale of GEFs and GAPs
.
Cell Signal
2012
;
24
:
353
62
.
5.
Gao
J
,
Aksoy
BA
,
Dogrusoz
U
,
Dresdner
G
,
Gross
B
,
Sumer
SO
, et al
Integrative analysis of complex cancer genomics and clinical profiles using the cBioPortal
.
Sci Signal
2013
;
6
:
pl1
.
6.
Arias-Romero
LE
,
Chernoff
J
. 
Targeting Cdc42 in cancer
.
Expert Opin Ther Targets
2013
;
17
:
1263
73
.
7.
De
P
,
Carlson
JH
,
Jepperson
T
,
Willis
S
,
Leyland-Jones
B
,
Dey
N
. 
RAC1 GTP-ase signals Wnt-beta-catenin pathway mediated integrin-directed metastasis-associated tumor cell phenotypes in triple negative breast cancers
.
Oncotarget
2017
;
8
:
3072
103
.
8.
Gadea
G
,
Blangy
A
. 
Dock-family exchange factors in cell migration and disease
.
Eur J Cell Biol
2014
;
93
:
466
77
.
9.
Lien
EC
,
Dibble
CC
,
Toker
A
. 
PI3K signaling in cancer: beyond AKT
.
Curr Opin Cell Biol
2017
;
45
:
62
71
.
10.
Mahajan
K
,
Mahajan
NP
. 
ACK1 tyrosine kinase: targeted inhibition to block cancer cell proliferation
.
Cancer Lett
2013
;
338
:
185
92
.
11.
Stengel
KR
,
Zheng
Y
. 
Essential role of Cdc42 in ras-induced transformation revealed by gene targeting
.
PLoS One
2012
;
7
:
e37317
.
12.
Wu
WJ
,
Tu
S
,
Cerione
RA
. 
Activated Cdc42 sequesters c-Cbl and prevents EGF receptor degradation
.
Cell
2003
;
114
:
715
25
.
13.
Bishop
AL
,
Hall
A
. 
Rho GTPases and their effector proteins
.
Biochem J
2000
;
348 Pt 2
:
241
55
.
14.
Dharmawardhane
S
,
Hernandez
E
,
Vlaar
C
. 
Development of EHop-016: a small molecule inhibitor of Rac
.
Enzym
2013
;
33
:
117
46
.
15.
Saci
A
,
Cantley
LC
,
Carpenter
CL
. 
Rac1 regulates the activity of mTORC1 and mTORC2 and controls cellular size
.
Mol Cell
2011
;
42
:
50
61
.
16.
Fritz
G
,
Henninger
C
. 
Rho gtpases: novel players in the regulation of the DNA damage response?
Biomolecules
2015
;
5
:
2417
34
.
17.
Porter
AP
,
Papaioannou
A
,
Malliri
A
. 
Deregulation of Rho GTPases in cancer
.
Small GTPases
2016
;
7
:
123
38
.
18.
Gao
Y
,
Xing
J
,
Streuli
M
,
Leto
TL
,
Zheng
Y
. 
Trp56 of Rac1 specifies interaction with a subset of guanine nucleotide exchange factors
.
J Biol Chem
2001
;
276
:
47530
41
.
19.
Gao
Y
,
Dickerson
JB
,
Guo
F
,
Zheng
J
,
Zheng
Y
. 
Rational design and characterization of a Rac GTPase-specific small molecule inhibitor
.
Proc Natl Acad Sci U S A
2004
;
101
:
7618
23
.
20.
Yoshida
T
,
Zhang
Y
,
Rivera Rosado
L a
,
Chen
J
,
Khan
T
,
Moon
SY
, et al
Blockade of Rac1 activity induces G1 cell cycle arrest or apoptosis in breast cancer cells through downregulation of cyclin D1, survivin, and X-linked inhibitor of apoptosis protein
.
Mol Cancer Ther
2010
;
9
:
1657
68
.
21.
Thomas
EK
,
Cancelas
JA
,
Chae
HD
,
Cox
AD
,
Keller
PJ
,
Perrotti
D
, et al
Rac guanosine triphosphatases represent integrating molecular therapeutic targets for BCR-ABL-induced myeloproliferative disease
.
Cancer Cell
2007
;
12
:
467
78
.
22.
Colomba
A
,
Giuriato
S
,
Dejean
E
,
Thornber
K
,
Delsol
G
,
Tronchère
H
, et al
Inhibition of Rac controls NPM–ALK-dependent lymphoma development and dissemination
.
Blood Cancer J
2011
;
1
:
e21
.
23.
Ji
J
,
Feng
X
,
Shi
M
,
Cai
Q
,
Yu
Y
,
Zhu
Z
, et al
Rac1 is correlated with aggressiveness and a potential therapeutic target for gastric cancer
.
Int J Oncol
2015
;
46
:
1343
53
.
24.
Karpel-Massler
G
,
Westhoff
M-A
,
Zhou
S
,
Nonnenmacher
L
,
Dwucet
A
,
Kast
RE
, et al
Combined inhibition of HER1/EGFR and RAC1 results in a synergistic antiproliferative effect on established and primary cultured human glioblastoma cells
.
Mol Cancer Ther
2013
;
12
:
1783
95
.
25.
Dutting
S
,
Heidenreich
J
,
Cherpokova
D
,
Amin
E
,
Zhang
SC
,
Ahmadian
MR
, et al
Critical off-target effects of the widely used Rac1 inhibitors NSC23766 and EHT1864 in mouse platelets
.
J Thromb Haemost
2015
;
13
:
827
38
.
26.
Hernández
E
,
De La Mota-Peynado
A
,
Dharmawardhane
S
,
Vlaar
CP
. 
Novel inhibitors of rac1 in metastatic breast cancer
.
P R Health Sci J
2010
;
29
:
348
56
.
27.
Montalvo-Ortiz
BL
,
Castillo-Pichardo
L
,
Hernández
E
,
Humphries-Bickley
T
,
De La Mota-Peynado
A
,
Cubano
LA
, et al
Characterization of EHop-016, novel small molecule inhibitor of Rac GTPase
.
J Biol Chem
2012
;
287
:
13228
38
.
28.
Castillo-Pichardo
L
,
Humphries-Bickley
T
,
De La Parra
C
,
Forestier-Roman
I
,
Martinez-Ferrer
M
,
Hernandez
E
, et al
The Rac inhibitor EHop-016 inhibits mammary tumor growth and metastasis in a nude mouse model
.
Transl Oncol
2014
;
7
:
546
55
.
29.
Maes
H
,
Van Eygen
S
,
Krysko
DV
,
Vandenabeele
P
,
Nys
K
,
Rillaerts
K
, et al
BNIP3 supports melanoma cell migration and vasculogenic mimicry by orchestrating the actin cytoskeleton
.
Cell Death Dis
2014
;
5
:
e1127
12
.
30.
Okada
T
,
AY
L
,
LX
Q
,
Agaram
N
,
AM
C
,
Shima
F
, et al
Integrin-alpha 10 dependency identifies RAC and RICTOR as therapeutic targets in high-grade myxofibrosarcoma
.
Cancer Discov
2017
;
6
:
1148
65
.
31.
Martin
H
,
Mali
RS
,
Ma
P
,
Chatterjee
A
,
Ramdas
B
,
Sims
E
, et al
Pak and Rac GTPases promote oncogenic KIT-induced neoplasms
.
J Clin Invest
2013
;
123
:
4449
63
.
32.
Manes
TD
,
Pober
JS
. 
TCR-driven transendothelial migration of human effector memory CD4 T cells involves Vav, Rac, and myosin IIA
.
J Immunol
2013
;
190
:
3079
88
.
33.
Humphries-Bickley
T
,
Castillo-Pichardo
L
,
Corujo-Carro
F
,
Duconge
J
,
Hernandez-O'Farrill
E
,
Vlaar
C
, et al
Pharmacokinetics of Rac inhibitor EHop-016 in mice by ultra-performance liquid chromatography tandem mass spectrometry
.
J Chromatogr B Anal Technol Biomed Life Sci
2015
;
981–982
:
19
26
.
34.
Humphries-Bickley
T
,
Castillo-Pichardo
L
,
Hernandez-O'Farrill
E
,
Borrero-Garcia
LD
,
Forestier-Roman
I
,
Gerena
Y
, et al
Characterization of a dual Rac/Cdc42 inhibitor MBQ-167 in metastatic cancer
.
Mol Cancer Ther
2017
;
16
:
805
18
.
35.
Zins
K
,
Lucas
T
,
Reichl
P
,
Abraham
D
,
Aharinejad
S
. 
A Rac1/Cdc42 GTPase-specific small molecule inhibitor suppresses growth of primary human prostate cancer xenografts and prolongs survival in mice
.
PLoS One
2013
;
8
:
e74924
.
36.
Zins
K
,
Gunawardhana
S
,
Lucas
T
,
Abraham
D
,
Aharinejad
S
. 
Targeting Cdc42 with the small molecule drug AZA197 suppresses primary colon cancer growth and prolongs survival in a preclinical mouse xenograft model by downregulation of PAK1 activity
.
J Transl Med
2013
;
11
:
295
.
37.
Cromm
PM
,
Spiegel
J
,
Grossmann
TN
,
Waldmann
H
. 
Direct modulation of small GTPase activity and function
.
Angew Chemie
2015
;
54
:
13516
37
.
38.
Cummings
CG
,
Ross
NT
,
Katt
WP
,
Hamilton
AD
. 
Synthesis and biological evaluation of a 5-6-5 imidazole-phenyl-thiazole based a-helix mimetic
.
Org Lett
2009
;
11
:
25
8
.
39.
Ferri
N
,
Corsini
A
,
Bottino
P
,
Clerici
F
,
Contini
A
. 
Virtual screening approach for the identification of new Rac1 inhibitors
.
J Med Chem
2009
;
52
:
4087
90
.
40.
Ferri
N
,
Contini
A
,
Bernini
SK
,
Corsini
A
. 
Role of small GTPase protein Rac1 in cardiovascular diseases: development of new selective pharmacological inhibitors
.
J Cardiovasc Pharmacol
2013
;
62
:
425
35
.
41.
Ferri
N
,
Bernini
SK
,
Corsini
A
,
Clerici
F
,
Erba
E
,
Stragliotto
S
, et al
3-Aryl-N-aminoylsulfonylphenyl-1H-pyrazole-5-carboxamides: a new class of selective Rac inhibitors
.
Medchemcomm
2013
;
4
:
537
.
42.
Ruffoni
A
,
Ferri
N
,
Bernini
SK
,
Ricci
C
,
Corsini
A
,
Maffuci
I
, et al
2 - Amino-3-(phenylsulfanyl)norbornane-2-carboxylate: an appealing sca ff old for the design of rac1 − tiam1 protein − protein interaction inhibitors
.
J Med Chem
2014
;
57
:
2953
62
.
43.
Cardama
G
,
Comin
M
,
Hornos
L
,
Gonzalez
N
,
Defelipe
L
,
Turjanski
A
, et al
Preclinical development of novel rac1-GEF signaling inhibitors using a rational design approach in highly aggressive breast cancer cell lines
.
Anticancer Agents Med Chem
2014
;
14
:
840
51
.
44.
Cardama
GA
,
Gonzalez
N
,
Ciarlantini
M
,
Donadío
LG
,
Comin
MJ
,
Alonso
DF
, et al
Proapoptotic and antiinvasive activity of Rac1 small molecule inhibitors on malignant glioma cells
.
Onco Targets Ther
2014
;
7
:
2021
33
.
45.
Cabrera
M
,
Echeverria
E
,
Lenicov
FR
,
Cardama
G
,
Gonzalez
N
,
Davio
C
, et al
Pharmacological Rac1 inhibitors with selective apoptotic activity in human acute leukemic cell lines
.
Oncotarget
2017
;
8
:
98509
23
.
46.
Gonzalez
N
,
Cardama
GA
,
Comin
MJ
,
Segatori
VI
,
Pifano
M
,
Alonso
DF
, et al
Pharmacological inhibition of Rac1-PAK1 axis restores tamoxifen sensitivity in human resistant breast cancer cells
.
Cell Signal
2017
;
30
:
154
61
.
47.
Bouquier
N
,
Vignal
E
,
Charrasse
S
,
Weill
M
,
Schmidt
S
,
Léonetti
JP
, et al
A cell active chemical GEF inhibitor selectively targets the trio/RhoG/Rac1 signaling pathway
.
Chem Biol
2009
;
16
:
657
66
.
48.
Poppe
D
,
Tiede
I
,
Fritz
G
,
Becker
C
,
Bartsch
B
,
Wirtz
S
, et al
Azathioprine suppresses ezrin-radixin-moesin-dependent T cell-APC conjugation through inhibition of Vav guanosine exchange activity on Rac proteins
.
J Immunol
2006
;
176
:
640
51
.
49.
Menna
PL
,
Parera
RL
,
Cardama
GA
,
Alonso
DF
,
Gomez
DE
,
Farina
HG
. 
Enhanced cytostatic activity of statins in mouse mammary carcinoma cells overexpressing beta2-chimaerin
.
Mol Med Rep
2009
;
2
:
97
102
.
50.
Nishikimi
A
,
Uruno
T
,
Duan
X
,
Cao
Q
,
Okamura
Y
,
Saitoh
T
, et al
Blockade of inflammatory responses by a small-molecule inhibitor of the Rac activator DOCK2
.
Chem Biol
2012
;
19
:
488
97
.
51.
Sakamoto
K
,
Adachi
Y
,
Komoike
Y
,
Kamada
Y
,
Koyama
R
,
Fukuda
Y
, et al
Novel DOCK2-selective inhibitory peptide that suppresses B-cell line migration
.
Biochem Biophys Res Commun
2017
;
483
:
183
90
.
52.
Tajiri
H
,
Uruno
T
,
Shirai
T
,
Takaya
D
,
Matsunaga
S
,
Setoyama
D
, et al
Targeting Ras-driven cancer cell survival and invasion through selective inhibition of DOCK1
.
Cell Rep
2017
;
19
:
969
80
.
53.
Peterson
JR
,
Lebensohn
AM
,
Pelish
HE
,
Kirschner
MW
. 
Biochemical suppression of small molecule inhibitors: a new strategy to identify inhibitor targets and signaling pathway components
.
Chem Biol
2006
;
13
:
443
52
.
54.
Sakamori
R
,
Yu
S
,
Zhang
X
,
Hoffman
A
,
Sun
J
,
Das
S
, et al
CDC42 inhibition suppresses progression of incipient intestinal tumors
.
Cancer Res
2014
;
74
:
5480
92
.
55.
Friesland
A
,
Zhao
Y
,
Chen
Y-H
,
Wang
L
,
Zhou
H
,
Lu
Q
. 
Small molecule targeting Cdc42-intersectin interaction disrupts Golgi organization and suppresses cell motility
.
Proc Natl Acad Sci U S A
2013
;
110
:
1261
6
.
56.
Shutes
A
,
Onesto
C
,
Picard
V
,
Leblond
B
,
Schweighoffer
F
,
Der
CJ
. 
Specificity and mechanism of action of EHT 1864, a novel small molecule inhibitor of Rac family small GTPases
.
J Biol Chem
2007
;
282
:
35666
78
.
57.
Rosenblatt
AE
,
Garcia
MI
,
Lyons
L
,
Xie
Y
,
Maiorino
C
,
Desire
L
, et al
Inhibition of the Rho GTPase, Rac1, decreases estrogen receptor levels and is a novel therapeutic strategy in breast cancer
.
Endocr Relat Cancer
2011
;
18
:
207
19
.
58.
Castoria
G
,
D'Amato
L
,
Ciociola
A
,
Giovannelli
P
,
Giraldi
T
,
Sepe
L
, et al
Androgen-induced cell migration: role of androgen receptor/filamin A association
.
PLoS One
2011
;
6
:
e17218
.
59.
Molnár
J
,
Fazakas
C
,
Haskó
J
,
Sipos
O
,
Nagy
K
,
Nyúl-Tóth
Á
, et al
Transmigration characteristics of breast cancer and melanoma cells through the brain endothelium: role of rac and PI3K
.
Cell Adhes Migr
2016
;
10
:
269
81
.
60.
Katz
E
,
Sims
AH
,
Sproul
D
,
Caldwell
H
,
Dixon
MJ
,
Meehan
RR
, et al
Targeting of Rac GTPases blocks the spread of intact human breast cancer
.
Oncotarget
2012
;
3
:
608
19
.
61.
Hampsch
RA
,
Shee
K
,
Bates
D
,
Lewis
LD
,
Désiré
L
,
Leblond
B
, et al
Therapeutic sensitivity to Rac GTPase inhibition requires consequential suppression of mTORC1, AKT, and MEK signaling in breast cancer
.
Oncotarget
2017
;
8
:
21806
17
.
62.
Surviladze
Z
,
Waller
A
,
Wu
Y
,
Romero
E
,
Edwards
BS
,
Wandinger-Ness
A
, et al
Identification of a small GTPase inhibitor using a high-throughput flow cytometry bead-based multiplex assay
.
J Biomol Screen
2010
;
15
:
10
20
.
63.
Arnst
JL
,
Hein
AL
,
Taylor
MA
,
Palermo
NY
,
Contreras
I
,
Sonawane
YA
, et al
Discovery and characterization of small molecule Rac1 inhibitors
.
Oncotarget
2017
;
8
:
34586
600
.
64.
Beausoleil
E
,
Chauvignac
C
,
Taverne
T
,
Lacombe
S
,
Pognante
L
,
Leblond
B
, et al
Structure-activity relationship of isoform selective inhibitors of Rac1/1b GTPase nucleotide binding
.
Bioorganic Med Chem Lett
2009
;
19
:
5594
8
.
65.
Singh
A
,
Karnoub
AE
,
Palmby
TR
,
Lengyel
E
,
Sondek
J
,
Der
CJ
. 
Rac1b, a tumor associated, constitutively active Rac1 splice variant, promotes cellular transformation
.
Oncogene
2004
;
23
:
9369
80
.
66.
Silva
AL
,
Carmo
F
,
Bugalho
MJ
. 
RAC1b overexpression in papillary thyroid carcinoma: a role to unravel
.
Eur J Endocrinol
2013
;
168
:
795
804
.
67.
Zhou
C
,
Licciulli
S
,
Avila
JL
,
Cho
M
,
Troutman
S
,
Jiang
P
, et al
The Rac1 splice form Rac1b promotes K-ras-induced lung tumorigenesis
.
Oncogene
2013
;
32
:
903
9
.
68.
Hong
L
,
Kenney
SR
,
Phillips
GK
,
Simpson
D
,
Schroeder
CE
,
Nöth
J
, et al
Characterization of a Cdc42 protein inhibitor and its use as a molecular probe
.
J Biol Chem
2013
;
288
:
8531
43
.
69.
Surviladze
Z
,
Waller
A
,
Strouse
JJ
,
Bologa
C
,
Ursu
O
,
Salas
V
, et al
A potent and selective inhibitor of Cdc42 GTPase
.
Bethesda, MD:
National Center for Biotechnology Information
; 
2010
.
70.
Oprea
TI
,
Sklar
LA
,
Agola
JO
,
Guo
Y
,
Silberberg
M
,
Roxby
J
, et al
Novel activities of select NSAID renantiomers against Rac1 and Cdc42 GTPases
.
PLoS One
2015
;
10
:
1
32
.
71.
Guo
Y
,
Kenney
SR
,
Muller
CY
,
Adams
S
,
Rutledge
T
,
Romero
E
, et al
R-ketorolac targets Cdc42 and Rac1 and alters ovarian cancer cell behaviors critical for invasion and metastasis
.
Mol Cancer Ther
2015
;
7
:
2215
28
.
72.
Peretti
AS
,
Dominguez
D
,
Grimes
MM
,
Hathaway
HJ
,
Prossnitz
ER
,
Rivera
MR
, et al
The R-enantiomer of ketorolac delays mammary tumor development in mouse mammary tumor virus-polyoma middle T antigen (MMTV-PyMT) Mice
.
Am J Pathol
2017
;
188
:
515
24
.
73.
Bidaud-Meynard
A
,
Arma
D
,
Taouji
S
,
Laguerre
M
,
Dessolin
J
,
Rosenbaum
J
, et al
A novel small-molecule screening strategy identifies mitoxantrone as a RhoGTPase inhibitor
.
Biochem J
2013
;
450
:
55
62
.
74.
Hodge
RG
,
Ridley
AJ
. 
Regulating Rho GTPases and their regulators
.
Nat Rev Mol Cell Biol
2016
;
17
:
496
510
.
75.
Zimonjic
DB
,
Chan
LN
,
Tripathi
V
,
Lu
J
,
Kwon
O
,
Popescu
NC
, et al
In vitro and in vivo effects of geranylgeranyltransferase I inhibitor P61A6 on non-small cell lung cancer cells
.
BMC Cancer
2013
;
13
:
198
.
76.
Lu
J
,
Chan
L
,
Fiji
HDG
,
Dahl
R
,
Kwon
O
,
Tamanoi
F
. 
In vivo antitumor effect of a novel inhibitor of protein geranylgeranyltransferase-I
.
Mol Cancer Ther
2009
;
8
:
1218
26
.
77.
Tanaka
S
,
Fukumoto
Y
,
Nochioka
K
,
Minami
T
,
Kudo
S
,
Shiba
N
, et al
Statins exert the pleiotropic effects through small GTP-binding protein dissociation stimulator upregulation with a resultant rac1 degradation
.
Arterioscler Thromb Vasc Biol
2013
;
33
:
1591
600
.
78.
Altwairgi
AK.
Statins are potential anticancerous agents (Review)
.
Oncol Rep
2015
;
33
:
1019
39
.
79.
Alfaqih
MA
,
Allott
EH
,
Hamilton
RJ
,
Freeman
MR
,
Freedland
SJ
. 
The current evidence on statin use and prostate cancer prevention: are we there yet?
Nat Rev Urol
2016
;
14
:
107
19
.
80.
Ahern
TP
,
Lash
TL
,
Damkier
P
,
Christiansen
PM
,
Cronin-Fenton
DP
. 
Statins and breast cancer prognosis: evidence and opportunities
.
Lancet Oncol
2014
;
15
:
e461
8
.
81.
Pelish
HE
,
Peterson
JR
,
Salvarezza
SB
,
Rodriguez-Boulan
E
,
Chen
J-L
,
Stamnes
M
, et al
Secramine inhibits Cdc42-dependent functions in cells and Cdc42 activation in vitro
.
Nat Chem Biol
2006
;
2
:
39
46
.
82.
Taniuchi
K
,
Yokotani
K
,
Saibara
T
. 
BART inhibits pancreatic cancer cell invasion by Rac1 inactivation through direct binding to active Rac1
.
Neoplasia
2012
;
14
:
440
50
.
83.
Murali
A
,
Rajalingam
K
. 
Small Rho GTPases in the control of cell shape and mobility
.
Cell Mol Life Sci
2014
;
71
:
1703
21
.
84.
Cheng
WY
,
Chiao
MT
,
Liang
YJ
,
Yang
YC
,
Shen
CC
,
Yang
CY
. 
Luteolin inhibits migration of human glioblastoma U-87 MG and T98G cells through downregulation of Cdc42 expression and PI3K/AKT activity
.
Mol Biol Rep
2013
;
40
:
5315
26
.
85.
Lin
Y
,
Shi
R
,
Wang
X
,
Shen
H-M
. 
Luteolin, a flavonoid with potential for cancer prevention and therapy
.
Curr Cancer Drug Targets
2008
;
8
:
634
46
.
86.
Porchia
LM
,
Guerra
M
,
Wang
Y-C
,
Zhang
Y
,
Espinosa
A V
,
Shinohara
M
, et al
2-amino-N-{4-[5-(2-phenanthrenyl)-3-(trifluoromethyl)-1H-pyrazol-1-yl]-phenyl} acetamide (OSU-03012), a celecoxib derivative, directly targets p21-activated kinase
.
Mol Pharmacol
2007
;
72
:
1124
31
.
87.
Maksimoska
J
,
Feng
L
,
Harms
K
,
Yi
C
,
Kissil
J
,
Marmorstein
R
, et al
Targeting large kinase active site with rigid, bulky octahedral ruthenium complexes
.
J Am Chem Soc
2008
;
130
:
15764
5
.
88.
Licciulli
S
,
Maksimoska
J
,
Zhou
C
,
Troutman
S
,
Kota
S
,
Liu
Q
, et al
FRAX597, a small molecule inhibitor of the p21-activated kinases, inhibits tumorigenesis of neurofibromatosis type 2 (NF2)-associated schwannomas
.
J Biol Chem
2013
;
288
:
29105
14
.
89.
Rudolph
J
,
Crawford
JJ
,
Hoeflich
KP
,
Chernoff
J
. 
p21-activated kinase inhibitors
.
Enzym
2013
;
34 Pt. B
:
157
80
.
90.
Park
J-M
,
Han
Y-M
,
Jeong
M
,
Chung
MH
,
Kwon
C
 Il
,
Ko
KH
, et al
Synthetic 8-hydroxydeoxyguanosine inhibited metastasis of pancreatic cancer through concerted inhibitions of ERM and Rho-GTPase
.
Free Radic Biol Med
2017
;
110
:
151
61
.
91.
Peterson
JR
,
Lokey
RS
,
Mitchison
TJ
,
Kirschner
MW
. 
A chemical inhibitor of N-WASP reveals a new mechanism for targeting protein interactions
.
Proc Natl Acad Sci U S A
2001
;
98
:
10624
9
.
92.
Peterson
JR
,
Bickford
LC
,
Morgan
D
,
Kim
AS
,
Ouerfelli
O
,
Kirschner
MW
, et al
Chemical inhibition of N-WASP by stabilization of a native autoinhibited conformation
.
Nat Struct Mol Biol
2004
;
11
:
747
55
.
93.
Murakami
T
,
Singh
AS
,
Kiyuna
T
,
Dry
SM
,
Li
Y
,
James
AW
, et al
Effective molecular targeting of CDK4/6 and IGF-1R in a rare FUS-ERG fusion CDKN2A-deletion doxorubicin-resistant Ewing's sarcoma patient-derived orthotopic xenograft (PDOX) nude-mouse model
.
Oncotarget
2016
;
7
:
47556
64
.
94.
Hiroshima
Y
,
Maawy
AA
,
Katz
MHG
,
Fleming
JB
,
Bouvet
M
,
Endo
I
, et al
Selective efficacy of zoledronic acid on metastasis in a patient-derived orthotopic xenograph (PDOX) nude-mouse model of human pancreatic cancer
.
J Surg Oncol
2015
;
111
:
311
5
.
95.
Kawaguchi
K
,
Murakami
T
,
Chmielowski
B
,
Igarashi
K
,
Kiyuna
T
,
Unno
M
, et al
Vemurafenib-resistant BRAF-V600E-mutated melanoma is regressed by MEK-targeting drug trametinib, but not cobimetinib in a patient-derived orthotopic xenograft (PDOX) mouse model
.
Oncotarget
2016
;
7
:
71737
43
.
96.
Kawaguchi
K
,
Igarashi
K
,
Murakami
T
,
Kiyuna
T
,
Nelson
SD
,
Dry
SM
, et al
Combination of gemcitabine and docetaxel regresses both gastric leiomyosarcoma proliferation and invasion in an imageable patient-derived orthotopic xenograft (iPDOX) model
.
Cell Cycle
2017
;
16
:
1063
9
.
97.
Yan
Y
,
Hein
AL
,
Etekpo
A
,
Burchett
KM
,
Lin
C
,
Enke
CA
, et al
Inhibition of RAC1 GTPase sensitizes pancreatic cancer cells to γ-irradiation
.
Oncotarget
2014
;
5
:
10251
70
.
98.
Yan
Y
,
Greer
PM
,
Cao
PT
,
Kolb
RH
,
Cowan
KH
. 
RAC1 GTPase plays an important role in γ-irradiation induced G2/M checkpoint activation
.
Breast Cancer Res
2012
;
14
:
R60
.
99.
Dokmanovic
M
,
Hirsch
DS
,
Shen
Y
,
Wu
WJ
. 
Rac1 contributes to trastuzumab resistance of breast cancer cells: rac1 as a potential therapeutic target for the treatment of trastuzumab-resistant breast cancer
.
Mol Cancer Ther
2009
;
8
:
1557
69
.
100.
Kaneto
N
,
Yokoyama
S
,
Hayakawa
Y
,
Kato
S
,
Sakurai
H
,
Saiki
I
. 
RAC1 inhibition as a therapeutic target for gefitinib-resistant non-small-cell lung cancer
.
Cancer Sci
2014
;
105
:
788
94
.
101.
Chen
HY
,
Yang
YM
,
Stevens
BM
,
Noble
M
. 
Inhibition of redox/Fyn/c-Cbl pathway function by Cdc42 controls tumour initiation capacity and tamoxifen sensitivity in basal-like breast cancer cells
.
EMBO Mol Med
2013
;
5
:
723
36
.
102.
Cerami
E
,
Gao
J
,
Dogrusoz
U
,
Gross
BE
,
Sumer
SO
,
Aksoy
BA
, et al
The cBio cancer genomics portal: an open platform for exploring multidimensional cancer genomics data
.
Cancer Discov
2012
;
2
:
401
4
.
103.
Cancer Genome Atlas Research Network
. 
Comprehensive molecular characterization of urothelial bladder carcinoma
.
Nature
2014
;
507
:
315
22
.
104.
Ciriello
G
,
Gatza
ML
,
Beck
AH
,
Wilkerson
MD
,
Rhie
SK
,
Pastore
A
, et al
Comprehensive molecular portraits of invasive lobular breast cancer
.
Cell
2015
;
163
:
506
19
.
105.
Brennan
CW
,
Verhaak
RGW
,
McKenna
A
,
Campos
B
,
Noushmehr
H
,
Salama
SR
, et al
The somatic genomic landscape of glioblastoma
.
Cell
2013
;
155
:
462
77
.
106.
Cancer Genome Atlas Research Network
. 
Comprehensive molecular characterization of gastric adenocarcinoma
.
Nature
2014
;
513
:
202
9
.
107.
Cancer Genome Atlas Research Network
. 
Comprehensive molecular profiling of lung adenocarcinoma
.
Nature
2014
;
511
:
543
50
.
108.
Bailey
P
,
Chang
DK
,
Nones
K
,
Johns
AL
,
Patch
A-M
,
Gingras
M-C
, et al
Genomic analyses identify molecular subtypes of pancreatic cancer
.
Nature
2016
;
531
:
47
52
.
109.
Robinson
D
,
Van Allen
EM
,
Wu
Y-M
,
Schultz
N
,
Lonigro
RJ
,
Mosquera
J-M
, et al
Integrative clinical genomics of advanced prostate cancer
.
Cell
2015
;
161
:
1215
28
.
110.
Azab
AK
,
Azab
F
,
Pitsillides
CM
,
Ngo
HT
,
Jia
XY
,
Melhem
MR
, et al
Rho-a and Rac-1 GTPases play major and differential roles in SDF1 alpha-induced cell adhesion and chemotaxis in multiple myeloma
.
Blood
2009
;
114
:
619
29
.
111.
Rozenveld-Geugien
M
,
Baas
IO
,
van Gosliga
D
,
Vellenga
E
,
Schuringa
JJ
. 
Expansion of normal and leukemic human hematopoietic stem/progenitor cells requires Rac-mediated interaction with stromal cells
.
Exp Hematol
2007
;
35
:
782
92
.
112.
Cardama
G
,
Gonzalez
N
,
Maggio
J
,
Menna
P
,
Gomez
D
. 
Rho GTPases as therapeutic targets in cancer (Review)
.
Int J Oncol
2017
;
51
:
1025
34
.
113.
Bid
HK
,
Roberts
RD
,
Manchanda
PK
,
Houghton
PJ
. 
RAC1: an emerging therapeutic option for targeting cancer angiogenesis and metastasis
.
Mol Cancer Ther
2013
;
12
:
1925
34
.
114.
Hinterleitner
C
,
Huelsenbeck
J
,
Henninger
C
,
Wartlick
F
,
Schorr
A
,
Kaina
B
, et al
Rac1 signaling protects monocytic AML cells expressing the MLL-AF9 oncogene from caspase-mediated apoptotic death
.
Apoptosis
2013
;
18
:
963
79
.
115.
Humphreys
KJ
,
McKinnon
RA
,
Michael
MZ
. 
Mir-18a inhibits CDC42 and plays a tumour suppressor role in colorectal cancer cells
.
PLoS One
2014
;
9
:
e112288
.
116.
Huang
Z
,
Zhang
L
,
Chen
Y
,
Zhang
H
,
Zhang
Q
,
Li
R
, et al
Cdc42 deficiency induces podocyte apoptosis by inhibiting the Nwasp/stress fibers/YAP pathway
.
Cell Death Dis
2016
;
7
:
e2142
.