Adoptive cellular immunotherapy inducing a graft-versus-tumor (GVT) effect is the therapeutic mainstay of allogeneic hematopoietic stem cell transplantation (ASCT) for high-risk leukemias. Autologous immunotherapies using vaccines or adoptive transfer of ex vivo–manipulated lymphocytes are clinically explored in patients with various cancer entities. Main reason for failure of ASCT and cancer immunotherapy is progression of the underlying malignancy, which is more prevalent in patients with advanced disease. Elucidating the molecular mechanisms contributing to immune escape will help to develop strategies for the improvement of immunologic cancer treatment. To this end, we have undertaken functional screening and expression cloning of factors mediating resistance to antigen-specific cytotoxic T lymphocytes (CTLs). We have identified Cdc42, a GTPase regulating actin dynamics and growth factor signaling that is highly expressed in invasive cancers, as determinator of cancer cell susceptibility to antigen-specific CTLs in vitro and adoptively transferred immune effectors in vivo. Cdc42 prevents CTL-induced apoptosis via mitogen-activated protein kinase (MAPK) signaling and posttranscriptional stabilization of Bcl-2. Pharmacologic inhibition of MAPK/extracellular signal–regulated kinase (ERK) kinase (MEK) overcomes Cdc42-mediated immunoresistance and activation of Bcl-2 in vivo. In conclusion, Cdc42 signaling contributes to immune escape of cancer. Targeting Cdc42 may improve the efficacy of cancer immunotherapies.

Major advances in the understanding of the immune system have fostered the application of immune interventions to the benefit of patients. Among those, cancer immunotherapy is of particular interest, as it aims to specifically search and destroy malignant cells while sparing the surrounding healthy tissues.1  In the past decade, passive immunotherapy using monoclonal antibodies has been successfully introduced into clinical oncology, and has become standard of care for patients suffering from certain breast cancers, B-cell lymphomas, colorectal cancer, and others. Adoptive cellular immunotherapy in the context of allogeneic hematopoietic stem cell transplantation (ASCT) is the standard treatment for patients with high-risk leukemias, and additional indications are clinically explored.2,3  In contrast, the clinical role of autologous cellular immunotherapy for cancer still remains to be defined. Current studies explore the application of various formats of cancer vaccines, as well as the adoptive transfer of ex vivo–manipulated tumor-reactive lymphocytes.4-6 

In the process of optimizing autologous and allogeneic cancer immunotherapies, much attention is paid to the identification of cancer-specific antigens and strategies to improve the activation of an immune response.6  Obviously, the effector phase of cancer immunity is of equal importance for therapeutic success. Malignant cells were reported to devise multiple strategies to escape immune recognition and to suppress antitumoral immune responses.7  Moreover, cancers may resist death signals imposed by cellular immune effectors using resistance mechanisms that interfere with endogenous signal transduction. Examples are the expression of serpins, which neutralize cytotoxic T-lymphocyte (CTL)–derived granzymes, and FLICE-inhibitory proteins, which interfere with death receptor signaling.8-10  Recently, it was shown that factors conferring resistance to anticancer drugs and radiation can also protect tumor cells against CTL-induced apoptosis.11,12  Accordingly, therapeutic targeting of such central mediators of resistance may increase the efficacy of both pharmacologic and immunologic cancer treatments. The definition of such targets requires a detailed understanding of the molecular mechanisms used by cancer cells to resist an immune attack. To this end, we have devised functional screening and expression cloning to reveal Cdc42 as a factor mediating cell-autonomous resistance to CTL-induced tumor suppression and cytotoxic therapies. Cdc42 is a Rho GTPase involved in the regulation of the actin cytoskeleton and growth factor signal transduction, which contributes to oncogenic transformation and cancer invasiveness.13-15  Here, we show that Cdc42 signals to mitogen-activated protein kinase (MAPK) and Bcl-2 to protect cancer cells against immune-mediated destruction in vitro and in vivo. Pharmacologic inhibition of MAPK/extracellular signal–regulated kinase (ERK) kinase (MEK) activity successfully sensitized established cancers to tumor suppression by adoptively transplanted cellular immune effectors, thus providing a lead for therapeutic modulation of Cdc42-mediated immunoresistance.

Cell lines

HLA-A*0201–transgenic murine embryonic fibroblasts (MEFs) generated from cA2Kb mice backcrossed on a C57BL/6 background were retrovirally transduced to express cooperating oncogenes as published11 ; cA2Kb is a transgenic line expressing a chimeric MHC class I molecule composed of the α1 and α2 domains of HLA-A*0201 and the α3, transmembrane, and cytoplasmic domains of H-2Kb.16  MEFs were maintained in Dulbecco modified Eagle medium (DMEM) supplemented with fetal bovine serum, l-glutamine, and penicillin and streptomycin (Invitrogen, Carlsbad, CA). Allo-A2Kb–reactive CD8+ CTL-lysing targets expressing the HLA-A*0201 antigen, HLA-A*0201/influenza matrix(58-66)–specific CTLlysing targets, and HLA-A*0201/p53(264-272) CTL lysing targets presenting the influenza matrix(58-66) epitope or the human p53(264-272) epitope in the context of HLA-A*0201 were generated by immunizing HLA-A*0201 transgenic mice with the respective peptide epitopes, as published previously.17,18  The HLA-A*0201+ human colorectal cancer cell line HCT116 has been generously provided by B. Vogelstein (Johns Hopkins Kimmel Cancer Center, Baltimore, MD).

Cytotoxicity and apoptosis assays

51Cr release cytotoxicity assays (5 hours) were carried out as described.19  Apoptosis was detected by cell-cycle analysis following staining with propidium iodide (PI). Cells with maintained mitochondrial transmembrane potential Δψm were quantitated flow cytometrically following staining with the dye TMRE (Invitrogen). To assay proliferative survival, 5000 adherent MEFs were incubated in 96-well plates with CTL effectors at the indicated effector-target (E/T) ratios for 4.5 hours. Following removal of CTLs, MEFs were harvested and replated in 35-mm dishes for a 7-day culture period. The resulting colonies were fixed, stained, and counted.11 

Plasmids, antibodies, and reagents

A cDNA encoding a GTPase-defective Cdc42 was generated by polymerase chain reaction (PCR) using pRK5-MYC.Cdc42 (Q61L; provided by P. Aspenström Uppsala University, Uppsala, Sweden) as template and cloned into the retroviral vector plasmids pMxIG (provided by T. Kitamura, University of Tokyo, Tokyo, Japan) and pQCxIN (Clontech, Mountain View, CA). All inserts were verified by sequencing. The following primary antibodies were used: actin (C4; MP Biomedicals, Irvine, CA); Bcl-2 (C2), Bcl-xL (H5), and Mcl-1 (all from Santa Cruz Biotechnology, Santa Cruz, CA); and Cdc42 (MAB3707; Chemicon, Temecula, CA). All phosphoepitope-specific antisera were purchased from Cell Signaling Technology (Danvers, MA). ABT-737 was generously provided by Abbott Laboratories (Abbott Park, IL); and PD98059, staurosporine, and etoposide were purchased from Calbiochem (San Diego, CA) and Sigma-Aldrich (St Louis, MO), respectively.

Generation of a retroviral expression library

A cDNA library was constructed from the leukapheresis product of a patient with chronic myeloid leukemia using the pCMV-Script XR Library Construction Kit (Stratagene, Cambridge, United Kingdom). Total RNA was isolated and poly(A) + mRNA was prepared by means of the Oligotex mRNA Kit (Qiagen, Hilden, Germany). The mRNA was reverse-transcribed using an oligo-dT primer containing an EcoRI site at its 5′ end and ligated to adaptors. Ligation products were then digested with XhoI and ligated into the EcoRI and XhoI sites of pMxIG, and recombinant plasmids were electroporated into Escherichia coli XL10-Gold Ultracompetent Cells (Stratagene). The resulting library of 3.8 × 105 cells represented 95% inserts with sizes between 0.5 and 4.0 kb. Replication-defective retroviral vectors were generated as published11  to transduce oncogene-transformed cA2Kb MEFs under conditions ensuring a multiplicity of infection of 1. All studies using human material were approved by the local ethics committee (Landesärztekammer Rheinland-Pfalz, Mainz, Germany); the patient provided written informed consent in accordance with the Declaration of Helsinki.

Splenocyte transplantation model

Irradiated (1.5 Gy [150 rad]) nonobese diabetic/severe combined immunodeficient (NOD/SCID) mice received subcutaneous injections of 106 cA2Kb MEFs (Cdc42 MEFs in left flank, control MEFs in right flank). Following the outgrowth of palpable fibrosarcomas (days 7-8), mice were treated by single tail-vein injections of 5 × 107 splenocytes from C57BL/6 or cA2Kb mice resuspended in saline, and a subcutaneous injection of 6 × 105 U interleukin-2 (Medicopharm, Nussdorf, Germany) dissolved in saline and incomplete Freund adjuvant. Tumor sizes were measured bidimensionally using a calliper. Differences between the resulting growth curves were calculated using analysis of variance (ANOVA) and Student t test. To obtain splenocyte preparations devoid of CD8+ T cells or CD49+ natural killer (NK) cells, splenocytes were depleted by means CD8 or CD49 microbeads, respectively, and a magnetic-activated cell sorter (MACS) system (Miltenyi Biotec, Bergisch-Gladbach, Germany) following the manufacturer's instructions. Daily intraperitoneal injections of PD98059 (5 mg/kg; Calbiochem) dissolved in DMSO (Sigma-Aldrich) and diluted in saline were administered for MEK inhibition in vivo. All animal studies were conducted in compliance with institutional guidelines and were approved by the responsible regulatory authority (Landesuntersuchungsamt Rheinland-Pfalz).

Functional expression cloning of Cdc42 as mediator of immunoresistance

To functionally identify resistance factors against CTL-mediated tumor suppression, we devised an expression cloning strategy (Figure 1A). In brief, oncogene-transformed HLA-A*0201–transgenic cA2Kb MEFs were retrovirally transduced to express a cDNA library and green fluorescent protein (GFP). Clones surviving coculture with allo-A2Kb–reactive CD8+ CTLs (allo-A2), which lyse targets expressing the HLA-A*0201 antigen,17,18  were expanded and analyzed for GFP expression. Vector-encoded cDNA sequences of CTL-resistant and GFP+ clones were retrieved by PCR and sequencing using vector-specific primers. Here, we report clone A4, which expressed a library sequence spanning the entire open reading frame of the Homo sapiens cell division cycle 42 (GTP-binding protein, 25 kDa; Cdc42), transcript variant 1, mRNA (GenBank accession no. NM_00179120 ; Figure 1B). Clone A4 exhibited marked resistance to allo-A2 CTL-induced cytotoxicity as compared with parental cA2Kb MEF (Figure 1C).

Figure 1

Functional screening and expression cloning of Cdc42 as mediator of immunoresistance. (A) Schematic diagram of the library expression and screening strategies. (B) PCR amplification (left panel; first lane: DNA marker) and sequence (right panel) of the vector-encoded cDNA of clone A4. The vector-encoded EcoRI restriction site is underlined; the Cdc42 open reading frame is shaded. (C) The 5-hour 51Cr release assay of cA2Kb MEFs (□ indicates parental MEFs; ■, clone A4) coincubated with allo-A2 CTLs. Mean values plus or minus SD of a representative experiment are shown.

Figure 1

Functional screening and expression cloning of Cdc42 as mediator of immunoresistance. (A) Schematic diagram of the library expression and screening strategies. (B) PCR amplification (left panel; first lane: DNA marker) and sequence (right panel) of the vector-encoded cDNA of clone A4. The vector-encoded EcoRI restriction site is underlined; the Cdc42 open reading frame is shaded. (C) The 5-hour 51Cr release assay of cA2Kb MEFs (□ indicates parental MEFs; ■, clone A4) coincubated with allo-A2 CTLs. Mean values plus or minus SD of a representative experiment are shown.

Close modal

Cdc42 protects cancer cells against CTL-induced tumor suppression in vitro and in vivo

To study whether Cdc42 in fact mediated protection against CTL-induced cytotoxicity, we constructed a bicistronic retroviral vector for stable expression of a constitutively active Cdc42 (Q61L) mutant21  and GFP. Following transduction of cA2Kb MEFs, the GFP+ population (cA2Kb-Cdc42 MEF) was isolated by fluorescence-activated cell sorting. This approach was taken to avoid antibiotic selection of drug-resistant clones. Such cA2Kb-Cdc42 MEFs were highly resistant to cytotoxicity induced by allo-A2 CTLs (Figure 2A). In addition, cA2Kb-Cdc42 MEFs cocultured with allo-A2 CTLs exhibited enhanced clonogenic survival as compared with cA2Kb-control MEF (Figure 2B). This protection was not explained by Cdc42-mediated down-modulation of MHC class I, as all MEF populations exhibited equal HLA-A*0201 expression (Figure S1A, available on the Blood website; see the Supplemental Materials link at the top of the online article). Similar results were obtained using the HLA-A*0201–positive human colorectal cancer cell line HCT116. HCT116 cells expressing Cdc42 (Q61L) were more resistant to immunologic tumor suppression by allo-A2 CTLs in vitro (Figure 2C).

Figure 2

Cdc42 confers immunoresistance of cancer in vitro. (A) The 5-hour 51Cr release assay of cA2Kb-Cdc42 (● ○) or cA2Kb-control MEFs (■ □ coincubated with allo-A2 CTLs (● ■) or A2 Flu control CTLs (○ □). Mean values plus or minus SD of 3 independent experiments are shown. (B) Clonogenic survival of cA2Kb-Cdc42 and cA2Kb-control MEFs coincubated with allo-A2 CTLs at the indicated E/T ratios (representative photograph of at least 3 independent experiments). (C) Clonogenic survival of HCT116-Cdc42 and HCT116-control human colorectal cancer cells coincubated with allo-A2 CTLs (representative photograph of 3 independent experiments).

Figure 2

Cdc42 confers immunoresistance of cancer in vitro. (A) The 5-hour 51Cr release assay of cA2Kb-Cdc42 (● ○) or cA2Kb-control MEFs (■ □ coincubated with allo-A2 CTLs (● ■) or A2 Flu control CTLs (○ □). Mean values plus or minus SD of 3 independent experiments are shown. (B) Clonogenic survival of cA2Kb-Cdc42 and cA2Kb-control MEFs coincubated with allo-A2 CTLs at the indicated E/T ratios (representative photograph of at least 3 independent experiments). (C) Clonogenic survival of HCT116-Cdc42 and HCT116-control human colorectal cancer cells coincubated with allo-A2 CTLs (representative photograph of 3 independent experiments).

Close modal

To extend these observations, we made use of an in vivo model recapitulating the graft-versus-tumor (GVT) effect of ASCT and adoptive lymphocyte transfer. In this model, the transplantation of unprimed splenocytes derived from C57BL/6 mice suppressed the growth of HLA-A*0201–transgenic C57BL/6 fibrosarcomas, which were established in NOD/SCID mice. The tumor suppressive activity of transplanted splenocytes mainly relied on HLA-A*0201–reactive CD8+ lymphocytes and CD49+ NK cells (Figure 3A,C) acting via perforin-dependent pathways.22  Following subcutaneous injection in NOD/SCID mice, cA2Kb-Cdc42 and cA2Kb-control MEFs readily formed HLA-A*0201–transgenic fibrosarcomas. The growth rate of these tumors was not altered by the expression of Cdc42 (Figure 3D). As expected, transplantation of allogeneic C57BL/6 splenocytes significantly suppressed the growth of cA2Kb-control fibrosarcomas, when compared with syngeneic cA2Kb splenocytes, which were tolerant to HLA-A*0201–expressing tumor cells (Figure 3A). In contrast, cA2Kb-Cdc42 fibrosarcomas largely resisted tumor suppression by transplanted allogeneic C57BL/6 splenocytes in vivo (Figure 3B). In summary, constitutively active Cdc42 protected cancer cells against cytotoxicity and tumor suppression by alloreactive CTLs in vitro and CTLs and NK cells in vivo.

Figure 3

Cdc42 confers resistance against CTL- and NK cell–mediated tumor suppression in vivo. (A) Growth of cA2Kb-control fibrosarcomas in NOD/SCID mice. The arrowhead indicates the time point of adoptive transfer of allogeneic C57BL/6-derived (■) or syngeneic HLA-A*0201–transgenic cA2Kb splenocytes (□). (B) Growth of cA2Kb-Cdc42 fibrosarcomas in NOD/SCID mice. The arrowhead indicates the time point of adoptive transfer of allogeneic C57BL/6-derived (●) or syngeneic HLA-A*0201–transgenic cA2Kb splenocytes (○). (C) Growth of cA2Kb-control fibrosarcomas in NOD/SCID mice. The arrowhead indicates the time point of adoptive transfer of allogeneic C57BL/6-derived (■), CD8-depleted (○), or NK cell–depleted (▲) splenocytes. The growth curves following undepleted and depleted splenocyte transfer differed significantly (P < .001; ANOVA). (D) Spontaneous growth of cA2Kb-control (□) and cA2Kb-Cdc42 (●) fibrosarcomas in NOD/SCID mice. Mean values plus or minus SE of bidimensional tumor sizes of 5 mice per group are shown for each experiment (*P < .001; **P = .025, Student t test).

Figure 3

Cdc42 confers resistance against CTL- and NK cell–mediated tumor suppression in vivo. (A) Growth of cA2Kb-control fibrosarcomas in NOD/SCID mice. The arrowhead indicates the time point of adoptive transfer of allogeneic C57BL/6-derived (■) or syngeneic HLA-A*0201–transgenic cA2Kb splenocytes (□). (B) Growth of cA2Kb-Cdc42 fibrosarcomas in NOD/SCID mice. The arrowhead indicates the time point of adoptive transfer of allogeneic C57BL/6-derived (●) or syngeneic HLA-A*0201–transgenic cA2Kb splenocytes (○). (C) Growth of cA2Kb-control fibrosarcomas in NOD/SCID mice. The arrowhead indicates the time point of adoptive transfer of allogeneic C57BL/6-derived (■), CD8-depleted (○), or NK cell–depleted (▲) splenocytes. The growth curves following undepleted and depleted splenocyte transfer differed significantly (P < .001; ANOVA). (D) Spontaneous growth of cA2Kb-control (□) and cA2Kb-Cdc42 (●) fibrosarcomas in NOD/SCID mice. Mean values plus or minus SE of bidimensional tumor sizes of 5 mice per group are shown for each experiment (*P < .001; **P = .025, Student t test).

Close modal

Cdc42 confers cross-resistance of cancer cells to CTL-mediated cytotoxicity and drug-induced apoptosis

Next, we asked whether protection of cancer cells by Cdc42 was restricted to cytotoxicity induced by alloreactive CTLs. To this end, cA2Kb-Cdc42 and cA2Kb-control MEFs were loaded with peptides representing the HLA-A*0201–restricted influenza matrix(58-66) or human p53(264-272) epitopes, followed by coculture with HLA-A*0201/influenza matrix(58-66)–specific CTLs (A2 Flu) and HLA-A*0201/p53(264-272) CTLs (A2 p53), respectively.17,18  Again, cA2Kb-Cdc42 MEFs exhibited significant protection against cytotoxicity induced by cellular immune effectors of different specificities (Figure 4A). This argued for an activity of Cdc42 interfering with essential CTL effector mechanisms.

Figure 4

Cdc42 prevents apoptosis as induced by CTLs of different specificities or cytotoxic drugs. (A) cA2Kb-Cdc42 (■) or cA2Kb-control MEFs (□) were loaded with different peptide concentrations representing HLA-A*0201–restricted influenza (top panel) and human p53 (bottom panel) epitopes, followed by coincubation with A2 Flu CTLs (top panel; E/T = 30) or A2 p53 CTLs (bottom panel; E/T = 15). Mean values plus or minus SD of 5-hour 51Cr release assays. (B) cA2Kb-Cdc42 (■) or cA2Kb-control MEFs (□) were treated for 24 hours with staurosporine (left) or etoposide (right). Cells with maintained Δψm (TMRE+; top panel) or apoptotic DNA fragmentation (sub-G1; bottom panels) were quantified by flow cytometry following staining with the fluorescent dyes TMRE or propidium iodide, respectively. Mean values plus or minus SD of 3 repetitions are shown.

Figure 4

Cdc42 prevents apoptosis as induced by CTLs of different specificities or cytotoxic drugs. (A) cA2Kb-Cdc42 (■) or cA2Kb-control MEFs (□) were loaded with different peptide concentrations representing HLA-A*0201–restricted influenza (top panel) and human p53 (bottom panel) epitopes, followed by coincubation with A2 Flu CTLs (top panel; E/T = 30) or A2 p53 CTLs (bottom panel; E/T = 15). Mean values plus or minus SD of 5-hour 51Cr release assays. (B) cA2Kb-Cdc42 (■) or cA2Kb-control MEFs (□) were treated for 24 hours with staurosporine (left) or etoposide (right). Cells with maintained Δψm (TMRE+; top panel) or apoptotic DNA fragmentation (sub-G1; bottom panels) were quantified by flow cytometry following staining with the fluorescent dyes TMRE or propidium iodide, respectively. Mean values plus or minus SD of 3 repetitions are shown.

Close modal

CTLs eliminate their specific targets by inducing apoptotic cell death. It was shown that CTLs can activate apoptotic caspases through several mechanisms, including the “mitochondrial” pathway, which is also triggered by anticancer drugs or radiation.11,23  Against this background, we assessed whether cA2Kb-Cdc42 MEFs were resistant to drug-induced apoptosis. Indeed, expression of Cdc42 protected cA2Kb MEFs against dissipation of the mitochondrial transmembrane potential Δψm (Figure 4B top panels) and apoptotic DNA fragmentation (Figure 4B bottom panels) triggered by the protein kinase inhibitor staurosporine and the topoisomerase II inhibitor etoposide. These findings suggested that Cdc42 mediated immunoresistance by inhibition of cancer cell–intrinsic apoptosis signaling, and not by interfering with the interaction between CTLs and cancer cells. As Cdc42-expressing cells maintained their Δψm, apoptotic signal transduction was possibly intercepted at or upstream of the permeabilization of the mitochondrial outer membrane (MOM).

Cdc42 protects cancer cells by posttranscriptional regulation of Bcl-2

The integrity of the MOM is regulated by the Bcl-2 family of proteins. Antiapoptotic family members, such as Bcl-xL, Mcl-1 or Bcl-2 itself, counteract the proapoptotic “multidomain” proteins Bax and Bak, which permeabilize the MOM through a mechanism, which is incompletely understood.24,25  Investigating the expression of antiapoptotic Bcl-2, Bcl-xL, and Mcl-1 in extracts derived from cA2Kb fibrosarcomas, we observed an increase of endogenous Bcl-2 expression in cA2Kb-Cdc42 tumors (Figure 5A). Using reverse transcription and quantitative PCR, no elevation of Bcl-2 RNA expression was detectable in cA2Kb-Cdc42 MEFs as compared with control MEFs (data not shown). To the contrary, expression levels and stability of endogenous Bcl-2 were markedly increased in Cdc42-expressing cA2Kb MEFs cultured in the presence of the translation inhibitor cycloheximide (Figure 5B). Thus, Cdc42 most likely inhibited apoptosis by posttranscriptional modulation of Bcl-2.

Figure 5

Cdc42 prevents apoptosis by posttranscriptional stabilization of Bcl-2. (A) Immunoblot analysis of extracts prepared from 3 cA2Kb-Cdc42 and 3 cA2Kb-control fibrosarcomas. Note the increase in Bcl-2 levels in the Cdc42-expressing tumors. (B) Immunoblot analysis of cA2Kb-Cdc42 and cA2Kb-control MEFs following treatment with the translation inhibitor cycloheximide (CHX; 200 μg/mL) for up to 6 hours. Note the enhancement of Bcl-2 stability in cA2Kb-Cdc42 MEFs. (C) Etoposide treatment (12.5 μM) of cA2Kb-Cdc42 (■) and cA2Kb-control MEFs (□) incubated with ABT-737 (6.25 μM) or vehicle under low serum conditions. Apoptotic DNA fragmentation was quantified by flow cytometry following staining with propidium iodide; mean values plus SD of 3 experiments are shown.

Figure 5

Cdc42 prevents apoptosis by posttranscriptional stabilization of Bcl-2. (A) Immunoblot analysis of extracts prepared from 3 cA2Kb-Cdc42 and 3 cA2Kb-control fibrosarcomas. Note the increase in Bcl-2 levels in the Cdc42-expressing tumors. (B) Immunoblot analysis of cA2Kb-Cdc42 and cA2Kb-control MEFs following treatment with the translation inhibitor cycloheximide (CHX; 200 μg/mL) for up to 6 hours. Note the enhancement of Bcl-2 stability in cA2Kb-Cdc42 MEFs. (C) Etoposide treatment (12.5 μM) of cA2Kb-Cdc42 (■) and cA2Kb-control MEFs (□) incubated with ABT-737 (6.25 μM) or vehicle under low serum conditions. Apoptotic DNA fragmentation was quantified by flow cytometry following staining with propidium iodide; mean values plus SD of 3 experiments are shown.

Close modal

To study whether Cdc42-mediated protection of cA2Kb MEFs in fact required the activity of antiapoptotic Bcl-2 proteins, we made use of the pharmacologic apoptosis sensitizer ABT-737. This compound was rationally developed as a mimetic of “BH3-only” proteins, such as Bim, Bid, or Bad, which act as inhibitors of antiapoptotic Bcl-2 proteins.26,27  Pretreatment of cA2Kb-control MEFs with ABT-737 failed to further enhance etoposide-induced apoptosis. In contrast, ABT-737 completely reversed the apoptosis resistance of cA2Kb-Cdc42 MEFs (Figure 5C). Recently, ABT-737 was shown to act exclusively on Bcl-2 and Bcl-xL, but not Mcl-1.28-30  As endogenous Bcl-xL levels of cA2Kb-Cdc42 MEFs remained unchanged (Figure 5A,B), these results confirmed that Cdc42-mediated inhibition of apoptosis indeed depended on Bcl-2 activity.

Cdc42-dependent immunoresistance requires MAPK signaling

Mammalian Cdc42 is a GTP-binding protein, which was initially described as a regulator of actin dynamics and cytoskeletal architecture in response to integrins and extracellular matrix signals.15  Recently, it was found to be involved in additional signal transduction pathways triggered by cytokine receptors and growth factor receptor tyrosine kinases.13  Cdc42 affects MAPK, which targets several regulators of apoptosis, including Bcl-2. Analyzing p42/44 mitogen-activated protein kinase (ERK) in extracts derived from cA2Kb-Cdc42 and cA2Kb-control fibrosarcomas, we observed activating phosphorylation of ERK in those tumors expressing Cdc42 (Figure 6A). Also, Cdc42-expressing HCT116 cells exhibited enhanced ERK phosphorylation in response to epidermal growth factor (EGF) stimulation (Figure S1D). As ERK-dependent phosphorylation of Bcl-2 was described to enhance its antiapoptotic activity,29,31  we assessed whether treatment with PD98059, a pharmacologic MEK inhibitor acting upstream of ERK in MAPK signal transduction, could overcome Cdc42-dependent immunoresistance. In control experiments, PD98059 did not influence the suppression of clonogenic survival of cA2Kb-control MEFs by allo-A2 CTLs. In contrast, cA2Kb-Cdc42 MEFs pretreated with PD98059 proved more susceptible to tumor suppression and cytotoxicity by alloreactive CTLs in vitro (Figures 6B,S1B).

Figure 6

Cdc42 confers immunoresistance by ERK-dependent up-regulation of Bcl-2. (A) Immunoblot analysis of extracts prepared from 3 cA2Kb-Cdc42 and 3 cA2Kb-control fibrosarcomas. Note that phosphoepitopes indicative of ERK activation are detectable in Cdc42-expressing tumors. (B) cA2Kb-control MEFs (left panel) and cA2Kb-Cdc42 MEFs (right panel) were pretreated with the MEK inhibitor PD98059 (10 μM for 2 hours) or vehicle, followed by coincubation with allo-A2 CTL. Clonogenic survival in 1 representative of at least 3 repetitions. (C) Growth of cA2Kb-control (■ □) and cA2Kb-Cdc42 (● ○) fibrosarcomas in NOD/SCID mice. Mice received daily intraperitoneal injections of PD98059 (● ■) or vehicle (○ □). The arrowhead indicates the time point of adoptive transfer of allogeneic C57BL/6-derived splenocytes. Mean values plus or minus SE of bidimensional tumor sizes of 5 mice per group are shown. Vehicle-treated cA2Kb-Cdc42 fibrosarcomas grew significantly faster than the other groups (P < .001, ANOVA; *P < .05, **P < .01, Tukey multiple comparison). (D) Immunblot analysis of extracts from cA2Kb-Cdc42 and cA2Kb-control fibrosarcomas derived from mice treated with PD98059 or vehicle. Note reduced ERK phosphorylation and decreased Cdc42-induced (brief exposure) as well as endogenous (long exposure) Bcl-2 expression following treatment with PD98059.

Figure 6

Cdc42 confers immunoresistance by ERK-dependent up-regulation of Bcl-2. (A) Immunoblot analysis of extracts prepared from 3 cA2Kb-Cdc42 and 3 cA2Kb-control fibrosarcomas. Note that phosphoepitopes indicative of ERK activation are detectable in Cdc42-expressing tumors. (B) cA2Kb-control MEFs (left panel) and cA2Kb-Cdc42 MEFs (right panel) were pretreated with the MEK inhibitor PD98059 (10 μM for 2 hours) or vehicle, followed by coincubation with allo-A2 CTL. Clonogenic survival in 1 representative of at least 3 repetitions. (C) Growth of cA2Kb-control (■ □) and cA2Kb-Cdc42 (● ○) fibrosarcomas in NOD/SCID mice. Mice received daily intraperitoneal injections of PD98059 (● ■) or vehicle (○ □). The arrowhead indicates the time point of adoptive transfer of allogeneic C57BL/6-derived splenocytes. Mean values plus or minus SE of bidimensional tumor sizes of 5 mice per group are shown. Vehicle-treated cA2Kb-Cdc42 fibrosarcomas grew significantly faster than the other groups (P < .001, ANOVA; *P < .05, **P < .01, Tukey multiple comparison). (D) Immunblot analysis of extracts from cA2Kb-Cdc42 and cA2Kb-control fibrosarcomas derived from mice treated with PD98059 or vehicle. Note reduced ERK phosphorylation and decreased Cdc42-induced (brief exposure) as well as endogenous (long exposure) Bcl-2 expression following treatment with PD98059.

Close modal

Based on these results, we reasoned whether pharmacologic MEK inhibition was a strategy to overcome Cdc42-mediated immunoresistance of cancer in vivo. To this end, cA2Kb-Cdc42 and cA2Kb-control fibrosarcomas were established in NOD/SCID mice. Daily intraperitoneal injections of PD98059 had no impact on the growth rate of these tumors, either in the presence or absence of cellular immune effectors (Figures 6C,S1C). Interestingly, treatment with PD98059 significantly delayed the growth rate of cA2Kb-Cdc42 fibrosarcomas in mice undergoing adoptive immunotherapy by transplantation of allogeneic C57BL/6 splenocytes (Figure 6C). These findings suggested that PD98059 sensitized cA2Kb-Cdc42 fibrosarcomas toward immune-mediated tumor suppression by modulating Cdc42 signaling to ERK. In support, cA2Kb-Cdc42 fibrosarcoma extracts prepared from NOD/SCID mice treated with PD98059 exhibited reduced ERK phosphorylation and Bcl-2 expression as compared with their vehicle-treated counterparts (Figure 6D). Thus, pharmacologic inhibition of Cdc42 signaling to the MEK-ERK pathway reduced Bcl-2 expression and sensitized established cancers to adoptive cellular immunotherapy.

The combination of target selectivity with long-term surveillance could make cellular immunotherapy an ideal cancer treatment. Accordingly, numerous approaches are explored to break tolerance to established cancers. Recent advances in the understanding of immune regulation have greatly added to the instrument, which can be applied to elicit an antigen-specific immune response in vivo. Further, the adoptive transfer of ex vivo–manipulated CTLs with improved target specificity is studied as a means to circumvent immunologic tolerance to cancer. A common prerequisite for these strategies is the assumption that once a cancer cell has successfully been targeted by lymphocytes, it will readily succumb to their various effector mechanisms. However, cancer cells can very well resist immune-mediated demise using cell-intrinsic resistance mechanisms, which parallel those conferring resistance to anticancer drugs and radiation.8,10-12  Hence, effective cancer immunotherapy may require sensitization of intrinsically resistant cancer cells to immune-mediated cytotoxicity. With the advancement of molecularly targeted therapies, the implementation of such a strategy has become feasible. Consequently, suitable targets have to be defined, which can be addressed to improve the efficacy of immunologic cancer treatments.

Against this background, we have devised functional screening and expression cloning to reveal Cdc42, a ubiquitously expressed Rho family GTPase, as mediator of resistance to CTL-mediated growth suppression of MEFs and HCT116 colorectal cancer cells. Cdc42 acts as a molecular switch that, upon activation by various cell surface receptors, cycles from the GDP-bound inactive state to the GTP-bound active state. Downstream effector pathways of Cdc42 regulate the actin cytoskeleton, migration, and cell-cycle progression.13-15  Numerous studies have implied Cdc42 in oncogenic transformation and tumor progression. Cdc42 function was required for full transformation of fibroblasts by oncogenic Ras,32  Cdc42 was expressed in multiple cancers and induced invasiveness of malignant cells.33,34  This was in keeping with the frequently observed overexpression of Cdc42 at the tumor invasive front and in particularly aggressive cancers.35-37 

Mice with targeted deletion of Cdc42 die at early embryonic stage, thus precluding the study of mammalian Cdc42 at an organismal level.38  However, the recent generation of conditional Cdc42-deficient and Cdc42GAP-deficient mice enabled the study of loss or gain of Cdc42 function in several cellular contexts. MEFs generated from these 2 mouse models featured defects in adhesion, migration, and establishment of cell polarity, thus confirming a critical role for the “fine-tuning” of Cdc42 signaling.39,40  Interestingly, surviving Cdc42GAP-deficient mice exhibited a phenotype reminiscent of premature aging. Enhanced Cdc42 activity in those mice either resulted in genomic instability, increased apoptosis, or cellular senescence, which was partially rescued by heterozygous loss of p53.41  Taken together, these findings are most likely explained by a replicative stress response to hyperactive Cdc42, which is similar to the one evoked by oncogenes.42  In turn, Cdc42-deficient MEFs exhibited defective MAPK signaling and impaired cell-cycle progression from G1 to S phase, demonstrating an important role of Cdc42 in growth factor receptor signal transduction.39 

In our experimental system, cA2Kb MEFs were transformed by the cooperating oncogenes Myc and H-Ras, and thus had already bypassed oncogene-induced senescence and apoptosis. Accordingly, these cells tolerated the expression of the Cdc42 (Q61L) mutant, which was reported to interfere with in vitro growth of primary fibroblasts or 3T3 cells.13  Despite resulting in enhanced ERK activation, constitutively active Cdc42 failed to accelerate proliferation of Myc/H-ras–transformed cA2Kb MEFs in vitro and in vivo. Further, pharmacologic MEK inhibition had no antiproliferative effect on these cells. Hence, Myc/H-ras–transformed cA2Kb MEFs apparently proliferated independently of Raf-MEK-ERK signaling and Cdc42. Nevertheless, we failed to establish cA2Kb MEFs with stable shRNA knock-down of endogenous Cdc42, suggesting that it was still required for additional cellular functions.

Cdc42 plays a key role in the regulation of actin dynamics.15  Recently, overexpression of ephrin-A1 and scinderin leading to reorganization of the actin cytoskeleton was reported to mediate resistance to CTL-induced tumor cell killing in vitro.43  However, cA2Kb-Cdc42 MEFs exhibited no apparent changes in cell morphology as compared with control cells, and were also protected against pharmacologic induction of cell death. Interestingly, we observed highly increased ERK activation in cA2Kb-Cdc42 MEFs, which correlated with resistance to cytotoxicity induced by CTL or proapoptotic drugs. MAPK signaling as well as constitutively active Cdc42 mutants have been shown to protect cells from death receptor-induced apoptosis.44,45  However, the CTL populations applied in our study predominantly act via granule-dependent effector mechanisms.11,22  Here, we have observed Cdc42-induced posttranscriptional stabilization of antiapoptotic Bcl-2, a known target of MAPK signaling.29,31  Pharmacologic inhibition of MEK reduced Bcl-2 levels and sensitized cA2Kb-Cdc42 MEFs to lymphocyte-induced tumor suppression in vitro and in vivo. Hence, Cdc42 conferred immunoresistance by MAPK-dependent inhibition of apoptosis, which may complement immune evasion through MAPK-induced secretion of immunosuppressive factors as observed in melanoma cells harboring the B-RafV600E mutation.46 

The online version of this article contains a data supplement.

The publication costs of this article were defrayed in part by page charge payment. Therefore, and solely to indicate this fact, this article is hereby marked “advertisement” in accordance with 18 USC section 1734.

We thank L. A. Sherman, P. Aspenström, S. W. Lowe, T. Kitamura, B. Vogelstein, and Abbott Laboratories for providing mice and reagents; S. Hoffarth, A. Hohberger, E. Antunes, R. Engel, A. Konur, and the staff of the Zentrale Versuchstiereinrichtung (central animal facility) for help with experiments; and all members of the Schuler and Theobald laboratories for support.

This work was supported by grants from the Deutsche Krebshilfe (no. 107993) and Deutsche Forschungsgemeinschaft (SCHU 1541/3-1).

National Institutes of Health

Contribution: C.A.M., P.S.H., S.T., and C.W. carried out research; C.W. and M.T. contributed unique reagents; M.T. and C.H. reviewed the data and the manuscript; and M.S. supervised the project, designed research, analyzed data, and wrote the paper.

Conflict-of-interest disclosure: The authors declare no competing financial interests.

Correspondence: Martin Schuler, Department of Medicine (Cancer Research), West German Cancer Center, University Hospital Essen, Hufelandstrasse 55, 45147 Essen, Germany; e-mail: martin.schuler@uk-essen.de.

1
Blattman
 
JN
Greenberg
 
PD
Cancer immunotherapy: a treatment for the masses.
Science
2004
, vol. 
305
 (pg. 
200
-
205
)
2
Appelbaum
 
FR
Haematopoietic cell transplantation as immunotherapy.
Nature
2001
, vol. 
411
 (pg. 
385
-
389
)
3
Bleakley
 
M
Riddell
 
SR
Molecules and mechanisms of the graft-versus-leukaemia effect.
Nat Rev Cancer
2004
, vol. 
4
 (pg. 
371
-
380
)
4
Dudley
 
ME
Wunderlich
 
JR
Yang
 
JC
, et al. 
Adoptive cell transfer therapy following non-myeloablative but lymphodepleting chemotherapy for the treatment of patients with refractory metastatic melanoma.
J Clin Oncol
2005
, vol. 
23
 (pg. 
2346
-
2357
)
5
Morgan
 
RA
Dudley
 
ME
Wunderlich
 
JR
, et al. 
Cancer regression in patients after transfer of genetically engineered lymphocytes.
Science
2006
, vol. 
314
 (pg. 
126
-
129
)
6
Leen
 
AM
Rooney
 
CM
Foster
 
AE
Improving T cell therapy for cancer.
Annu Rev Immunol
2007
, vol. 
25
 (pg. 
243
-
265
)
7
Rabinovich
 
GA
Gabrilovich
 
D
Sotomayor
 
EM
Immunosuppressive strategies that are mediated by tumor cells.
Annu Rev Immunol
2007
, vol. 
25
 (pg. 
267
-
296
)
8
Irmler
 
M
Thome
 
M
Hahne
 
M
, et al. 
Inhibition of death receptor signals by cellular FLIP.
Nature
1997
, vol. 
388
 (pg. 
190
-
195
)
9
Medema
 
JP
de Jong
 
J
Peltenburg
 
LTC
, et al. 
Blockade of the granzyme B/perforin pathway through overexpression of the serine protease inhibitor PI-9/SPI-6 constitutes a mechanism for immune escape by tumors.
Proc Natl Acad Sci U S A
2001
, vol. 
98
 (pg. 
11515
-
11520
)
10
Bots
 
M
Kolfschoten
 
IGM
Bres
 
SA
, et al. 
SPI-CI and SPI-6 cooperate in the protection from effector cell-mediated cytotoxicity.
Blood
2005
, vol. 
105
 (pg. 
1153
-
1161
)
11
Huber
 
C
Bobek
 
N
Kuball
 
J
, et al. 
Inhibitors of apoptosis confer resistance to tumour suppression by adoptively transplanted cytotoxic T lymphocytes in vitro and in vivo.
Cell Death Differ
2005
, vol. 
12
 (pg. 
317
-
325
)
12
Ravi
 
R
Fuchs
 
EJ
Jain
 
A
, et al. 
Resistance of cancers to immunologic cytotoxicity and adoptive immunotherapy via X-linked inhibitor of apoptosis protein expression and coexisting defects in mitochondrial death signaling.
Cancer Res
2006
, vol. 
66
 (pg. 
1730
-
1739
)
13
Cerione
 
RA
Cdc42: new roads to travel.
Trends Cell Biol
2004
, vol. 
14
 (pg. 
127
-
132
)
14
Hall
 
A
Rho GTPases and the control of cell behaviour.
Biochem Soc Trans
2005
, vol. 
33
 (pg. 
891
-
895
)
15
Ridley
 
AJ
Rho GTPases and actin dynamics in membrane protrusions and vesicle trafficking.
Trends Cell Biol
2006
, vol. 
16
 (pg. 
522
-
529
)
16
Vitiello
 
A
Marchesini
 
D
Furze
 
J
Sherman
 
LA
Chesnut
 
RW
Analysis of the HLA-restricted influenza-specific cytotoxic T lymphocyte response in transgenic mice carrying a chimeric human-mouse class I major histocompatibility complex.
J Exp Med
1991
, vol. 
173
 (pg. 
1007
-
1015
)
17
Theobald
 
M
Biggs
 
J
Hernandez
 
J
Lustgarten
 
J
Labadie
 
C
Sherman
 
LA
Tolerance to p53 by A2. 1-restricted cytotoxic T lymphocytes.
J Exp Med
1997
, vol. 
185
 (pg. 
833
-
841
)
18
Drexler
 
I
Antunes Ferreira
 
E
Schmitz
 
M
, et al. 
Modified vaccinia virus Ankara for delivery of human tyrosinase as melanoma-associated antigen: induction of tyrosinase- and melanoma-specific human leukocyte antigen A*0201-restricted cytotoxic T cells in vitro and in vivo.
Cancer Res
1999
, vol. 
59
 (pg. 
4955
-
4963
)
19
Theobald
 
M
Biggs
 
J
Dittmer
 
D
Levine
 
AJ
Sherman
 
LA
Targeting p53 as a general tumor antigen.
Proc Natl Acad Sci U S A
1995
, vol. 
92
 (pg. 
11993
-
11997
)
20
NCBI
Accessed 2006 
21
Aspenström
 
P
Lindberg
 
U
Hall
 
A
Two GTPases, Cdc42 and Rac, bind directly to a protein implicated in the immunodeficiency disorder Wiskott-Aldrich syndrome.
Curr Biol
1996
, vol. 
6
 (pg. 
70
-
75
)
22
Hähnel
 
PS
Hoffarth
 
S
Thaler
 
S
Huber
 
C
Theobald
 
M
Schuler
 
M
Targeting protein kinase B to improve therapeutic efficacy of adoptively transplanted immune effectors.
Onkologie
2006
, vol. 
29
 pg. 
49
 
23
Sutton
 
VR
Davis
 
JE
Cancilla
 
M
, et al. 
Initiation of apoptosis by granzyme B requires direct cleavage of bid, but not direct granzyme B-mediated caspase activation.
J Exp Med
2000
, vol. 
192
 (pg. 
1403
-
1414
)
24
Cory
 
S
Adams
 
JM
The Bcl2 family: regulators of the cellular life-or-death switch.
Nat Rev Cancer
2002
, vol. 
2
 (pg. 
647
-
656
)
25
Green
 
DR
Kroemer
 
G
The pathophysiology of mitochondrial cell death.
Science
2004
, vol. 
305
 (pg. 
626
-
629
)
26
Oltersdorf
 
T
Elmore
 
SW
Shoemaker
 
AR
, et al. 
An inhibitor of Bcl-2 family proteins induces regression of solid tumours.
Nature
2005
, vol. 
435
 (pg. 
677
-
681
)
27
Willis
 
SN
Fletcher
 
JI
Kaufmann
 
T
, et al. 
Apoptosis initiated when BH3 ligands engage multiple Bcl-2 homologs, not Bax or Bak.
Science
2007
, vol. 
315
 (pg. 
856
-
859
)
28
van Delft
 
MF
Wei
 
AH
Mason
 
KD
, et al. 
The BH3 mimetic ABT-737 targets selective Bcl-2 proteins and efficiently induces apoptosis via Bak/Bax if Mcl-1 is neutralized.
Cancer Cell
2006
, vol. 
10
 (pg. 
389
-
399
)
29
Konopleva
 
M
Contractor
 
R
Tsao
 
T
, et al. 
Mechanisms of apoptosis sensitivity and resistance to the BH3 mimetic ABT-737 in acute myeloid leukemia.
Cancer Cell
2006
, vol. 
10
 (pg. 
375
-
388
)
30
Wesarg
 
E
Hoffarth
 
S
Wiewrodt
 
R
, et al. 
Targeting BCL-2 family proteins to overcome drug resistance in non-small cell lung cancer.
Int J Cancer
2007
, vol. 
121
 (pg. 
2387
-
2394
)
31
Deng
 
X
Ruvolo
 
P
Carr
 
B
May
 
WS
Survival function of ERK1/2 as IL-3-activated, staurosporine-resistant Bcl2 kinases.
Proc Natl Acad Sci U S A
2000
, vol. 
97
 (pg. 
1578
-
1583
)
32
Qiu
 
RG
Abo
 
A
McCormick
 
F
Symons
 
M
Cdc42 regulates anchorage-independent growth and is necessary for Ras transformation.
Mol Cell Biol
1997
, vol. 
17
 (pg. 
3449
-
3458
)
33
Fritz
 
G
Just
 
I
Kaina
 
B
Rho GTPases are over-expressed in human tumors.
Int J Cancer
1999
, vol. 
81
 (pg. 
682
-
687
)
34
Keely
 
PJ
Westwick
 
JK
Whitehead
 
IP
Der
 
CJ
Parise
 
LV
Cdc42 and Rac1 induce integrin-mediated cell motility and invasiveness through PI(3)K.
Nature
1997
, vol. 
390
 (pg. 
632
-
636
)
35
Vasko
 
V
Espinosa
 
AV
Scouten
 
W
, et al. 
Gene expression and functional evidence of epithelial-to-mesenchymal transition in papillary thyroid carcinoma invasion.
Proc Natl Acad Sci U S A
2007
, vol. 
104
 (pg. 
2803
-
2808
)
36
Kamai
 
T
Yamanishi
 
T
Shirataki
 
H
, et al. 
Overexpression of RhoA, Rac1, and Cdc42 GTPases is associated with progression in testicular cancer.
Clin Cancer Res
2004
, vol. 
10
 (pg. 
4799
-
4805
)
37
Wang
 
W
Goswami
 
S
Sahai
 
E
Wyckoff
 
JB
Segall
 
JE
Condeelis
 
JS
Tumor cells caught in the act of invading: their strategy for enhanced cell motility.
Trends Cell Biol
2005
, vol. 
15
 (pg. 
138
-
145
)
38
Chen
 
F
Ma
 
L
Parrini
 
MC
, et al. 
Cdc42 is required for PIP2-induced actin polymerization and early development but not for cell viability.
Curr Biol
2000
, vol. 
10
 (pg. 
758
-
765
)
39
Yang
 
L
Wang
 
L
Zheng
 
Y
Gene targeting of Cdc42 and Cdc42GAP affirms the critical involvement of Cdc42 in filopodia induction, directed migration, and proliferation in primary mouse embryonic fibroblasts.
Mol Biol Cell
2006
, vol. 
17
 (pg. 
4675
-
4685
)
40
Wang
 
L
Yang
 
L
Burns
 
K
Kuan
 
CY
Zheng
 
Y
Cdc42GAP regulates c-Jun N-terminal kinase (JNK)-mediated apoptosis and cell number during mammalian perinatal growth.
Proc Natl Acad Sci U S A
2005
, vol. 
102
 (pg. 
13484
-
13489
)
41
Wang
 
L
Yang
 
L
Debidda
 
M
Witte
 
D
Zheng
 
Y
Cdc42 GTPase-activating protein deficiency promotes genomic instability and premature aging-like phenotypes.
Proc Natl Acad Sci U S A
2007
, vol. 
104
 (pg. 
1248
-
1253
)
42
Bartkova
 
J
Rezaei
 
N
Liontos
 
M
, et al. 
Oncogene-induced senescence is part of the tumorigenesis barrier imposed by DNA damage checkpoints.
Nature
2006
, vol. 
444
 (pg. 
633
-
637
)
43
Abouzahr
 
S
Bismuth
 
G
Gaudin
 
C
, et al. 
Identification of target actin content and polymerization status as a mechanism of tumor resistance after cytolytic T lymphocyte pressure.
Proc Natl Acad Sci U S A
2006
, vol. 
103
 (pg. 
1428
-
1433
)
44
Holmström
 
TH
Schmitz
 
I
Soderström
 
TS
, et al. 
MAPK/ERK signaling in activated T cells inhibits CD95/Fas-mediated apoptosis downstream of DISC assembly.
EMBO J
2000
, vol. 
19
 (pg. 
5418
-
5428
)
45
Tu
 
SS
Wu
 
WJ
Yang
 
W
Nolbant
 
P
Hahn
 
K
Cerione
 
RA
Antiapoptotic Cdc42 mutants are potent activators of cellular transformation.
Biochemistry
2002
, vol. 
41
 (pg. 
12350
-
12358
)
46
Sumimoto
 
H
Imabayashi
 
F
Iwata
 
T
Kawakami
 
Y
The BRAF-MAPK signaling pathway is essential for cancer-immune evasion in human melanoma cells.
J Exp Med
2006
, vol. 
203
 (pg. 
1651
-
1656
)

Supplemental data

Sign in via your Institution