E XP ER I ME NTAL C E LL RE S E ARCH

34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60 61 62 63 64 65 66 67 68 69 70 71 72 73 74 75 76 77 78 79 80 81 82 83 84 85 86 87 88 89 90 91 92 93

] (]]]]) ]]]–]]]

Available online at www.sciencedirect.com

journal homepage: www.elsevier.com/locate/yexcr

Research Article

A novel PI3K inhibitor iMDK suppresses non-small cell lung Cancer cooperatively with A MEK inhibitor

Q1

Naomasa Ishidaa, Takuya Fukazawaa,n, Yutaka Maedad, Tomoki Yamatsujia, Munenori Takaokaa, Minoru Haisaa, Etsuko Yokotaa, Kaori Shigemitsua, Ichiro Moritaa, Katsuya Katoc, Kenichi Matsumotoe, Tsuyoshi Shimoe, Tatsuo Okuie,f, Xiao-Hong Baog, Huifang Haoh, Shawn N. Grantd, Nagio Takigawab, Jeffrey A. Whitsettd, Yoshio Naomotoa a

Department of General Surgery, Kawasaki Medical School, Okayama 700-8505, Japan Department of General Internal Medicine 4, Kawasaki Medical School, Okayama 700-8505, Japan c Department of Diagnostic Radiology 2, Kawasaki Medical School, Okayama 700-8505, Japan Q2 dDivision of Pulmonary Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229-3039 United States e Department of Oral and Maxillofacial Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan f Division of Hematology and Oncology, Indiana University School of Medicine, Indianapolis, IN 46202, United States g Department of Biochemistry, School of Basic Medicine, Liaoning Medical University, Jinzhou 121001, China h Department of Biology, College of Life Science, Inner Mongolia University, Hohhot 010021, China b

article information

abstract

Article Chronology:

The PI3K–AKT pathway is expected to be a therapeutic target for non-small cell lung cancer

Received 5 October 2014

(NSCLC) treatment. We previously reported that a novel PI3K inhibitor iMDK suppressed NSCLC

Received in revised form

cells in vitro and in vivo without harming normal cells and mice. Unexpectedly, iMDK activated

4 March 2015

the MAPK pathway, including ERK, in the NSCLC cells. Since iMDK did not eradicate such NSCLC

Accepted 23 March 2015

cells completely, it is possible that the activated MAPK pathway confers resistance to the NSCLC cells against cell death induced by iMDK. In the present study, we assessed whether suppressing

Keywords:

of iMDK-mediated activation of the MAPK pathway would enhance anti-tumorigenic activity of

PI3K

iMDK. PD0325901, a MAPK inhibitor, suppressed the MAPK pathway induced by iMDK and

MEK

cooperatively inhibited cell viability and colony formation of NSCLC cells by inducing apoptosis

NSCLC

in vitro. HUVEC tube formation, representing angiogenic processes in vitro, was also cooperatively inhibited by the combinatorial treatment of iMDK and PD0325901. The combinatorial treatment of iMDK with PD0325901 cooperatively suppressed tumor growth and tumor-associated angiogenesis in a lung cancer xenograft model in vivo. Here, we demonstrate a novel treatment strategy using iMDK and PD0325901 to eradicate NSCLC. & 2015 Published by Elsevier Inc.

n

Corresponding author. Fax: þ81 86 232 8343. E-mail address: [email protected] (T. Fukazawa).

http://dx.doi.org/10.1016/j.yexcr.2015.03.019 0014-4827/& 2015 Published by Elsevier Inc.

Please cite this article as: N. Ishida, et al., A novel PI3K inhibitor iMDK suppresses non-small cell lung Cancer cooperatively with A MEK inhibitor, Exp Cell Res (2015), http://dx.doi.org/10.1016/j.yexcr.2015.03.019

94 95 96 97 98 99 100

2

101 102 103 104 105 106 107 108 109 110 111 112 113 114 115 116 117 118 119 120 121 122 123 124 125 126 127 128 129 130 131 132 133 134 135 136 137 138 139 140 141 142 143 144 145 146 147 148 149 150 151 152 153 154 155 156 157 158 159 160

E XP E RI ME N TAL CE L L R ES E ARC H

Introduction Lung cancer is the leading cause of cancer-related mortality worldwide [1]. Recent discoveries of molecularly targeted therapies, including erlotinib/gefitinib for EGFR-mutant NSCLC [2,3] and crizotinib/ceritinib for ALK-fusion NSCLC [4,5] have demonstrated that such therapies using small molecule drugs extend survival of NSCLC patients significantly, compared to conventional chemotherapy [6,7]. In addition to EGFR-mutant or ALK-fusion NSCLCs, projection for RET-fusion and ROS1-fusion NSCLCs mortality are also considered to be favorable since they can be targeted by small molecules, such as crizotinib [7,8]. A drawback of current therapy for NSCLC is that there is no effective treatment for KRAS-mutant NSCLC; the most common type in Caucasians and the second most common type in Asians [9], and squamous NSCLC [10]. Molecularly targeted therapies for KRAS-mutant NSCLC have been tested [11,12]. For example, inhibition of MEK (a MAPKK; downstream of KRAS) by selumetinib, a MEK inhibitor, in addition to docetaxel (a chemotherapeutic drug) extended survival of KRAS-mutant NSCLC patients by 4 months compared to docetaxel alone. However, selumetinib plus docetaxel caused more adverse events than docetaxel alone [13], indicating that further improvements in this therapeutic strategy are needed to extend survival and reduce the adverse events associated with KRAS-mutant NSCLC. In addition to the KRAS–MAPK pathway, the KRAS–PI3K–AKT pathway is considered to contribute to KRAS lung tumorigenesis [14]. While not mutually exclusive, these pathways often interact with each other [15]. For example, a PI3K inhibitor PI-103 inhibited the PI3K pathway while activating the MAPK pathway [16], suggesting a potential mechanism for tumor survival by the compensatory activation of an alternative tumorigenic pathway. We recently reported a novel PI3K inhibitor iMDK, which was originally identified as a small molecule inhibiting the growth factor MDK (also known as midkine or MK) [17]. While its direct targets are unknown, iMDK inhibited phosphorylation of PI3K and AKT, indicating that iMDK serves as a PI3K inhibitor [17]. Similarly to PI-103 [16], iMDK activated phosphorylation of ERK and P38MAPK while inhibiting the PI3K pathway [17]. Since the combined inhibition of the PI3K and MAPK pathways by PI-103 and a MAPK inhibitor PD0325901 enhanced cell killing of NSCLC [16], in the present study, we tested whether the novel PI3K inhibitor iMDK further enhanced cell death of NSCLC in the presence of PD0325901. Combined treatment of iMDK and PD0325901 significantly suppressed tumor growth of NSCLC, including KRAS-mutant NSCLC and squamous NSCLC. The combinatorial treatment also inhibited endothelial cell-mediated angiogenesis, a requirement for tumor progression in vitro and in vivo. Here, we demonstrate a novel strategy to treat NSCLC by simultaneously targeting both PI3K and MAPK pathways by iMDK and PD0325901.

Materials and methods Reagents 3-[2-(4-fluorobenzyl)imidazo[2,1–b][1,3]thiazol-6-yl]-2H-chromen-2-one (here after termed iMDK) purchased from ChemDiv

] (]]]]) ]]]–]]]

(San Diego, CA) and MEK inhibitor PD0325901 obtained from Selleck Chemicals (Houston, TX) were dissolved in DMSO.

Cell lines and culture conditions H441 (lung adenocarcinoma; KRASG12V), H2009 (non-small cell carcinoma; KRASG12A), A549 (lung carcinoma; KRASG12S) and H520 (lung squamous cell carcinoma; KRASWT) were obtained from the American Type Culture Collection (Manassas, VA) and grown in high glucose Dulbecco's modified Eagle medium (H441 and A549) or RPMI 1640 (H2009 and H520) supplemented with 10% heatinactivated fetal bovine serum. Cell information was obtained from COSMIC (http://cancer.sanger.ac.uk/cancergenome/projects/ cosmic/). Human umbilical vein endothelial cells (HUVECs) pur chased from Life Technologies (Grand Island, NY) were grown in HUVEC growth medium (MCDB-104 medium supplemented with 10 ng/ml EGF, 100 μg/ml heparin, 100 ng/ml endothelial cell growth factor and 10% heat-inactivated fetal bovine serum). All cell lines were cultured in 10% CO2 at 37 1C.

Immunoblot analysis Cells were lysed in ice-cold M-PER lysis buffer purchased from Thermo Fisher Scientific (Rockford, IL). Cell lysates were clarified by centrifugation (20 min at 15,000g at 4 1C) and protein concentration determined using the BCA protein assay (Thermo Fisher Scientific). Equal amounts of protein were separated on an SDS-PAGE gel. The gel was electrophoretically transferred to a Hybond PVDF transfer membrane (GE. Healthcare Ltd., Piscataway, NJ) and incubated with primary and secondary antibodies according to the Supersignals West Pico chemiluminescence protocol (Pierce, Rockford, IL). Antibody specific for β-actin antibody was obtained from Sigma (St. Louis, MO). Antibody specific for AKT, phosphorylated-AKT (Ser473), ERK, and phosphorylated-ERK (Ser473) were obtained from Cell Signaling Technology (Beverly, MA). Secondary horseradish peroxidaseconjugated antibodies were obtained from Jackson Immunoresearch Laboratories (West Grove, PA).

Cell viability assay H441 cells and H2009 cells were plated in 96-well plates at a density of 1.5  103 cells and cultured at 37 1C for 24 h. H441 and H2009 cells were treated with or without iMDK (2.5 μM) in the presence or absence of PD0325901 (0.5 μM). H520 cells were treated with or without iMDK (0.125 μM) in the presence or absence of PD0325901 (0.25 μM). A549 cells were treated with or without iMDK (0.25 μM) in the presence or absence of PD0325901 (0.125 μM). Cells were treated for 72 h. Viable cells were assessed by WST-1 assays (Roche Molecular Biochemicals, Laval, Quebec, Canada) according to the manufacturer's protocol.

Colony formation assay Cells were first plated at a density of 5  104 cells for H441 and 1  105 cells for H2009 cells per well in 12-well plates 24 h before treatment. H441 cells were treated with iMDK at a concentration of 1 μM and/or PD0325901 at a concentration of 500 nM for 24 h and then released from the dish by incubation with trypsin/EDTA, counted, plated in triplicate at a density of 5  103 cells in six-well plates for 14 days. H2009 cells were treated with iMDK and/or PD0325901 at a

Please cite this article as: N. Ishida, et al., A novel PI3K inhibitor iMDK suppresses non-small cell lung Cancer cooperatively with A MEK inhibitor, Exp Cell Res (2015), http://dx.doi.org/10.1016/j.yexcr.2015.03.019

161 162 163 164 165 166 167 168 169 170 171 172 173 174 175 176 177 178 179 180 181 182 183 184 185 186 187 188 189 190 191 192 193 194 195 196 197 198 199 200 201 202 203 204 205 206 207 208 209 210 211 212 213 214 215 216 217 218 219 220

E X PE R IM EN TA L C ELL R E S EA RC H

221 222 223 224 225 226 227 228 229 230 231 232 233 234 235 236 237 238 239 240 241 242 243 244 245 246 247 248 249 250 251 252 253 254 255 256 257 258 259 260 261 262 263 264 265 266 267 268 269 270 271 272 273 274 275 276 277 278 279 280

concentration of 1 μM for 24 h and then released from the dish by incubation with trypsin/EDTA, counted, plated in triplicate at a density of 1  103 cells in six-well plates for 14 days. The cells were fixed with 100% methanol and allowed to air dry. The cells were then stained with 0.1% crystal violet. Colonies (a group of aggregated cells numbering at least 50) were then counted. The mean number of the control group was arbitrarily set to 100%, and all other numbers were normalized and percentage-specific cytotoxicity compared to colony formation in the control group was calculated.

Flow cytometric analysis for activated caspase-3 H441 cells were plated in 12-well plates at a density of 1  105 cells per well 1 day before the treatments. Cells were treated with iMDK (10 nM) or PD0325901 (10 nM) alone, or iMDK (10 nM) in combination with PD0325901 (10 nM). After 72 h, cells were harvested and washed twice with PBS. Cleaved caspase 3 was labeled in situ with phycoerythrin (PE)–conjugated anti-activated caspase-3 antibodies purchased from BD Bioscience (San Jose, CAs) and analyzed by FACS Verse (BD Bioscience)[18,19].

TUNEL staining Terminal deoxynucleotidyltransferase-mediated dUTP-biotin nick end labeling (TUNEL) staining was performed to detect apoptosis using the DeadEnd colorimetric TUNEL system (Promega, Madison, WI) according to the manufacturer's protocol.

] (]]]]) ]]]–]]]

3

immersion steps. After being autoclaved in 0.2% citrate buffer for 15 min, sections were incubated with 3% hydrogen peroxide for 30 min to block endogenous peroxidase activity. The antibodies for CD31/PECAM-1 were obtained from Abcam. Specimens were incubated overnight at 4 1C with a 1:100 dilution of antibody followed by three washes with TBS. The slides were treated with streptavidin-biotin complex (Envision System labeled polymer, horseradish peroxidase [HRP], Dako, Carpinteria, CA) for 60 min at a dilution of 1:100. Immunoreactions were visualized using a 3,39-diaminobenzidine (DAB) substrate-chromogen solution (Dako Cytomation LiquidDAB Substrate Chromogen System, Dako) and counterstained with hematoxylin. Sections were immersed in an ethanol and xylene bath and mounted for examination.

Ras activation assay H441 cells were treated with iMDK (250 nM and 500 nM) or PD0325901 (50 nM and 250 nM) for 6 h and 24 h. Cells were rinsed in PBS before cell lysis. Ras activation was determined using a RasGTPase Chemi ELISA kit, according to manufacturer's protocol (Active Motif, Carlsbad, CA).

Statistical analysis Significance of group differences was evaluated using ANOVA for multiple comparisons. Statistical significance was defined as po0.05 (#).

HUVEC tube formation assay HUVECs were treated with iMDK (10 μM) and/or PD0325901 (10 μM) for 5 h and a tube formation assay was performed using an in vitro angiogenesis kit (Millipore Corp. Bedford, MA). Tube formation was quantified by measuring the total length of the tube network and the number of branching points using Image J software (NIH, Bethesda) [20,21].

Mouse experiments The experimental protocol was approved by the Ethics Review Committee for Animal Experimentation of Okayama University Graduate School of Medicine and Dentistry (Ethics Committee reference number: OKU-2013104). Human lung cancer xenografts were established in 6-wk-old female BALB/c nude mice (Charles River Laboratories Japan, Kanagawa, Japan) by subcutaneous (s.c.) inoculation of H441 cells (1  106/ 50 μl) into the right dorsal flank. The mice were randomly assigned into four groups (n ¼8 per group) 9 days after tumor inoculations. The mice were intraperitoneally injected with 100 μl solution containing iMDK (9 mg/kg) everyday and/or orally administered PD0325901 (5 mg/kg) in 0.5% [w/v] methylcellulose solution with 0.2% [v/v] polysorbate 80 [Tween 80] five times per week (on days 1, 2, 3, 4, 5, 7, 8, 9, 10, 11). DMSO was intraperitoneally injected and methylcellulose solution with polysorbate 80 was orally administered in the control group. Tumors were measured every day, and tumor volume was calculated as a  b2  0.5, where a and b were large and small diameters, respectively. On day 11, all mice were sacrificed and tumors were removed and prepared for histology. For immunohistochemistry, sections were sequentially deparaffinized through a series of xylene, graded ethanol, and water

Results PD0325901 inhibited activation of ERK induced by iMDK treatment in H441 lung adenocarcinoma cells In the previous study, we demonstrated that iMDK suppressed the PI3K–AKT pathway (12 h after treatment) while inducing the MAPK pathway (48 h after treatment), as detected by the phosphorylation of ERK (MAPK), in H441 lung adenocarcinoma cells that carry a KRAS mutation (KRASG12V) [22]. Phosphorylated ERK (p-ERK) was slightly increased less than 1 h after treatment (Supplemental Fig. 1); however, p-ERK was robustly increased 72 h after treatment (Fig. 1A), suggesting that the effect of iMDK on the MAPK pathway in a short time is limited. RAS-GTP levels were not affected by iMDK (less than 24 h; Supplemental Fig. 2). These results suggest that iMDK primarily targets the PI3K pathway but not KRAS itself, which in turn influences alternative tumorigenic pathways. Induction of an alternative tumorigenic pathway after suppression of a tumorigenic pathway by therapeutic cancer drugs is considered to be a mechanism by which tumors evade cell death from the molecularly targeted drugs. Thus, a combinatorial treatment using drugs targeting multiple pathways may be required to block tumor cell resistance [15,23– 25]. In order to test this concept, we assessed whether the MAPK pathway activated by iMDK is suppressed by PD0325901, a MEK (MAPKK) inhibitor, in H441 cells. As shown in Fig. 1B, PD0325901 suppressed iMDK-induced phosphorylation of ERK in a dosedependent fashion. These results indicate that the combinatorial treatment of iMDK with PD0325901 simultaneously inhibits two major KRAS tumorigenic pathways in H441 cells.

Please cite this article as: N. Ishida, et al., A novel PI3K inhibitor iMDK suppresses non-small cell lung Cancer cooperatively with A MEK inhibitor, Exp Cell Res (2015), http://dx.doi.org/10.1016/j.yexcr.2015.03.019

281 282 283 284 285 286 287 288 289 290 291 292 293 294 295 296 297 298 299 300 301 302 303 304 305 306 307 308 309 310 311 312 313 314 315 316 317 318 319 320 321 322 323 324 325 326 327 328 329 330 331 332 333 334 335 336 337 338 339 340

4

341 342 343 344 345 346 347 348 349 350 351 352 353 354 355 356 357 358 359 360 361 362 363 364 365 366 367 368 369 370 371 372 373 374 375 376 377 378 379 380 381 382 383 384 385 386 387 388 389 390 391 392 393 394 395 396 397 398 399 400

E XP E RI ME N TAL CE L L R ES E ARC H

] (]]]]) ]]]–]]]

cell viability of NSCLC cells independently of KRAS mutations or MDK expression. In addition to cell viability, colony formation of H441 and H2009 cells were also significantly reduced by the combinatorial treatment compared to either iMDK or PD0325901.

Combinatorial treatment of iMDK with PD0325901 increased apoptosis in H441 cells

Fig. 1 – PD0325901 inhibited ERK1/2 phosphorylation induced by iMDK treatment in H441 lung adenocarcinoma cells. A. iMDK suppressed AKT phosphorylation in a dose-dependent manner (0–500 nM) in H441 lung adenocarcinoma cells after treatment for 72 h. In contrast, ERK1/2 phosphorylation was increased by iMDK as assessed by immunoblot analysis. B. PD0325901 (10–250 nM) suppressed ERK1/2 phosphorylation induced by iMDK (200 nM) in H441 cells. Conversely, iMDK suppressed AKT phosphorylation induced by PD0325901. H441 cells were treated with iMDK in the presence or absence of PD0325901 for 72 h.

PD0325901 enhanced iMDK-mediated suppression of cell proliferation and colony formation in H441 cells Having demonstrated that the combinatorial treatment of iMDK and PD0325901 inhibited both the PI3K pathway and the iMDKmediated activation of the MAPK pathway in H441 cells carrying KRASG12V (Fig. 1), we assessed whether the combinatorial treatment would inhibit growth of H441 cells as well as other KRAS mutated NSCLC cells, including H2009 (non-small cell carcinoma; KRASG12A) and A549 (lung carcinoma; KRASG12S) cells, and KRAS wild type H520 (lung squamous cell carcinoma; KRASWT) cells. First, we assessed whether iMDK inhibited the PI3K pathway not only in H441 cells but also other cells, including H2009 and H520 cells. As shown in Supplemental Fig. 3, AKT (downstream of PI3K) activation was inhibited by iMDK, indicating that iMDK inhibits the PI3K pathway in a broad range of NSCLC cells regardless of KRAS mutations. Next, we treated the cells with iMDK and/or a MAPK inhibitor PD0325901. As shown in Fig. 2A, PD0325901 further inhibited iMDK-mediated suppression of cell viability in KRAS mutated H441 and H2009 cells as well as KRAS wild type H520 cells. Though iMDK alone did not inhibit cell viability of A549 cells, the combinatorial treatment of iMDK with PD0325901 significantly inhibited that of A549 cells compared to the single treatment of PD0325901. As shown previously [17], MDK is expressed in H441, H2009 and H520 cells but not in A549 cells. These results indicate that the combinatorial treatment inhibits

In order to analyze the mechanism by which PD0325901 enhances iMDK-mediated cell growth inhibition in the NSCLC cells, we measured the degree of apoptosis after the combinatorial treatment in H441 cells by quantifying activated caspase-3 and TUNEL positive cells, which are markers of apoptosis. As shown in Fig. 3, flow cytometric analysis demonstrated that single use of iMDK or PD0325901 (10 nM) did not induce activation of caspase3 (Fig. 3A). Combinatorial treatment with iMDK and PD0325901 synergistically increased levels of activated caspapse-3. Combinatorial treatment significantly increased the number of TUNEL positive cells compared to either iMDK or PD0325901 alone (Fig. 3B). These results indicate that the inhibition of H441 cell growth by the combinatorial treatment of iMDK with PD0325901 is likely attributed to apoptosis.

iMDK with PD0325901 suppressed tube formation of HUVECs in vitro Endothelial cells contribute to tumor progression by creating blood vessels leading to angiogenesis to fuel tumor cells [26]. We sought to determine whether the combinatorial treatment with iMDK and PD0325901 influences not only NSCLC cells but also angiogenesis constituted from such endothelial cells. First, using TUNEL staining, we assessed whether the treatment of iMDK, PD0325901 or both in combination would induce cell death in HUVEC (Human Umbilical Vein Endothelial Cells). iMDK, PD0325901 or a combination of the two did not induce cell death in HUVEC in vitro (Fig. 4A), indicating that iMDK does not cause apoptosis in normal cells, consistent with our previous report [17]. Next, we assessed whether treatment with iMDK, PD0325901 or both in combination influenced angiogenesis by assessing tube formation in HUVECs, which mimics angiogenesis in vitro. Neither iMDK nor PD0325901, when used alone, inhibited angiogenesis as assessed by HUVEC tube formation. In contrast, combinatorial treatment with iMDK and PD0325901 significantly disrupted tube formation 5 h after treatment (Fig. 4B). Protein expression of MDK was not detected in HUVEC cells (data not shown), indicating that the combinatorial treatment disrupts the tube formation independently of MDK. These results indicate that simultaneous inhibition of the PI3K and MAPK pathways suppresses angiogenesis as well as tumor cell growth, both of which are required for tumor growth.

The combinatorial treatment of iMDK with PD0325901 significantly suppressed tumor growth and tumorassociated angiogenesis in lung Cancer xenograft model in vivo In order to determine whether combinatorial treatment of iMDK with PD0325901 cooperatively suppresses lung tumor growth in vivo, we treated nude mice carrying xenograft tumors derived from H441 cells with iMDK and/or PD0325901. As shown in Fig. 5A, tumor growth

Please cite this article as: N. Ishida, et al., A novel PI3K inhibitor iMDK suppresses non-small cell lung Cancer cooperatively with A MEK inhibitor, Exp Cell Res (2015), http://dx.doi.org/10.1016/j.yexcr.2015.03.019

401 402 403 404 405 406 407 408 409 410 411 412 413 414 415 416 417 418 419 420 421 422 423 424 425 426 427 428 429 430 431 432 433 434 435 436 437 438 439 440 441 442 443 444 445 446 447 448 449 450 451 452 453 454 455 456 457 458 459 460

E X PE R IM EN TA L C ELL R E S EA RC H

461 462 463 464 465 466 467 468 469 470 471 472 473 474 475 476 477 478 479 480 481 482 483 484 485 486 487 488 489 490 491 492 493 494 495 496 497 498 499 500 501 502 503 504 505 506 507 508 509 510 511 512 513 514 515 516 517 518 519 520

] (]]]]) ]]]–]]]

5

Fig. 2 – Combination of iMDK and PD0320901 cooperatively suppressed cell viability and colony formation of NSCLC cells. A. PD0325901 enhanced growth inhibition in H441, H2009 and H520 and A549 NSCLC cells after iMDK treatment. Treatment conditions were described in Materials and Methods. Cell viability was assessed by the WST-1 assay 72 h after exposure to iMDK. Data are presented as % growth inhibition as described in Methods. Statistical significance was defined as po0.05 (#). B. Colony formation of H441 and H2009 cells treated with iMDK (1 μM) and/or PD0325901 (500 nM for H441 cells, 1 μM for H2009 cells). Fourteen days after the treatment, cells were fixed and stained with crystal violet. Representative images of experiments performed in triplicate are shown. C. Mean colony number was derived from quantitation of triplicate dishes for each treatment and arbitrarily set to 100%. Data are shown relative to controls. Statistical significance was defined as po0.05 (#).

Please cite this article as: N. Ishida, et al., A novel PI3K inhibitor iMDK suppresses non-small cell lung Cancer cooperatively with A MEK inhibitor, Exp Cell Res (2015), http://dx.doi.org/10.1016/j.yexcr.2015.03.019

521 522 523 524 525 526 527 528 529 530 531 532 533 534 535 536 537 538 539 540 541 542 543 544 545 546 547 548 549 550 551 552 553 554 555 556 557 558 559 560 561 562 563 564 565 566 567 568 569 570 571 572 573 574 575 576 577 578 579 580

6

581 582 583 584 585 586 587 588 589 590 591 592 593 594 595 596 597 598 599 600 601 602 603 604 605 606 607 608 609 610 611 612 613 614 615 616 617 618 619 620 621 622 623 624 625 626 627 628 629 630 631 632 633 634 635 636 637 638 639 640

E XP E RI ME N TAL CE L L R ES E ARC H

] (]]]]) ]]]–]]]

Fig. 3 – PD0325901 enhanced apoptosis induced by iMDK in H441 lung adenocarcinoma cells. A. H441 lung adenocarcinoma cells were fixed and stained with anti-activated caspase 3 antibodies 72 h after iMDK (10 nM) and/or PD0325901 (10 nM) and analyzed by flow cytometry. The percentage of cells staining for active caspase-3 is indicated. B. PD0325901 enhanced apoptosis induced by iMDK in H441 cells. Cells were treated for 72 h at a concentration of 10 nM iMDK. TUNEL staining was performed as described in Methods. Statistical significance was defined as po0.05 (#).

was significantly suppressed after daily intraperitoneal injection of iMDK or oral administration of PD0325901 5 times a week. The combinatorial treatment of iMDK with PD0325901 further inhibited the tumor growth compared to either iMDK or PD0325901 alone. Angiogenesis in the xenografted tumor, which was assessed by the expression of CD31/PECAM-1, was also cooperatively inhibited by the combinatorial treatment of iMDK with PD0325901 compared to either iMDK or PD0325901 (Fig. 5B and C). These results indicate that the combinatorial treatment of iMDK with PD0325901

significantly suppresses tumor growth and tumor-associated angiogenesis of NSCLC in vivo.

Discussion Molecularly targeted therapies are extending survival of some NSCLC patients compared to conventional chemotherapy [7]; however, a therapeutic target for KRAS-mutant NSCLC and squamous NSCLC,

Please cite this article as: N. Ishida, et al., A novel PI3K inhibitor iMDK suppresses non-small cell lung Cancer cooperatively with A MEK inhibitor, Exp Cell Res (2015), http://dx.doi.org/10.1016/j.yexcr.2015.03.019

641 642 643 644 645 646 647 648 649 650 651 652 653 654 655 656 657 658 659 660 661 662 663 664 665 666 667 668 669 670 671 672 673 674 675 676 677 678 679 680 681 682 683 684 685 686 687 688 689 690 691 692 693 694 695 696 697 698 699 700

E X PE R IM EN TA L C ELL R E S EA RC H

701 702 703 704 705 706 707 708 709 710 711 712 713 714 715 716 717 718 719 720 721 722 723 724 725 726 727 728 729 730 731 732 733 734 735 736 737 738 739 740 741 742 743 744 745 746 747 748 749 750 751 752 753 754 755 756 757 758 759 760

] (]]]]) ]]]–]]]

7

Fig. 4 – iMDK suppressed angiogenesis with PD0325901 in vitro. A. Treatment of HUVEC cells with iMDK, PD0325901 or a combination of the two did not induce cell death as detected by TUNEL staining. Cells were treated for 5 h at a concentration of 10 μM. TUNEL staining was performed as described in Materials and methods. B. Combinatorial treatment of iMDK with PD0325901 significantly inhibited tube formation of HUVECs. HUVEC cells were treated with iMDK (10 μM), PD0325901 (10 μM) or both for 5 h. Tube length was quantified as detailed in Materials and methods. Statistical significance was defined as po0.05 (#).

which are the most common lung cancers, has not been clinically established. A recent clinical trial using a MAPK inhibitor selumetinib, a molecularly targeted drug, in combination with docetaxel (chemotherapeutic [non-molecularly targeted] drug) extended survival by 4 months for patients whose lung adenocarcinomas carry KRAS mutations compared to those using docetaxel alone; however, the combinatorial treatment resulted in increased incidence of adverse effects compared to docetaxel only treatment [13]. There is an urgent need for better therapeutic strategies to treat NSCLCs whose molecular targets are not known.

Previously, we reported that iMDK, which suppresses the expression of MDK, inhibited the PI3K pathway and in turn suppressed the growth of KRAS-mutant NSCLC cells [17]. Unexpectedly, iMDK also activated the MAPK pathway, which suggests the compensatory activation of an alternative tumorigenic pathway for KRAS-mutant NSCLC cell survival. In the present study, killing of KRAS-mutant NSCLC cells was enhanced by inhibiting iMDK-mediated activation of the MAPK pathway using PD0325901 in addition to iMDKmediated suppression of the PI3K pathway. The combinatorial treatment was also effective in reducing tumor growth of KRAS wild

Please cite this article as: N. Ishida, et al., A novel PI3K inhibitor iMDK suppresses non-small cell lung Cancer cooperatively with A MEK inhibitor, Exp Cell Res (2015), http://dx.doi.org/10.1016/j.yexcr.2015.03.019

761 762 763 764 765 766 767 768 769 770 771 772 773 774 775 776 777 778 779 780 781 782 783 784 785 786 787 788 789 790 791 792 793 794 795 796 797 798 799 800 801 802 803 804 805 806 807 808 809 810 811 812 813 814 815 816 817 818 819 820

8

821 822 823 824 825 826 827 828 829 830 831 832 833 834 835 836 837 838 839 840 841 842 843 844 845 846 847 848 849 850 851 852 853 854 855 856 857 858 859 860 861 862 863 864 865 866 867 868 869 870 871 872 873 874 875 876 877 878 879 880

E XP E RI ME N TAL CE L L R ES E ARC H

] (]]]]) ]]]–]]]

Fig. 5 – The combination treatment of iMDK and PD0325901 effectively reduced lung tumor growth in a xenograft mouse model. A. Volume of the tumors derived from H441 lung adenocarcinoma cells was significantly reduced after the combination treatment with iMDK (9 mg/kg) and PD0325901 (5 mg/kg) compared to that of single compound in a xenograft mouse model. Eight mice were used in each group. Tumor volume was monitored every day after inoculation of H441 cells. Tumor growth is expressed as mean tumor volume; bars represent SD. Statistical significance was defined as po0.05 (#). B. Shown is an image of xenograft tumors derived from H441 cells, which are dissected from the xenograft mice 11 days after treatment with iMDK. Expression of an angiogenesis marker CD31/PECAM-1 was detected by immunohistochemistry (scale bar shows 200 μm). C. Expression of CD31/PECAM-1 was significantly inhibited by the combination treatment of iMDK and PD0325901 compared to that of single compound treatment. Statistical significance was defined as po0.05 (#).

type H520 squamous NSCLC cells, indicating that the combinatorial treatment can be applicable for different types of NSCLC. iMDK did not reduce body weight or cause toxicity in mice [17], suggesting that the combinatorial use of iMDK with PD0325901 or other MAPK inhibitors, such as selumetinib, might be superior to the combinatorial use of docetaxel and selumetinib to prevent the adverse effects. Combinatorial therapy with iMDK may also enable treatment using reduced amounts of a MAPK inhibitor. The simultaneous inhibition of both the PI3K and MAPK pathways by small molecule drugs was previously shown to suppress KRAS-mutant NSCLC in vitro and in vivo [15,27]. For example, the combinatorial use of NVPBEZ235 (a PI3K inhibitor) and ARRY-142886 (a MAPK inhibitor) synergistically reduced Kras-mutant lung tumors in a transgenic mouse model [28]. NVP-BEZ235 is currently being tested for safety in clinical trials [29]. While both NVP-BEZ235 and iMDK inhibit the

PI3K pathway, the chemical structure of iMDK is distinct from that of NVP-BEZ235, indicating that iMDK may influence the PI3K pathway in a manner distinct from NVP-BEZ235. Clinical development of iMDK or related compounds as a next generation PI3K drug may be useful. Angiogenesis creates a tumor microenvironment that enhances tumor progression, developing new blood vessels consisting of endothelial cells and thereby supplies nutrients and oxygen to tumor cells [30]. Previously, we demonstrated in a xenograft mouse model using H441 cells that iMDK suppressed expression of CD31/PECAM-1, a marker of angiogenesis [17]. While iMDK suppresses angiogenesis in vivo, it remains unknown whether iMDK directly influences growth or activities of endothelial cells. In the present study, we sought to determine whether the combinatorial treatment of iMDK with PD0325901 suppressed

Please cite this article as: N. Ishida, et al., A novel PI3K inhibitor iMDK suppresses non-small cell lung Cancer cooperatively with A MEK inhibitor, Exp Cell Res (2015), http://dx.doi.org/10.1016/j.yexcr.2015.03.019

881 882 883 884 885 886 887 888 889 890 891 892 893 894 895 896 897 898 899 900 901 902 903 904 905 906 907 908 909 910 911 912 913 914 915 916 917 918 919 920 921 922 923 924 925 926 927 928 929 930 931 932 933 934 935 936 937 938 939 940

E X PE R IM EN TA L C ELL R E S EA RC H

941 942 943 944 945 946 947 948 949 950 951 952 953 954 955 956 957 958 959 960 961 962 963 964 965 966 967 968 969 970 971 972 973 974 975 976 977 978 979 980 981 982 983 984 985 986 987 988 989 990 991 992 993 994 995 996 997 998 999 1000

angiogenesis. Tube formation assays using HUVEC (endothelial cells) recapitulate the angiogenesis process in vitro [31]. The combinatorial treatment of iMDK with PD0325901 suppressed HUVEC tube formation but not HUVEC growth compared to either iMDK or PD0325901 (Fig. 4). The combinatorial treatment also suppressed tumor-associated angiogenesis in the xenograft lung tumor derived from H441 cells compared to those of treatment with a single compound in vivo (Fig. 5). These results suggest that the combinatorial treatment suppresses NSCLC growth by not only directly causing apoptosis of NSCLC cells but also by directly disrupting tumor-associated angiogenesis. In summary, we demonstrated that the combinatorial treatment of a novel PI3K inhibitor iMDK with a MAPK inhibitor PD0325901 cooperatively suppressed the growth of NSCLC cells regardless of KRAS mutations or MDK expression. The combinatorial treatment also disrupted in vitro and in vivo angiogenesis that may influence the tumor-associated environment to limit tumor survival. The strategy using iMDK or related compounds with a MAPK inhibitor may be a promising approach to treat NSCLC whose molecular targets are not identified.

[6] [7] [8]

[9]

[10]

[11]

Acknowledgments Q3 This study was supported by the Ministry of Education, Science, Q4 and Culture, Japan, the National Institute of Health, the American

Lung Association, the University of Cincinnati Postdoctoral Fellow Research Program, and the Cincinnati Children's Hospital Medical Center. The authors thank N. Miyake, S. Ikeda, Y. Kishimoto and M. Durbin for technical assistance and discussion.

Appendix A.

[12]

Supporting information

Supplementary data associated with this article can be found in the online version at http://dx.doi.org/10.1016/j.yexcr.2015.03.019.

[13]

references [14] [1] R. Siegel, D. Naishadham, A. Jemal, Cancer statistics, CA: Cancer J. Clin. 63 (2013) 11–30. [2] M.F. Zakowski, M. Ladanyi, M.G. Kris, EGFR mutations in smallcell lung cancers in patients who have never smoked, N. Engl. J. Med. 355 (2006) 213–215. [3] M.S. Tsao, A. Sakurada, J.C. Cutz, C.Q. Zhu, S. Kamel-Reid, J. Squire, I. Lorimer, T. Zhang, N. Liu, M. Daneshmand, P. Marrano, G. da Cunha Santos, A. Lagarde, F. Richardson, L. Seymour, M. Whitehead, K. Ding, J. Pater, F.A. Shepherd, Erlotinib in lung cancer— molecular and clinical predictors of outcome, N. Engl. J. Med. 353 (2005) 133–144. [4] E.L. Kwak, Y.J. Bang, D.R. Camidge, A.T. Shaw, B. Solomon, R.G. Maki, S.H. Ou, B.J. Dezube, P.A. Janne, D.B. Costa, M. VarellaGarcia, W.H. Kim, T.J. Lynch, P. Fidias, H. Stubbs, J.A. Engelman, L.V. Sequist, W. Tan, L. Gandhi, M. Mino-Kenudson, G.C. Wei, S.M. Shreeve, M.J. Ratain, J. Settleman, J.G. Christensen, D.A. Haber, K. Wilner, R. Salgia, G.I. Shapiro, J.W. Clark, A.J. Iafrate, Anaplastic lymphoma kinase inhibition in non-small-cell lung cancer, N. Engl. J. Med. 363 (2010) 1693–1703. [5] L. Friboulet, N. Li, R. Katayama, C.C. Lee, J.F. Gainor, A.S. Crystal, P.Y. Michellys, M.M. Awad, N. Yanagitani, S. Kim, A.C. Pferdekamper, J. Li, S. Kasibhatla, F. Sun, X. Sun, S. Hua,

[15]

[16]

[17]

[18]

] (]]]]) ]]]–]]]

9

P. McNamara, S. Mahmood, E.L. Lockerman, N. Fujita, M. Nishio, J.L. Harris, A.T. Shaw, J.A. Engelman, The ALK inhibitor ceritinib overcomes crizotinib resistance in non-small cell lung cancer, Cancer Discov. 4 (2014) 662–673. T. Mitsudomi, K. Suda, Y. Yatabe, Surgery for NSCLC in the era of personalized medicine, Nat. Rev. Clin. Oncol. 10 (2013) 235–244. K. Suda, T. Mitsudomi, Successes and limitations of targeted cancer therapy in lung cancer, Prog. Tumor Res. 41 (2014) 62–77. K. Takeuchi, M. Soda, Y. Togashi, R. Suzuki, S. Sakata, S. Hatano, R. Asaka, W. Hamanaka, H. Ninomiya, H. Uehara, Y. Lim Choi, Y. Satoh, S. Okumura, K. Nakagawa, H. Mano, Y. Ishikawa, RET, ROS1 and ALK fusions in lung cancer, Nat. Med. 18 (2012) 378– 381. N. Yamaguchi, P.A. Vanderlaan, E. Folch, D.H. Boucher, H.M. Canepa, M.S. Kent, S.P. Gangadharan, A. Majid, O.N. Kocher, M.A. Goldstein, M.S. Huberman, D.B. Costa, Smoking status and selfreported race affect the frequency of clinically relevant oncogenic alterations in non-small-cell lung cancers at a United Statesbased academic medical practice, Lung Cancer 82 (2013) 31–37. R.G. Liao, H. Watanabe, M. Meyerson, P.S. Hammerman, Targeted therapy for squamous cell lung cancer, Lung Cancer Manag. 1 (2012) 293–300. N.T. Ihle, L.A. Byers, E.S. Kim, P. Saintigny, J.J. Lee, G.R. Blumenschein, A. Tsao, S. Liu, J.E. Larsen, J. Wang, L. Diao, K.R. Coombes, L. Chen, S. Zhang, M.F. Abdelmelek, X. Tang, V. Papadimitrakopoulou, J.D. Minna, S.M. Lippman, W.K. Hong, R. S. Herbst, J.V. Wistuba II, G. Heymach, Powis, Effect of KRAS oncogene substitutions on protein behavior: implications for signaling and clinical outcome, J. Natl. Cancer Inst. 104 (2012) 228–239. M.L. Sos, K. Michel, T. Zander, J. Weiss, P. Frommolt, M. Peifer, D. Li, R. Ullrich, M. Koker, F. Fischer, T. Shimamura, D. Rauh, C. Mermel, S. Fischer, I. Stuckrath, S. Heynck, R. Beroukhim, W. Lin, W. Winckler, K. Shah, T. LaFramboise, W.F. Moriarty, M. Hanna, L. Tolosi, J. Rahnenfuhrer, R. Verhaak, D. Chiang, G. Getz, M. Hellmich, J. Wolf, L. Girard, M. Peyton, B.A. Weir, T.H. Chen, H. Greulich, J. Barretina, G.I. Shapiro, L.A. Garraway, A.F. Gazdar, J.D. Minna, M. Meyerson, K.K. Wong, R.K. Thomas, Predicting drug susceptibility of non-small cell lung cancers based on genetic lesions, J. Clin. Investig. 119 (2009) 1727–1740. P.A. Janne, A.T. Shaw, J.R. Pereira, G. Jeannin, J. Vansteenkiste, C. Barrios, F.A. Franke, L. Grinsted, V. Zazulina, P. Smith, I. Smith, L. Crino, Selumetinib plus docetaxel for KRAS-mutant advanced non-small-cell lung cancer: a randomised, multicentre, placebocontrolled, phase 2 study, Lancet Oncol. 14 (2013) 38–47. A.G. Stephen, D. Esposito, R.K. Bagni, F. McCormick, Dragging ras back in the ring, Cancer cell 25 (2014) 272–281. E. Rozengurt, H.P. Soares, J. Sinnet-Smith, Suppression of feedback loops mediated by PI3K/mTOR induces multiple overactivation of compensatory pathways: an unintended consequence leading to drug resistance, Mol. Cancer Ther. 13 (2014) 2477– 2488. M.L. Sos, S. Fischer, R. Ullrich, M. Peifer, J.M. Heuckmann, M. Koker, S. Heynck, I. Stuckrath, J. Weiss, F. Fischer, K. Michel, A. Goel, L. Regales, K.A. Politi, S. Perera, M. Getlik, L.C. Heukamp, S. Ansen, T. Zander, R. Beroukhim, H. Kashkar, K.M. Shokat, W.R. Sellers, D. Rauh, C. Orr, K.P. Hoeflich, L. Friedman, K.K. Wong, W. Pao, R.K. Thomas, Identifying genotype-dependent efficacy of single and combined PI3K- and MAPK-pathway inhibition in cancer, Proc. Natl. Acad. Sci. USA 106 (2009) 18351–18356. H. Hao, Y. Maeda, T. Fukazawa, T. Yamatsuji, M. Takaoka, X.H. Bao, J. Matsuoka, T. Okui, T. Shimo, N. Takigawa, Y. Tomono, M. Nakajima, I.M. Fink-Baldauf, S. Nelson, W. Seibel, R. Papoian, J.A. Whitsett, Y. Naomoto, Inhibition of the growth factor MDK/ midkine by a novel small molecule compound to treat non-small cell lung cancer, PloS One 8 (2013) e71093. A.K. Pandurangan, P. Dharmalingam, S.K. Sadagopan, M. Ramar, A. Munusamy, S. Ganapasam, Luteolin induces growth arrest in

Please cite this article as: N. Ishida, et al., A novel PI3K inhibitor iMDK suppresses non-small cell lung Cancer cooperatively with A MEK inhibitor, Exp Cell Res (2015), http://dx.doi.org/10.1016/j.yexcr.2015.03.019

1001 1002 1003 1004 1005 1006 1007 1008 1009 1010 1011 1012 1013 1014 1015 1016 1017 1018 1019 1020 1021 1022 1023 1024 1025 1026 1027 1028 1029 1030 1031 1032 1033 1034 1035 1036 1037 1038 1039 1040 1041 1042 1043 1044 1045 1046 1047 1048 1049 1050 1051 1052 1053 1054 1055 1056 1057 1058 1059 1060

10

1061 1062 1063 1064 1065 1066 1067 1068 1069 1070 1071 1072 1073 1074 1075 1076 1077 1078 1079 1080 1081 1082 1083 1084 1085 1086 1087 1088 1089 1090 1091 1092 1093

[19]

[20]

[21]

[22]

[23] [24]

[25]

[26] [27]

E XP E RI ME N TAL CE L L R ES E ARC H

colon cancer cells through involvement of Wnt/beta-catenin/ GSK-3beta signaling, J. Environ. Pathol. Toxicol. Oncol.: Off. Organ Int. Soc. Environ. Toxicol. Cancer 32 (2013) 131–139. F. Belloc, M.A. Belaud-Rotureau, V. Lavignolle, E. Bascans, E. BrazPereira, F. Durrieu, F. Lacombe, Flow cytometry detection of caspase 3 activation in preapoptotic leukemic cells, Cytometry 40 (2000) 151–160. N. D'Haene, S. Sauvage, C. Maris, I. Adanja, M. Le Mercier, C. Decaestecker, L. Baum, I. Salmon, VEGFR1 and VEGFR2 involvement in extracellular galectin-1- and galectin-3-induced angiogenesis, PloS One 8 (2013) e67029. M.J. Lee, S. Thangada, K.P. Claffey, N. Ancellin, C.H. Liu, M. Kluk, M. Volpi, R.I. Sha’afi, T. Hla, Vascular endothelial cell adherens junction assembly and morphogenesis induced by sphingosine1-phosphate, Cell 99 (1999) 301–312. Z. Zhang, G. Jiang, F. Yang, J. Wang, Knockdown of mutant K-ras expression by adenovirus-mediated siRNA inhibits the in vitro and in vivo growth of lung cancer cells, Cancer Biol. Ther. 5 (2006) 1481–1486. S. Grant, Cotargeting survival signaling pathways in cancer, J. Clin. Investig. 118 (2008) 3003–3006. K. Suda, K. Tomizawa, M. Fujii, H. Murakami, H. Osada, Y. Maehara, Y. Yatabe, Y. Sekido, T. Mitsudomi, Epithelial to mesenchymal transition in an epidermal growth factor receptormutant lung cancer cell line with acquired resistance to erlotinib, J. Thorac. Oncol.: Off. Publ. Int. Assoc. Study Lung Cancer 6 (2011) 1152–1161. E. Halilovic, Q.B. She, Q. Ye, R. Pagliarini, W.R. Sellers, D.B. Solit, N. Rosen, PIK3CA mutation uncouples tumor growth and cyclin D1 regulation from MEK/ERK and mutant KRAS signaling, Cancer Res. 70 (2010) 6804–6814. R.S. Kerbel, Tumor angiogenesis, N. Engl. J. Med. 358 (2008) 2039–2049. J. Meng, B. Dai, B. Fang, B.N. Bekele, W.G. Bornmann, D. Sun, Z. Peng, R.S. Herbst, V. Papadimitrakopoulou, J.D. Minna, M. Peyton, J.A. Roth, Combination treatment with MEK and AKT

[28]

[29] [30]

[31]

] (]]]]) ]]]–]]]

inhibitors is more effective than each drug alone in human nonsmall cell lung cancer in vitro and in vivo, PloS One 5 (2010) e14124. J.A. Engelman, L. Chen, X. Tan, K. Crosby, A.R. Guimaraes, R. Upadhyay, M. Maira, K. McNamara, S.A. Perera, Y. Song, L.R. Chirieac, R. Kaur, A. Lightbown, J. Simendinger, T. Li, R.F. Padera, C. Garcia-Echeverria, R. Weissleder, U. Mahmood, L.C. Cantley, K.K. Wong, Effective use of PI3K and MEK inhibitors to treat mutant Kras G12D and PIK3CA H1047R murine lung cancers, Nat. Med. 14 (2008) 1351–1356. P.I.P.I.C.Z.T. Safety Study of BEZ235 With Everolimus in Subjects With Advanced Solid Tumors Phase: Phase II, NCT01508104. K. Mittal, J. Ebos, B. Rini, Angiogenesis and the tumor microenvironment: vascular endothelial growth factor and beyond, Semin. Oncol. 41 (2014) 235–251. D.E. Morales, K.A. McGowan, D.S. Grant, S. Maheshwari, D. Bhartiya, M.C. Cid, H.K. Kleinman, H.W. Schnaper, Estrogen promotes angiogenic activity in human umbilical vein endothelial cells in vitro and in a murine model, Circulation 91 (1995) 755–763.

Please cite this article as: N. Ishida, et al., A novel PI3K inhibitor iMDK suppresses non-small cell lung Cancer cooperatively with A MEK inhibitor, Exp Cell Res (2015), http://dx.doi.org/10.1016/j.yexcr.2015.03.019

1094 1095 1096 1097 1098 1099 1100 1101 1102 1103 1104 1105 1106 1107 1108 1109 1110 1111 1112 1113 1114 1115 1116 1117 1118 1119 1120 1121 1122 1123 1124 1125

A novel PI3K inhibitor iMDK suppresses non-small cell lung Cancer cooperatively with A MEK inhibitor.

The PI3K-AKT pathway is expected to be a therapeutic target for non-small cell lung cancer (NSCLC) treatment. We previously reported that a novel PI3K...
5MB Sizes 0 Downloads 10 Views