<<

RESEARCH BRIEF

HER2 Activating Mutations Are Targets for Treatment

Shyam M. Kavuri 1 , Naveen Jain 1 , Francesco Galimi 2,3 , Francesca Cottino 3 , Simonetta M. Leto 2,3 , Giorgia Migliardi 2,3 , Adam C. Searleman 1 , Wei Shen 1, John Monsey 1 , Livio Trusolino 2,3 , Samuel A. Jacobs 4 , Andrea Bertotti 2,3,5 , and Ron Bose 1,6

ABSTRACT The Cancer Genome Atlas project identifi ed HER2 somatic mutations and gene amplifi cation in 7% of patients with colorectal cancer. Introduction of the HER2 mutations S310F, L755S, V777L, V842I, and L866M into colon epithelial cells increased signaling pathways and anchorage-independent cell growth, indicating that they are activating mutations. Intro- duction of these HER2 activating mutations into colorectal cancer cell lines produced resistance to and panitumumab by sustaining MAPK phosphorylation. HER2 mutants are potently inhib- ited by low nanomolar doses of the irreversible inhibitors and . HER2 gene sequencing of 48 cetuximab-resistant, quadruple (KRAS, NRAS, BRAF, and PIK3CA) wild-type (WT) colorectal cancer patient-derived xenografts (PDX) identifi ed 4 PDXs with HER2 mutations. HER2- targeted therapies were tested on two PDXs. Treatment with a single HER2-targeted drug (trastuzu- mab, neratinib, or ) delayed tumor growth, but dual HER2- with plus tyrosine kinase inhibitors produced regression of these HER2-mutated PDXs.

SIGNIFICANCE: HER2 activating mutations cause EGFR antibody resistance in colorectal cell lines, and PDXs with HER2 mutations show durable tumor regression when treated with dual HER2-targeted therapy. These data provide a strong preclinical rationale for clinical trials targeting HER2 activating mutations in metastatic colorectal cancer. Cancer Discov; 5(8); 832–41. ©2015 AACR.

See related commentary by Pectasides and Bass, p. 799.

INTRODUCTION found that 7% of colorectal cancer patients have HER2 somatic mutations or HER2 gene amplifi cation (3 ). HER2 Cancer genome sequencing is identifying new genetic gene amplifi cation in colorectal cancer is known to produce alterations and new driver events in human cancers ( 1, 2 ). resistance to the EGFR monoclonal antibodies cetuximab The Cancer Genome Atlas (TCGA) colorectal cancer project and panitumumab ( 4, 5 ). To our knowledge, the impact of

1Division of Oncology, Department of Medicine, Washington University Corresponding Authors: Ron Bose, Washington University School of Medi- School of Medicine, St. Louis, Missouri. 2 Department of Oncology, Univer- cine, 660 S. Euclid Avenue, Campus Box 8069, St. Louis, MO 63110. Phone: sity of Torino Medical School, Torino, Italy. 3Translational Cancer Medicine, 314-747-9308; Fax: 314-747-9320; E-mail: [email protected] ; and Candiolo Cancer Institute – FPO IRCCS, Torino, Italy. 4NSABP Foundation, Andrea Bertotti, Translational Cancer Medicine, Candiolo Cancer Institute – Pittsburgh, Pennsylvania. 5National Institute of Biostructures and Biosys- FPO IRCCS, 10060 Candiolo, Torino, Italy. Phone 39-011-993-3242; Fax: tems, Rome, Italy. 6 Alvin J. Siteman Cancer Center, Washington University 39-011-993-3225; E-mail: [email protected] School of Medicine, St. Louis, Missouri. doi: 10.1158/2159-8290.CD-14-1211 Note: Supplementary data for this article are available at Cancer Discovery © 2015 American Association for Cancer Research. Online (http://cancerdiscovery.aacrjournals.org/). Current address for S.M. Kavuri: Lester and Sue Smith Breast Cancer Center, Baylor College of Medicine, Houston, TX.

832 | CANCER DISCOVERYAUGUST 2015 www.aacrjournals.org

Downloaded from cancerdiscovery.aacrjournals.org on September 26, 2021. © 2015 American Association for Cancer Research. HER2 Activating Mutations Are Targets in Colorectal Cancer RESEARCH BRIEF

HER2 somatic mutations in colorectal cancer has not been (KRAS, NRAS, BRAF, PIK3CA; Supplementary Table S1). studied, and it is an open question as to whether HER2 muta- The co-occurring KRAS mutations included 4 cases of codon tions are clinically important in colorectal cancer. 12/13 mutations and 2 cases of exon 4 mutations (KRASK117N HER2 somatic mutations have been previously studied and KRAS A146T). The co-occurring BRAF mutations in HER2- in breast cancer and non–small cell lung cancer (NSCLC). mutated cases included one case with BRAFV600E (TCGA- The majority of these mutations were shown to be activat- AA-3947) and one case with BRAFF247L (TCGA-AG-A002). The ing based on their ability to increase intracellular signaling, BRAF F247L mutation is located in the C1 domain of BRAF; it induce oncogenic transformation, and accelerate xenograft has not been reported in any other sample in the cBioPortal tumor growth ( 6–8 ). HER2 activating mutations tend to fall and Catalogue of Somatic Mutations in Cancer (COSMIC) in several hotspots (residues 309–310 in the extracellular databases to date, and, to our knowledge, it has not been domain and residues 755–781 and 842 in the kinase domain), functionally characterized to date ( 16, 17 ). HER2-mutated and they are responsive to HER2 tyrosine kinase inhibitors ( 6, colorectal cancers occurred in both the right and left sides of 7 ). These preclinical data have led to two multi-institutional, the colon as well as in the rectum (Supplementary Table S1). phase II clinical trials that will screen patients with metastatic The microsatellite stability (MSS) or instability (MSI) status of breast cancer for HER2 mutations and treat the mutation- these cancers was reported, and 83% (10/12) were MSS (Sup- positive patients with the second-generation HER2/EGFR plementary Table S1). MSS colorectal cancers have a worse tyrosine kinase inhibitor neratinib (9, 10). Further, phase I prognosis than MSI colorectal cancers and show clinical ben- and II clinical trials for HER2 mutations in NSCLC are dem- efi t from 5-fl uorouracil–containing adjuvant onstrating the clinical effi cacy of combining neratinib with (18, 19). the mTOR inhibitor ( 11, 12 ). To analyze the effects of HER2 mutations, we introduced In this study, we determined the effect of HER2 somatic these mutations into Immortalized Mouse Colon Epithelial mutations in colorectal cancer. The HER2 mutations found (IMCE) cells and measured their effect on cell signaling and in colorectal cancer are similar to those found in breast can- anchorage-independent growth. IMCE cells are nontrans- cer. We demonstrate that these HER2 mutations cause onco- formed colon epithelial cells and can be transformed by the genic transformation of colon epithelial cells and produce introduction of oncogenes (20, 21). We stably introduced WT resistance to cetuximab and panitumumab in two colorectal or four HER2 mutations into IMCE cells using a retroviral cancer cell lines. We identifi ed HER2 activating mutations vector ( Fig. 1C ). All four HER2 mutations increased HER2 in colorectal cancer patient-derived xenografts (PDX) and signaling pathways, with increased HER2, MAPK, and AKT demonstrated that dual HER2-targeted therapy causes tumor phosphorylation seen relative to the HER2WT transduced cells regression. These data form a strong preclinical rationale ( Fig. 1C ). Phosphorylation of the immediate HER2 substrate, for clinical trials targeting HER2 activating mutations in phospholipase γ C1 (PLCγ), was greatest in the HER2V777L cells, patients with metastatic colorectal cancer. but was also increased in the other HER2 mutations. The effect of HER2 mutations on soft-agar colony formation was also RESULTS tested. All HER2 mutations dramatically increased the number of colonies formed in soft agar, demonstrating enhanced HER2 Mutations Identifi ed from Patients anchorage-independent growth ( Fig. 1D ; Supplementary Fig. with Colorectal Cancer Cause Oncogenic S1A). Interestingly, in our prior study that used an MCF10A Transformation of Colon Epithelial Cells immortalized breast epithelial cell line, the HER2L755S muta- The TCGA colorectal cancer project identifi ed HER2 altera- tion did not increase soft-agar colony formation or alter colony tions in 7% (14/212) of cases. Six cases had HER2 somatic morphology in Matrigel ( 6 ). This difference in soft-agar colony mutations, fi ve had HER2 gene amplifi cation, and three had formation between MCF10A–HER2L755S and IMCE–HER2L755S both HER2 mutations and HER2 amplifi cation ( Fig. 1A ). likely is due to subtle differences between the two cell lines. The Concurrent mutation and amplifi cation have been described effect of trastuzumab and neratinib on soft-agar colony forma- for other oncogenes, including RAS and EGFR ( 13 ). In addi- tion was also tested ( Fig. 1D ). Trastuzumab produced statisti- tion to the TCGA colorectal cancer study, a recent sequenc- cally signifi cant reductions in colony formation with L755S, ing study on patients with colorectal cancer performed at L866M, and S310F mutations, whereas neratinib prevented Memorial Sloan Kettering Cancer Center identifi ed 3 more colony formation with all of the mutations tested here. The HER2-mutated cases ( 14 ). The HER2 mutations reported in effect of neratinib on IMCE–KRAS cells was also tested. IMCE– both studies are combined in Fig. 1B, and several of these KRAS cells are relatively resistant to neratinib, with no effect mutations are identical to HER2 mutations found in patients on soft-agar colony formation at 50 to 100 nmol/L neratinib, V842I with breast cancer, including the kinase domain mutations whereas IMCE–HER2 cells show an IC50 of approximately V842I, V777L, and L755S, and the extracellular domain muta- 10 nmol/L neratinib in the soft-agar colony-formation assay tion S310F. Unlike the HER2 kinase domain mutations found (Supplementary Fig. S1B). in NSCLC, no HER2 kinase domain in-frame insertions/dele- tions were reported in these two colorectal cancer sequencing HER2 Mutations Cause Resistance studies ( 8 , 15 ). The HER2 mutations V777L and V842I were to EGFR Monoclonal Antibodies in seen in multiple patients, with 2 cases with HER2V777L and 4 Colorectal Cancer Cell Lines cases with HER2V842I identifi ed (Fig. 1B). Half (6/12) of these To determine whether HER2 mutations cause resistance HER2-mutated colorectal cancer cases are KRAS wild-type to the EGFR monoclonal antibodies cetuximab and panitu- (WT), and one third (4/12) of these cases are quadruple WT mumab, we fi rst introduced the HER2V842I mutation into the

AUGUST 2015CANCER DISCOVERY | 833

Downloaded from cancerdiscovery.aacrjournals.org on September 26, 2021. © 2015 American Association for Cancer Research. RESEARCH BRIEF Kavuri et al.

A Any HER2 alterations in B colorectal cancer TCGA ERBB2 Amp+ TCGA ERBB2 Amp− V777L V842I 14/212 = 7% MSKCC Amp N/A 2 patients 4 patients

S310F L755S Ampli- Mutation fication I263T A466T R678Q R868W N1219S 6 35 Recep_L FU Recep_L FU FU TM Tyrosine kinase 212 212 212 0 200 400 600 800 1,000 1,200 Mutation position

755S WT V777L V842I L S310F MW CDParental HER2 HER2 HER2 HER2 HER2 [kDa] Phospho HER2 150 500 Phospho Mock γ * PLC 450 Trastuzumab 150 ns Phospho 400 * Neratinib MAPK 37 † Phospho 350 AKT * 50 300 † HER2 250 150 Lighter 200 * expo. 150 Number of colonies 150 γ PLC 100 *ns 150 50 † MAPK † †††††† 37 0 AKT WT L755S V777L V842I L866M S310F 50

Actin 37

Figure 1. HER2 mutations identifi ed by colorectal cancer genome sequencing studies increase cell signaling and anchorage-independent growth in a colonic epithelial cell line. A, HER2 alterations identifi ed by the TCGA colorectal cancer project. B, HER2 somatic mutations observed in 12 patients with colorectal cancer are shown. Red circles, TCGA cases with HER2 gene amplifi cation; blue circles, TCGA cases that do not have HER2 gene amplifi cation; green circles, cases from Brannon et al. (14 ), and gene amplifi cation is not reported on these cases. One TCGA case had concurrent V842I plus V777L mutations along with gene amplifi cation. FU, Furin-like domains; TM, transmembrane region. C, IMCE cells were retrovirally transduced with HER2 WT or mutants. Total lysates (8–10 μg) were analyzed by Western blot. D, IMCE HER2 WT or HER2 mutants were seeded in soft agar in duplicate, treated with trastuzumab (100 μg/mL) or neratinib (500 nmol/L), allowed to grow for 12 days, and stained with crystal violet. Photographs of the stained wells are shown in Supplementary Fig S1A. *, statistically signifi cant from HER2 WT at >99% probability; †, statistically signifi cant from mock treated at >99% probability; ns, not signifi cant as compared with mock treated. cetuximab-sensitive colorectal cell line DiFi using a retroviral Introduction of HER2 mutations into these cells increased vector (Fig. 2A and B). We initially focused on the HER2V842I MAPK and EGFR phosphorylation, and this was sustained mutation because it was the most prevalent mutation identi- even in the presence of cetuximab. fi ed in colorectal cancer cases by TCGA (3 ). Introduction of HER2WT into DiFi cells caused a modest (2–4-fold) change HER2 Mutants Are Highly Sensitive to the in the cells’ sensitivity to cetuximab and panitumumab (red Irreversible HER2/EGFR Tyrosine Kinase curves, Fig. 2A and B). In contrast, introduction of the V842I Inhibitors Neratinib and Afatinib mutation caused a 40- to 100-fold shift in the IC50 values Prior studies in breast cancer and NSCLC showed that (blue curves, Fig. 2A and B ). NCI-H508 is another cetuximab- HER2 mutations can be potently inhibited by nanomolar sensitive, colorectal cancer cell line, and they are more readily doses of neratinib or afatinib, which are irreversible HER2/ transduced with retroviral vectors than DiFi cells. Five HER2 EGFR tyrosine kinase inhibitors ( 6, 7 ). We therefore tested mutations were tested in NCI-H508 cells, and all mutations the effect of neratinib and afatinib on DiFi and NCI-H508 produced resistance to cetuximab and panitumumab ( Fig. 2C cells transduced with HER2WT or HER2 mutations. Neratinib and D). Western blots of the EGFR–HER2 signaling pathways and afatinib inhibited the growth of parental DiFi cells, DiFi– WT V842I suggest the mechanism of EGFR antibody drug resistance. HER2 cells, and DiFi–HER2 cells, with IC 50 values Cetuximab treatment of parental DiFi or NCI-H508 cells ranging from 2 to 5 nmol/L (Fig. 3A and B). Similarly, growth reduced MAPK and EGFR phosphorylation ( Fig. 2E and F ). of parental NCI-H508 cells and cells transduced with WT or

834 | CANCER DISCOVERYAUGUST 2015 www.aacrjournals.org

Downloaded from cancerdiscovery.aacrjournals.org on September 26, 2021. © 2015 American Association for Cancer Research. HER2 Activating Mutations Are Targets in Colorectal Cancer RESEARCH BRIEF

A B

120 µ IC50 ( g/mL) 120 IC50 (µg/mL) + − Parental 0.030 / 0.005 Parental 0.031 +/− 0.004 WT + − 100 HER2 0.078 / 0.026 HER2WT 0.14 +/− 0.032 V842I + − 100 HER2 2.7 / 1.6 HER2V842I 1.26 +/− 0.50 80 80

60 60

DiFi cells 40

% Cell growth % Cell growth 40

20 20

0 0 0.001 0.01 0.1 1 10 100 1,000 0.001 0.01 0.1 1 10 100 1,000 Cetuximab (µg/mL) Panitumumab (µg/mL) CD 120 120

100 100

80 80

60 60 IC (µg/mL) 50 IC50 (µg/mL) + − Parental 0.42 / 0.04 40 Parental 0.18 +/− 0.02 40 WT 1.30 +/− 0.10 WT 0.31 +/− 0.03 % Cell growth V842I > 70 % Cell growth > 70

NCl-H508 cells V842I L866M > 200 L866M > 200 20 L755S > 200 20 L755S > 200 V777L > 200 V777L > 200 S310F > 200 S310F > 200 0 0 0 0.01 0.1 1 10 100 0 0.01 0.1 1 10 100 Cetuximab (µg/mL) Panitumumab (µg/mL)

EFDiFi cells NCI-H508 cells

WT V842I Parental HER2 HER2 Parental HER2WT HER2V777L HER2V842I HER2L755S HER2S310F HER2L866M Cetuximab Cetuximab MW µ .5 (µg/mL) 20 0 20 20 20 ( g/ml) 0 0.1 0.25 0.5 0 0.1 0.25 0 0 0.1 0.25 0.5 [kDa] 0 5 10 0 5 10 2 0 5 10 20 0 5 10 0 5 10 0 5 10 0 5 10 20 Phospho Phospho MAPK p44/p42 37 MAPK Phospho MAPK HER2 150 Phospho Phospho EGFR HER2 150 Phospho HER2 AKT 50 Phospho MAPK EGFR 37 HER2 EGFR 150 EGFR Actin 150 AKT 50 Actin 37

Figure 2. HER2 mutations cause resistance to EGFR antibodies. A and B, DiFi cell lines were treated with cetuximab (A) or panitumumab (B) for 5 days, and cell growth was measured by Alamar blue. C and D, NCI-H508 cell lines were treated with cetuximab (C) or panitumumab (D) for 5 days, and cell growth was measured by crystal violet assay. E, DiFi parental, HER2WT , or HER2V842I cells were treated with cetuximab for 24 hours and then lysed. Cell lysates were analyzed by Western blot. F, identical experiment performed on NCI-H508 cells.

mutant HER2 was inhibited by neratinib and afatinib, with The effect of neratinib and afatinib on cell signaling was

IC50 values ranging from 0.2 to 3 nmol/L (Fig. 3C and D). tested. Both drugs strongly inhibited HER2, EGFR, AKT, and Parental DiFi and NCI-H508 cells are EGFR-dependent cell MAPK phosphorylation in DiFi and NCI-H508 cells ( Fig. 3E lines, and their growth is inhibited by neratinib or afatinib and F ). The effect of neratinib or cetuximab on NCI-H508 because these drugs inhibit both EGFR and HER2. cell growth was also tested in vivo using cell line xenografts

AUGUST 2015CANCER DISCOVERY | 835

Downloaded from cancerdiscovery.aacrjournals.org on September 26, 2021. © 2015 American Association for Cancer Research. RESEARCH BRIEF Kavuri et al.

AB 120 IC50 (nmol/L) IC50 (nmol/L) Parental + − 5.5 / 1.1 120 Parental + − WT + − 1.8 / 0.7 HER2 3.0 / 0.5 WT + − 100 V842I HER2 2.4 / 0.6 HER2 2.4 +/− 0.4 V842I 100 HER2 3.6 +/− 0.9 80 80 60 60 DiFi cells

40 % Cell growth % Cell growth 40

20 20

0 0 0.01 0.1 1 10 100 1,000 0.01 0.1 1 10 100 1,000 Neratinib (nmol/L) Afatinib (nmol/L) CD

120 IC (nmol/L) 120 50 IC50 (nmol/L) 1.20 +/− 0.10 0.40 +/− 0.06 100 0.56 +/− 0.05 100 0.30 +/− 0.04 1.90 +/− 0.30 0.42 +/− 0.05 0.16 +/− 0.01 0.19 +/− 0.03 2.80 +/− 0.20 3.00 +/− 0.30 80 0.29 +/− 0.04 80 0.56 +/− 0.06 0.20 +/− 0.03 0.29 +/− 0.03 60 60

40 % Cell growth 40 % Cell growth NCI-H508 cells 20 20

0 0 0 0.001 0.01 0.1 1 10 100 1,000 0 0.01 0.1 1 10 100 1,000 Neratinib (nmol/L) Afatinib (nmol/L)

DiFi cells NCI-H508 cells

EFWT V842I 7L 55S 10F 77 842I WT V V L7 S3 L866M Parental HER2 HER2

Parental HER2 HER2 HER2 HER2 HER2 HER2

ratinib MW 0 NeratinibAfatinib 0 NeratinibAfatinib 0 Ne Afatinib [kDa] inib inib b inib MW Phospho fatinib fatinib fatinib erat fatini 0 NeratinibA 0 Nerat A 0 NeratinibAfatinib0 NeratinibA 0 NeratinibAfat 0 NeratinibAfatinib0 N A [kDa] MAPK 37 Phospho MAPK Phospho 37 HER2 Phospho 150 HER2 Phospho 150 Phospho EGFR 150 EGFR Phospho 150 MAPK AKT 50 37 MAPK HER2 37 150 HER2 EGFR 150 150 Actin EGFR 37 150 AKT 50 Actin 37

Figure 3. HER2 mutants are highly sensitive to second-generation, irreversible HER2/EGFR tyrosine kinase inhibitors. A and B, DiFi cell lines were treated with neratinib (A) or afatinib (B) for 5 days, and cell growth was measured by Alamar blue. C and D, NCI-H508 cell lines were treated with neratinib (C) or afatinib (D) for 5 days, and cell growth was measured by crystal violet assay. E, DiFi parental, HER2WT , or HER2V842I cells were treated with neratinib (0.5 μmol/L) or afatinib (0.5 μmol/L) for 4 hours, and total lysates (8–10 μg) were harvested and analyzed by Western blot. F, identical experiment per- formed on NCI-H508 cells.

836 | CANCER DISCOVERYAUGUST 2015 www.aacrjournals.org

Downloaded from cancerdiscovery.aacrjournals.org on September 26, 2021. © 2015 American Association for Cancer Research. HER2 Activating Mutations Are Targets in Colorectal Cancer RESEARCH BRIEF

(Supplementary Fig. S2). Neratinib or the combination of zumab, neratinib, or lapatinib alone delayed tumor growth, neratinib plus trastuzumab inhibited the growth of NCI- but after 30 days, the mice developed large tumors and had H508 cells transduced with HER2 V777L . Cetuximab inhibited to be sacrifi ced. In contrast, dual HER2-targeted therapy the growth of NCI-H508 parental cells (Supplementary Fig. with either trastuzumab plus neratinib or trastuzumab plus S2A), whereas NCI-H508–HER2 V777L and HER2 WT cells grew lapatinib produced tumor regression and absence of tumor in the presence of cetuximab (Supplementary Fig. S2B and regrowth during the 41-day window of this experiment. For S2C). PDX M051, which has HER2L866M kinase domain mutation Comparison of the effect of neratinib between KRASWT and plus HER2 gene amplifi cation (Fig. 4D), treatment with mutant colorectal cancer cell lines was made (Supplementary trastuzumab had minimal effect on tumor growth. Neratinib Fig. S3). DiFi cells are KRAS, NRAS, BRAF, and PIK3CA WT as a single agent resulted in stable tumor size, whereas the (22 ). NCI-H508 cells are KRASWT and NRAS WT, have an inac- combination of trastuzumab plus neratinib caused tumor tivating BRAF mutation (G596R), and have a PIK3CAE545K regression, which was sustained over the duration of the helical domain mutation ( 23, 24 ). SW480 and HCT116 color- experiment. After the fi nal time point in both PDX experi- ectal cancer cells have KRAS G12V and G13D mutations, ments, the mice were sacrifi ced and the tumors excised. The respectively (23 ). These KRAS-mutated cell lines are relatively tumor histology with both PDXs demonstrates that dual

resistant to neratinib (IC50 values of 430 nmol/L) compared HER2-targeted therapy caused reduction in tumor cellular- with the KRASWT cell lines, paralleling the results obtained ity and acquisition of more differentiated features (Supple- with IMCE–KRAS cells (Supplementary Figs. S1B and S3). mentary Figs. S5 and S6). IHC on PDX M122 showed that These results show that HER2-mutated cell lines, but not treatment with neratinib or lapatinib (alone and, to a greater KRAS-mutated cell lines, are sensitive to the tyrosine kinase extent, when combined with trastuzumab) strongly reduced inhibitors neratinib and afatinib. Ki-67, phosphoMAPK, and phosphoS6 immunoreactivity (Supplementary Figs. S5 and S6) but did not induce detect- Colorectal PDXs with HER2 Mutations able signs of (not shown). Trastuzumab alone was Multiple mechanisms of resistance to EGFR antibodies poorly effective at decreasing Ki-67, phosphoMAPK, and have been reported, such as mutations in KRAS, NRAS, phosphoS6 levels. This lack of pharmacodynamic activity of BRAF, and PIK3CA, or gene amplifi cations in HER2 and trastuzumab alone was particularly evident in the IHC results MET (4 , 25 ). Cetuximab response rate in patients lacking from PDX M051, consistent with less therapeutic effi cacy in these genetic alterations is approximately 20% to 25%, sug- vivo (Supplementary Figs. S5 and S6). gesting that there are additional factors contributing to drug In order to understand this difference in trastuzumab resistance (4 ). We sequenced the HER2 gene in 48 colorectal effect between these two PDXs, we confi rmed that HER2L866M cancer PDX samples that are cetuximab resistant and are activated intracellular signaling pathways in IMCE and NCI- WT for KRAS, NRAS, BRAF, and PIK3CA (quadruple WT). H508 cells, produced resistance to EGFR monoclonal anti- Four of these PDXs had HER2 mutations, and the allele fre- bodies, and was sensitive to neratinib or afatinib in DiFi and quency of the HER2 mutation in the primary tumor (prior NCI-H508 cells, similar to the other HER2 mutations tested to implantation) and in the xenograft grown in the mice was in this study (Supplementary Fig. S7; Figs. 2 and 3 ). Compari- measured by next-generation DNA sequencing (Fig. 4A). The son of trastuzumab sensitivity of HER2L866M versus HER2S310F HER2S310Y mutation, found in PDX M122, was previously mutation transduced cells suggests that the S310F mutation shown to be an activating mutation ( 7 ) and functions the may have greater sensitivity to trastuzumab. NCI-H508 cells same as the S310F mutation studied in IMCE cells (Fig. 1C with HER2S310F were more sensitive to trastuzumab than and D ). The allele frequency of this mutation increased in HER2 WT transduced cells, whereas resistance to trastuzumab the PDX, likely due to enrichment of malignant cells in the was produced in cells transduced with HER2L866M (Supple- xenograft relative to the primary tumor. PDX M051 had both mentary Fig. S8). Similarly, in soft-agar assays on IMCE cells, HER2 amplifi cation and a novel kinase domain mutation, trastuzumab had a greater effect on S310F-containing cells L866M. The allele frequency of L866M (0.968 to 0.986) indi- than L866M cells (Fig. 1D). Trastuzumab has multiple mech- cates that the mutation is located on the amplifi ed copies of anisms of action on HER2-expressing cells (26 ). Detailed the HER2 gene. HER2L866M is homologous to the EGFRL858R studies on these mechanisms are beyond the scope of this mutation, which is a well-known EGFR-activating mutation study and will be examined in the future. found in NSCLC ( Fig. 4B ; ref. 15 ). An in vitro kinase assay To assess the specifi city of these HER2-targeted drugs in demonstrated that HER2L866M produced a 3-fold increase in this colorectal cancer PDX model system, we tested the effects tyrosine kinase activity relative to HER2WT (Fig. 4B). Both of cetuximab, neratinib, and trastuzumab plus neratinib on PDX M102 and M107 contained HER2V777L kinase domain a KRAS-mutant PDX ( Fig. 4E ). Unlike HER2-mutant PDXs, mutations, and the allele frequency of 0.315 to 0.324 in M107 the KRAS-mutant PDX (PDX M551) continued to grow when may represent a subclonal mutation. Cetuximab treatment of treated with neratinib or trastuzumab plus neratinib. KRAS these four PDXs was previously performed ( 4 ) and demon- mutation is a known mechanism of resistance to cetuximab, strated that these PDXs have de novo resistance to cetuximab and compared with the cetuximab treatment arm, the nerat- (Supplementary Fig. S4A–S4D). inib or trastuzumab plus neratinib arms had similar or slightly We tested the effect of HER2-targeted drugs on PDX M122 greater tumor growth. In total, these PDX experiments sug- and M051. PDX M102 and M107 had previously been cryop- gest that dual HER2-targeted therapy with trastuzumab plus reserved and could not be recovered during the timeframe of neratinib may be an effective treatment for HER2-mutated, this project. For PDX M122 ( Fig. 4C ), treatment with trastu- but not KRAS-mutated, colorectal cancers.

AUGUST 2015CANCER DISCOVERY | 837

Downloaded from cancerdiscovery.aacrjournals.org on September 26, 2021. © 2015 American Association for Cancer Research. RESEARCH BRIEF Kavuri et al.

A B 30

PDX M122 M051 M102 M107 25

20 L866M + HER2 mutation S310Y V777L V777L amplification 15 10

Primary Specific activity 0.787 0.968 0.651 0.315 Variant allele tumor 5 frequency (pmol product/ µ g HER2/min) 0 Xenograft 0.998 0.986 0.997 0.324 WT L866M

L866M HER2 863 DFGLARLLDIDETEYHA 879 EGFR 855 DFGLAKLLGAEEKEYHA 871 C S310Y PDX M122. HER2 mutation L858R L861Q 1,600

1,400 Placebo 1,200 Trastuzumab ) 3 1,000 Neratinib

800 Trastuzumab + Lapatinib 600 Lapatinib Tumor size (mm

400 Trastuzumab + Neratinib 200

0 −20 −10 0 10 20 30 40 50 Time (days)

DEG12D PDX M051. HER2L866M + amplification PDX M551. KRAS 2,200 2,000 Placebo 2,000 Placebo 1,800 Cetuximab 1,800 Trastuzumab 1,600 Neratinib

Neratinib ) ) 3 3 1,600 1,400 + Trastuzumab + Trastuzumab 1,400 Neratinib 1,200 Neratinib 1,200 1,000 1,000 800 800 600 Tumor size (mm Tumor size (mm 600 400 400 200 200 0 0 −20 −10 0 10 20 30 −20 −10 0 10 20 30 40 Time (days) Time (days)

Figure 4. Drug treatment of HER2- or KRAS-mutant colorectal cancer PDXs. A, HER2 gene–specifi c sequencing of 48 cetuximab-refractory, quadruple WT PDXs identifi ed 4 PDXs with HER2 mutations. Allele frequencies from next-generation DNA sequencing on the primary tumor prior to implantation or of the PDX grown in the mice are shown. B, in vitro kinase assay on WT or L866M HER2 kinase domain. HER2L866M is homologous to EGFRL858R . C–E, tumor growth curves for PDX tumors (n = 5 for each treatment arm). Data, mean ± SEM. Drug doses are: trastuzumab 30 mg/kg weekly, neratinib 40 mg/kg orally daily, lapatinib 100 mg/kg orally daily, and cetuximab 20 mg/kg twice weekly.

DISCUSSION resistance to the EGFR monoclonal antibodies cetuximab and panitumumab in colon cell lines. HER2-mutant transduced The TCGA project has identifi ed HER2 somatic mutations DiFi and NCI-H508 cells are inhibited by low nanomolar from colorectal cancer patients, but the clinical effect of these doses of the second-generation, irreversible tyrosine kinase mutations was unknown. Here, we show that these HER2 inhibitors neratinib and afatinib. Further, HER2 gene mutations activate intracellular signaling pathways, increase sequencing on 48 cetuximab-resistant, quadruple WT colo- anchorage-independent growth in soft agar, and produce rectal cancer PDXs identifi ed 4 PDXs with HER2 activating

838 | CANCER DISCOVERYAUGUST 2015 www.aacrjournals.org

Downloaded from cancerdiscovery.aacrjournals.org on September 26, 2021. © 2015 American Association for Cancer Research. HER2 Activating Mutations Are Targets in Colorectal Cancer RESEARCH BRIEF

mutations (4/48 = 8.3%). The effect of HER2-targeted thera- metastatic colorectal cancer for multiple genes is now practi- pies on two of these PDXs was tested. Single-agent HER2-tar- cal. KRAS-mutated samples show resistance to neratinib in geted therapy, with either trastuzumab, neratinib, or lapatinib, our experiments, and it would be prudent for current clinical delayed the growth of these PDXs, and dual HER2-targeted trials to focus on colorectal cancer patients whose tumor is therapy with either trastuzumab plus neratinib or trastuzu- KRASWT . NSABP Oncology Genome Assessment Guided Med- mab plus lapatinib produced durable tumor regression in the icine (N-GAMe) Program and the NSABP Colorectal Cancer mice. These results are consistent with recent analyses of the Biospecimen Profi ling Repository Trial (MPR-1 trial) include genomic landscape of response to EGFR therapy that identi- testing for HER2 mutations, and this will lead to prospective fi ed sequence alterations in HER2 as a mechanism of resist- clinical trials for colorectal cancer patients. ance to cetuximab in colorectal cancer (Bertotti, Papp, and colleagues, Nature, in press). More importantly, these data sug- gest that HER2 activating mutations may themselves be a drug METHODS target for the treatment of colorectal cancer. These preclinical Antibodies and Inhibitors fi ndings should be tested in colorectal cancer clinical trials. Antibodies for Western blots were purchased from Cell Signaling Potential caveats and limitations of this study include the Technologies: phosphoPLCγ (Tyr783), PLCγ, phosphoEGFR (Tyr1173), following. First, we acknowledge that retroviral transduction EGFR, phosphop44/42 MAPK (Thr202/Tyr204), p44/42 MAPK, phos- of HER2 into cell lines can produce overexpression of HER2. phoAKT (Ser473), and AKT; Millipore: phosphoHER2 (pY1248); and However, the colorectal cancer PDX samples contain endog- Thermo Fisher: HER2 antibody (Ab-17). Antibodies for IHC were enous HER2 mutations that are expressed at their native levels. purchased from Cell Signaling Technology (phosphoS6 Ser235/236, clone D57.2.2E; phosphoERK1/2 Thr202/Tyr204, clone D13.14.4E) or The growth suppression of the PDXs by HER2-targeted agents Dako (Ki-67, clone MIB-1). Cetuximab, panitumumab, trastuzumab, demonstrates that HER2 activating mutations are required for and lapatinib were obtained from the hospital pharmacy. Afatinib was the growth of these cancers. Second, next-generation genome obtained from Selleckchem. Neratinib was provided by Puma Biotech- sequencing has identifi ed large numbers of HER2 somatic muta- nology, Inc., under a Materials Transfer Agreement. tions, and it is not practical to experimentally test every muta- tion. Insights from structural biology and homology to known Cell Lines activating mutations in related genes as well as mutation impact IMCE and IMCE–KRAS cells were a generous gift from Dr. Robert prediction algorithms can generate hypotheses about the effect Whitehead (Vanderbilt University, Nashville). Dates of receipt were of novel mutations ( 6 , 27 , 28 ). Five mutations that were seen in August 13, 2013, and November 7, 2014, respectively, for these two only one patient with colorectal cancer (I263T, A466T, R678Q, cell lines, and they were cultured in RPMI-1640 supplemented with 5% R866W, and N1219S) were not tested in this study and should FCS, 1 μg/mL , 10 μmol/L a-thioglycerol, 1 μmol/L hydrocorti- currently be regarded as variants of unknown signifi cance. sone, 5 units per mL of mouse gamma interferon, and 1% penicillin/ ° There are many important implications of this study. HER2 streptomycin (P/S) in a 5% CO 2 humidifi ed atmosphere at 33 C. Cell line authentication of the IMCE cells (performed by Promega/ATCC somatic mutations are found in a wide range of solid tumors, on January 15, 2015) confi rmed that they were a nonhuman cell line. including 9% of bladder cancer cases, 7% of glioblastoma cases, DiFi cells were a gift from Dr. Alberto Bardelli (University of Torino, 5% of gastric cancer cases, 4% of lung adenocarcinoma cases, Italy; date of receipt May 15, 2013), and short tandem repeat profi ling 3% of esophageal cancer cases, and 1.5% to 2% of breast cancer performed by Promega/ATCC on January 15, 2015, showed that they cases ( 16 , 29 ). The broad distribution of HER2 somatic muta- had a D5S818 11,12; D13S317 8,11; D7S820 10,12; D16S539 12; vWA tions suggests that HER2 activating mutations are drivers in a 17,18; THO1 7, 9.3; AMEL X; TPOX 8,9; and CSF1PO 10,11 profi le. wide range of cancer types. The data presented here on color- NCI-H508 cells were purchased from the ATCC (date of receipt May 29, ectal cancer combined with prior functional studies on HER2 2014) and not further authenticated. The ATCC performs authentica- mutations in breast cancer and NSCLC support this hypoth- tion on its own cell lines, and the NCI-H508 cells were used for fewer esis ( 6, 7 ). Further, a multi-institutional, phase II is than 6 months after receipt and resuscitation from cryopreservation. SW480 and HCT116 cells were obtained from Drs. Jieya Shao and currently evaluating neratinib therapy in patients with a broad David Piwnica-Worms (Washington University School of Medicine, spectrum of solid tumors that harbor HER2 mutations ( 30 ). St. Louis, MO; date of receipt May 2014), and no authentication was These data form a strong preclinical rationale for clinical performed on these cells. DiFi and NCI-H508 cells were maintained in a o trials targeting HER2 activating mutations in patients with 5% CO 2 humidifi ed atmosphere at 37 C, and culture media for these two metastatic colorectal cancer. While this article was under cell lines are as follows: DiFi cells: F12 medium supplemented with 5% review, Kloth and colleagues published a study indicating that FCS, 1% P/S; NCI-H508 cells: RPMI-1640 supplemented with 5% FCS, 15% of Lynch syndrome or Lynch-like colorectal cancers have 1% P/S. Inhibition of cell growth by cetuximab, panitumumab, nerat- HER2 mutations, and they showed that the HER2-mutant inib, and afatinib was measured by Alamar blue or crystal violet assay colorectal cancer cell lines CW-2 and CCK-81 are sensitive to ( 35 ). IC50 values were calculated by a 4-parameter nonlinear regression treatment with neratinib and afatinib (31 ). Our PDX results conducted using SigmaPlot version 11 software (Systat software, Inc). suggest that dual HER2-targeted therapy may be needed Retroviral Transduction of HER2 Mutants in Colorectal to achieve optimum antitumor effect. Several large clinical Cancer Cell Lines trials on HER2-amplifi ed breast cancer have demonstrated HER2 WT or mutant retroviral vectors that we published were improved patient outcomes with dual HER2-targeted therapy transfected in ØNX amphotropic packaging cell line. HER2 WT or (32, 33). Metastatic colorectal cancers are routinely tested for mutant recombinant retroviral supernatants were transduced in KRAS, NRAS, and BRAF mutations ( 34 ). Our fi ndings suggest IMCE, DiFi, and NCI-H508 cell lines as described previously ( 6 ). that HER2 gene sequencing should also be included in this After 2 to 3 weeks of zeocin selection of bulk infected cultures, trans- testing. With the growing availability of gene panels, testing gene expression was verifi ed by FACS analysis for GFP expression

AUGUST 2015CANCER DISCOVERY | 839

Downloaded from cancerdiscovery.aacrjournals.org on September 26, 2021. © 2015 American Association for Cancer Research. RESEARCH BRIEF Kavuri et al.

(always >90%). Western blot analysis was performed on polyclonal approved by the Ethical Commission of the Candiolo Cancer Insti- cell lines to confi rm HER2 expression. tute and by the Italian Ministry of Health.

Soft-Agar Colony-Forming Assay Disclosure of Potential Confl icts of Interest Six-well plates were fi rst layered with 0.6% bacto agar in IMCE No potential confl icts of interest were disclosed. growth medium. After the solidifi cation of the bottom layer, a top layer containing 5 × 103 to 10 × 10 3 IMCE HER2 WT or mutant cells Authors’ Contributions in IMCE growth medium plus 0.4% bacto agar was added. Assays were Conception and design: S.M. Kavuri, N. Jain, A.C. Searleman, carried out in duplicate. Cells were allowed to form colonies for 12 days S.A. Jacobs, A. Bertotti, R. Bose and were photographed and quantifi ed as shown previously ( 6 ). Development of methodology: S.M. Kavuri, N. Jain, A.C. Searleman Statistical analysis of the colony count data was modeled using Acquisition of data (provided animals, acquired and managed Poisson regression using the MCMCglmm package (version 2.16; ref. patients, provided facilities, etc.): S.M. Kavuri, N. Jain, F. Galimi, 36 ), of the R statistical environment (version 2.15.1) using “Genotype” F. Cottino, S.M. Leto, G. Migliardi, W. Shen, J. Monsey, L. Trusolino and a “Genotype:Treatment” interaction as fi xed predictors. No inter- Analysis and interpretation of data (e.g., statistical analysis, biosta- cept was included to force explicit measurements for each “Genotype.” tistics, computational analysis): S.M. Kavuri, N. Jain, S.M. Leto, MCMCglmm uses fully Bayesian modeling and parameter estimates A.C. Searleman, J. Monsey, L. Trusolino, A. Bertotti, R. Bose generated using Gibbs sampling Markov Chain Monte Carlo with Writing, review, and/or revision of the manuscript: S.M. Kavuri, 100,000 iterations, a burn-in of 3,000, and a thin of 10. Diagnostics N. Jain, A.C. Searleman, L. Trusolino, S.A. Jacobs, A. Bertotti, R. Bose revealed the lack of autocorrelation and excellent chain mixing. The Administrative, technical, or material support (i.e., reporting or default prior was used for fi xed effects, which is a multivariate normal organizing data, constructing databases): W. Shen distribution with a 0 mean vector and diagonal variance matrix with Study supervision: A. Bertotti, R. Bose variances of 1010 and covariances of 0, as this ensures that fi xed effects are independent and estimated almost entirely from the data. Over- Acknowledgments dispersion and replicates were accounted for in the residual variance The authors thank Runjun Kumar for assistance with Fig. 1A and structure with an improper inverse Wishart prior with nu = 0 and V = B and Francesco Sassi for IHC analysis . 1, which implicitly assumes each well is a random effect. Parameters and parameter contrasts were considered to be statistically signifi cant when the 95% highest posterior density interval did not contain 0. The Grant Support conclusions were robust to changes in the minimal colony size from 1 This work was supported by the NIH (R01CA161001, to R. Bose); to 10 (the global median colony size). the Ohana Breast Cancer Research Fund and the Foundation for Barnes-Jewish Hospital (to R. Bose); the Fight Colorectal Cancer-AACR HER2 Gene Sequencing and In Vitro Kinase Assay Career Development Award (to A. Bertotti); the Associazione Italiana Initial screening of PDX samples for HER2 mutations was con- per la Ricerca sul Cancro (AIRC) 2010 Special Program Molecular × ducted by Sanger sequencing. Genomic DNA was extracted with the Clinical Oncology 5 1000, project 9970 (to L. Trusolino); AIRC Inves- Wizard Purifi cation System (Promega). Primers for HER2 exons 8 and tigator Grants, project 14205 (to L. Trusolino); AIRC Investigator 18–24 were designed with Primer3 software and synthesized by Sigma. Grants, project 15571 (to A. Bertotti); and Fondazione Piemontese per × Purifi ed PCR products were sequenced with a BigDye Terminator la Ricerca sul Cancro-ONLUS, 5 1000 Ministero della Salute 2011 (to version 3.1 Cycle Sequencing (Applied Biosystems) and analyzed L. Trusolino). with a 3730 ABI capillary electrophoresis system. Confi rmation of HER2 mutations and determination of allele frequency were performed Received October 13, 2014; revised May 27, 2015; accepted June by next-generation sequencing using an Illumina MiSeq instrument. 01, 2015; published online August 4, 2015. Briefl y, all ERBB2 exons were PCR amplifi ed in triplicate (75 ng DNA per reaction) on a BioMark HD system (Fluidigm). All samples were pooled and cleaned using bead purifi cation. The samples were loaded REFERENCES on an Illumina MiSeq instrument and sequenced. Total read counts at 1. Kandoth C , McLellan MD , Vandin F , Ye K , Niu B , Lu C , et al. the nucleotide position of the identifi ed HER2 mutations ranged from Mutational landscape and signifi cance across 12 major cancer types. 13,600 to 65,100 reads. The HER2 tyrosine kinase domain was recom- Nature 2013 ; 502 : 333 – 9 . binantly expressed in Sf9 cells using a baculoviral vector and purifi ed to 2. Weinstein JN , Collisson EA , Mills GB , Shaw KR , Ozenberger BA , Ell- >80% purity, as previously described (6 , 37 ). In vitro kinase assays were rott K , et al. The Cancer Genome Atlas Pan-Cancer analysis project. performed using γ-32 P-ATP and a synthetic peptide substrate ( 37 ). Nat Genet 2013 ; 45 : 1113 – 20 . 3. The Cancer Genome Atlas Consortium. Comprehensive molecular char- Xenograft Models and In Vivo Treatments acterization of human colon and rectal cancer. Nature 2012 ; 487 : 330 – 7 . 4. Bertotti A , Migliardi G , Galimi F , Sassi F , Torti D , Isella C , et al. A Tumor implantation and expansion were performed as previ- molecularly annotated platform of patient-derived xenografts (“xen- ously described ( 4 ). Established tumors (average volume, 400–600 opatients”) identifi es HER2 as an effective therapeutic target in 3 mm ) were treated with the following regimens, either single-agent cetuximab-resistant colorectal cancer. Cancer Discov 2011 ; 1 : 508 – 23 . or in combination: trastuzumab (Roche) 30 mg/kg weekly (vehicle: 5. Yonesaka K , Zejnullahu K , Okamoto I , Satoh T , Cappuzzo F , Souglakos physiologic saline); neratinib (Puma Biotechnology) 40 mg/kg orally J , et al. Activation of ERBB2 signaling causes resistance to the EGFR- daily (vehicle, 0.5% methylcellulose, 0.4% Tween-80). Tumor size was directed therapeutic antibody cetuximab . Sci Transl Med 2011 ; 3 : 99ra86 . evaluated once weekly by caliper measurements, and the volume of 6. Bose R , Kavuri SM , Searleman AC , Shen W , Shen D , Koboldt DC , the mass was calculated using the formula 4/3 × π × (d/2)2 × (D/2), et al. Activating HER2 mutations in HER2 gene amplifi cation nega- where d is the minor tumor axis and D is the major tumor axis. All tive breast cancer. Cancer Discov 2013 ; 3 : 224 – 37 . values for tumor growth curves were recorded blindly. In vivo proce- 7. Greulich H , Kaplan B , Mertins P , Chen TH , Tanaka KE , Yun CH, dures and related biobanking data were managed using the Labora- et al. Functional analysis of mutations in tory Assistant Suite (LAS), a web-based proprietary data management lung cancer identifi es oncogenic extracellular domain mutations of system for automated data tracking ( 38 ). Animal procedures were ERBB2. Proc Natl Acad Sci U S A 2012 ; 109 : 14476 – 81 .

840 | CANCER DISCOVERYAUGUST 2015 www.aacrjournals.org

Downloaded from cancerdiscovery.aacrjournals.org on September 26, 2021. © 2015 American Association for Cancer Research. HER2 Activating Mutations Are Targets in Colorectal Cancer RESEARCH BRIEF

8. Wang SE , Narasanna A , Perez-Torres M , Xiang B , Wu FY , 23. Ahmed D , Eide PW , Eilertsen IA , Danielsen SA , Eknaes M , Hektoen Yang S , et al. HER2 kinase domain mutation results in constitutive M , et al. Epigenetic and genetic features of 24 colon cancer cell lines . phosphorylation and activation of HER2 and EGFR and resistance to Oncogenesis 2013 ; 2 : e71 . EGFR tyrosine kinase inhibitors. Cancer Cell 2006 ; 10 : 25 – 38 . 24. Moretti S , De Falco V , Tamburrino A , Barbi F , Tavano M , Avenia 9. ClinicalTrials.gov [homepage on the Internet]. Neratinib in meta- N , et al. Insights into the molecular function of the inactivating static HER2 non-amplifi ed but HER2 mutant breast cancer. Available mutations of B-Raf involving the DFG motif . Biochim Biophys Acta from: http://www.clinicaltrials.gov/ct2/show/NCT01670877 . Access 2009 ; 1793 : 1634 – 45 . date Sept. 28, 2014. 25. Bardelli A , Corso S , Bertotti A , Hobor S , Valtorta E , Siravegna G , et al. 10. ClinicalTrials.gov [homepage on the Internet]. An open-label, phase 2 Amplifi cation of the MET receptor drives resistance to anti-EGFR study of neratinib in patients with solid tumors with somatic human therapies in colorectal cancer. Cancer Discov 2013 ; 3 : 658 – 73 . epidermal receptor (EGFR, HER2, HER3) mutations 26. Sliwkowski MX , Lofgren JA , Lewis GD , Hotaling TE , Fendly BM , Fox or EGFR gene amplifi cation. Available from: http://clinicaltrials.gov/ JA . Nonclinical studies addressing the mechanism of action of trastu- ct2/show/NCT01953926 . Access date Oct. 2, 2014. zumab (Herceptin). Semin Oncol 1999 ; 26 : 60 – 70 . 11. Gandhi L , Bahleda R , Tolaney SM , Kwak EL , Cleary JM , Pandya SS , 27. Reva B , Antipin Y , Sander C . Predicting the functional impact of et al. Phase I study of neratinib in combination with temsirolimus in protein mutations: application to cancer genomics. Nucleic Acids Res patients with human epidermal 2-dependent 2011 ; 39 : e118 . and other solid tumors. J Clin Oncol 2014 ; 32 : 68 – 75 . 28. Sim NL , Kumar P , Hu J , Henikoff S , Schneider G , Ng PC . SIFT web 12. Besse B , Soria J-C , Yao B , Kris M , Chao B , Cortot A , et al. Neratinib server: predicting effects of amino acid substitutions on proteins . (N) with or without temsirolimus (TEM) in patients (pts) with non- Nucleic Acids Res 2012 ; 40 : W452 – 7 . small cell lung cancer (NSCLC) carrying HER2 somatic mutations: 29. The Cancer Genome Atlas cBio Portal search performed August 30 , An international randomized phase II study . Annals of Oncology 2104 . Available from: http://www.cbioportal.org/public-portal/ . 2014 ; 25 : v1 – v41 ; abstract LBA39 PR. 30. ClinicalTrials.gov [homepage on the Internet]. An Open-label, Phase 2 13. Chong CR , Janne PA . The quest to overcome resistance to EGFR- Study of Neratinib in Patients With Solid Tumors With Somatic Human targeted therapies in cancer. Nat Med 2013 ; 19 : 1389 – 400 . Receptor (EGFR, HER2, HER3) Mutations or 14. Brannon AR , Vakiani E , Sylvester BE , Scott SN , McDermott G , Shah EGFR Gene Amplifi cation . Available from: http://clinicaltrials.gov/ct2/ RH , et al. Comparative sequencing analysis reveals high genomic con- show/NCT01953926 . Access date May 15, 2015. cordance between matched primary and metastatic colorectal cancer 31. Kloth M , Ruesseler V , Engel C , Koenig K , Peifer M , Mariotti E , et al. lesions. Genome Biol 2014 ; 15 : 454 . Activating ERBB2/HER2 mutations indicate susceptibility to pan- 15. Paez JG , Janne PA , Lee JC , Tracy S , Greulich H , Gabriel S , et al. EGFR HER inhibitors in Lynch and Lynch-like colorectal cancer . Gut 2015 mutations in lung cancer: correlation with clinical response to gefi t- Apr 28. [Epub ahead of print] . inib therapy. Science 2004 ; 304 : 1497 – 500 . 32. Baselga J , Cortes J , Kim SB , Im SA , Hegg R , Im YH , et al. 16. Cerami E , Gao J , Dogrusoz U , Gross BE , Sumer SO , Aksoy BA, e t a l . plus trastuzumab plus docetaxel for metastatic breast cancer . N Engl The cBio cancer genomics portal: an open platform for exploring J Med 2012 ; 366 : 109 – 19 . multidimensional cancer genomics data . Cancer Discov 2012 ; 2 : 401 – 4 . 33. Blackwell KL , Burstein HJ , Storniolo AM , Rugo H , Sledge G , 17. Forbes SA , Bindal N , Bamford S , Cole C , Kok CY , Beare D , et al. COS- Koehler M , et al. Randomized study of Lapatinib alone or in MIC: mining complete cancer genomes in the Catalogue of Somatic combination with trastuzumab in women with ErbB2-positive, Mutations in Cancer. Nucleic Acids Res 2011 ; 39 : D945 – 50 . trastuzumab-refractory metastatic breast cancer. J Clin Oncol 18. Guastadisegni C , Colafranceschi M , Ottini L , Dogliotti E . Microsatellite 2010 ; 28 : 1124 – 30 . instability as a marker of prognosis and response to therapy: a meta- 34. Karapetis CS , Khambata-Ford S , Jonker DJ , O’Callaghan CJ , Tu D , analysis of colorectal cancer survival data . Eur J Cancer 2010 ; 46 : 2788 – 98 . Tebbutt NC , et al. K-ras mutations and benefi t from cetuximab in 19. Sargent DJ , Marsoni S , Monges G , Thibodeau SN , Labianca R , Ham- advanced colorectal cancer. N Engl J Med 2008 ; 359 : 1757 – 65 . ilton SR , et al. Defective mismatch repair as a predictive marker for 35. Kavuri SM , Geserick P , Berg D , Dimitrova DP , Feoktistova M , S ieg- lack of effi cacy of fl uorouracil-based adjuvant therapy in colon can- mund D , et al. Cellular FLICE-inhibitory Protein (cFLIP) isoforms cer. J Clin Oncol 2010 ; 28 : 3219 – 26 . block CD95- and TRAIL death receptor-induced gene induction 20. D’Abaco GM , Whitehead RH , Burgess AW . Synergy between Apc min irrespective of processing of caspase-8 or cFLIP in the death-inducing and an activated ras mutation is suffi cient to induce colon carcino- signaling complex. J Biol Chem 2011 ; 286 : 16631 – 46 . mas. Mol Cell Biol 1996 ; 16 : 884 – 91 . 36. Hadfi eld JD . MCMC methods for multi-response generalized linear 21. Whitehead RH , VanEeden PE , Noble MD , Ataliotis P , Jat PS . Estab - mixed models: the MCMCglmm R package. J Stat Software 2010 ; 33 : lishment of conditionally immortalized epithelial cell lines from both 1– 22 . colon and small intestine of adult H-2Kb-tsA58 transgenic mice . Proc 37. Monsey J , Shen W , Schlesinger P , Bose R . Her4 and Her2/neu tyrosine Natl Acad Sci U S A 1993 ; 90 : 587 – 91 . kinase domains dimerize and activate in a reconstituted in vitro sys- 22. Misale S , Yaeger R , Hobor S , Scala E , Janakiraman M , Liska D , et al. tem. J Biol Chem 2010 ; 285 : 7035 – 44 . Emergence of KRAS mutations and acquired resistance to anti-EGFR 38. Baralis E , Bertotti A , Fiori A , Grand A . LAS: a software platform to therapy in colorectal cancer. Nature 2012 ; 486 : 532 – 6 . support oncological data management. J Med Syst 2012 ; 36 : S81 – 90 .

AUGUST 2015CANCER DISCOVERY | 841

Downloaded from cancerdiscovery.aacrjournals.org on September 26, 2021. © 2015 American Association for Cancer Research. HER2 Activating Mutations Are Targets for Colorectal Cancer Treatment

Shyam M. Kavuri, Naveen Jain, Francesco Galimi, et al.

Cancer Discovery 2015;5:832-841.

Updated version Access the most recent version of this article at: http://cancerdiscovery.aacrjournals.org/content/5/8/832

Supplementary Access the most recent supplemental material at: Material http://cancerdiscovery.aacrjournals.org/content/suppl/2015/07/25/5.8.832.DC1

Cited articles This article cites 33 articles, 14 of which you can access for free at: http://cancerdiscovery.aacrjournals.org/content/5/8/832.full#ref-list-1

Citing articles This article has been cited by 32 HighWire-hosted articles. Access the articles at: http://cancerdiscovery.aacrjournals.org/content/5/8/832.full#related-urls

E-mail alerts Sign up to receive free email-alerts related to this article or journal.

Reprints and To order reprints of this article or to subscribe to the journal, contact the AACR Publications Department at Subscriptions [email protected].

Permissions To request permission to re-use all or part of this article, use this link http://cancerdiscovery.aacrjournals.org/content/5/8/832. Click on "Request Permissions" which will take you to the Copyright Clearance Center's (CCC) Rightslink site.

Downloaded from cancerdiscovery.aacrjournals.org on September 26, 2021. © 2015 American Association for Cancer Research.