Jong Ho Cho, Wei Zhou, Yoon-La Choi, Jong-Mu Sun, Hyejoo Choi, Tae-Eun Kim, Marisa Dolled-Filhart, Kenneth Emancipator, Mary Anne Rutkowski, Jhingook Kim
Cancer Res Treat. 2018;50(1):95-102. Published online March 17, 2017
Purpose
Data are limited on programmed death ligand 1 (PD-L1) expression in epidermal growth factor receptor (EGFR)-mutant non-small cell lung cancer (NSCLC).
Materials and Methods
We retrospectively evaluated the relationship between PD-L1 expression and recurrence-free survival (RFS) and overall survival in 319 patients with EGFR-mutant NSCLC who were treated at Samsung Medical Center from 2006 to 2014. Membranous PD-L1 expression on tumor cells was measured using the PD-L1 IHC 22C3 pharmDx antibody and reported as tumor proportion score (TPS). Kaplan-Meier methods, log-rank test, and Cox proportional hazards models were used for survival analysis.
Results
All patients had ≥ 1 EGFR mutation—54% in exon 19 and 39% in exon 21. Overall, 51% of patients had PD-L1–positive tumors. The prevalence of PD-L1 positivity was higher among patients with stages II-IV versus stage I disease (64% vs. 44%) and among patients with other EGFR mutations (75%) than with L858R mutation (39%) or exon 19 deletion (52%). PD-L1 positivity was associated with shorter RFS, with an adjusted hazard ratio of 1.52 (95% confidence interval [CI], 0.81 to 2.84; median, 18 months) for the PD-L1 TPS ≥ 50% group, 1.51 (95% CI, 1.02 to 2.21; median, 31 months) for the PD-L1 TPS 1%-49% group, and 1.51 (95% CI, 1.05 to 2.18) for the combined PD-L1–positive groups (TPS ≥ 1%) compared with the PD-L1–negative group (median, 35 months).
Conclusion
PD-L1 expression is associated with disease stage and type of EGFR mutation. PD-L1 positivity might be associated with worse RFS among patients with surgically treated EGFR-mutant NSCLC.
Citations
Citations to this article as recorded by
Immunological features of EGFR-mutant non-small cell lung cancer and clinical practice: a narrative review Yi Dong, Liaqat Khan, Yi Yao Journal of the National Cancer Center.2024; 4(4): 289. CrossRef
Activatable near-infrared fluorescence probe for real-time imaging of PD-L1 expression in tumors Hyunjin Kim, Maixian Liu, Chan Hyeok Park, Byung Il Lee, Hyonchol Jang, Yongdoo Choi Journal of Materials Chemistry B.2024; 12(42): 10877. CrossRef
Influence of PD‐L1 expression on the efficacy of EGFR‐TKIs in EGFR‐mutant non‐small cell lung cancer Si‐Yu Lei, Hai‐Yan Xu, Hong‐Shuai Li, Ya‐Ning Yang, Fei Xu, Jun‐Ling Li, Zhi‐Jie Wang, Pu‐Yuan Xing, Xue‐Zhi Hao, Yan Wang Thoracic Cancer.2023; 14(24): 2327. CrossRef
Association of PD‐L1 tumor proportion score ≥20% with early resistance to osimertinib in patients with EGFR‐mutated NSCLC Yusuke Hamakawa, Yoko Agemi, Aya Shiba, Toshiki Ikeda, Yuko Higashi, Masaharu Aga, Kazuhito Miyazaki, Yuri Taniguchi, Yuki Misumi, Yukiko Nakamura, Tsuneo Shimokawa, Yusuke Saigusa, Nobuaki Kobayashi, Hiroaki Okamoto, Takeshi Kaneko Cancer Medicine.2023; 12(17): 17788. CrossRef
Determining Risk Factors Associated with Depression and Anxiety in Young Lung Cancer Patients: A Novel Optimization Algorithm Yu-Wei Fang, Chieh-Yu Liu Medicina.2021; 57(4): 340. CrossRef
Anti-PD1/PD-L1 Immunotherapy for Non-Small Cell Lung Cancer with Actionable Oncogenic Driver Mutations Edouard Dantoing, Nicolas Piton, Mathieu Salaün, Luc Thiberville, Florian Guisier International Journal of Molecular Sciences.2021; 22(12): 6288. CrossRef
Programmed Death Ligand 1 Expression and Related Markers in Pleuropulmonary Blastoma Zahra Alipour, Kris Ann P Schultz, Ling Chen, Anne K Harris, Ivan A Gonzalez, John Pfeifer, D Ashley Hill, Mai He, Louis P Dehner Pediatric and Developmental Pathology.2021; 24(6): 523. CrossRef
The predictive and prognostic effects of PD-L1 expression on TKI treatment and survival of EGFR-mutant NSCLC Bo Lan, Yongfang Wang, Jingni Wu, Kai Wang, Pingli Wang Medicine.2021; 100(34): e27038. CrossRef
The Clinicopathological and Molecular Associations of PD-L1 Expression in Non-small Cell Lung Cancer: Analysis of a Series of 10,005 Cases Tested with the 22C3 Assay Matthew Evans, Brendan O’Sullivan, Frances Hughes, Tina Mullis, Matthew Smith, Nicola Trim, Philippe Taniere Pathology & Oncology Research.2020; 26(1): 79. CrossRef
PD-L1 expression and response to pembrolizumab in patients with EGFR-mutant non-small cell lung cancer Eriko Miyawaki, Haruyasu Murakami, Keita Mori, Nobuaki Mamesaya, Takahisa Kawamura, Haruki Kobayashi, Shota Omori, Kazushige Wakuda, Akira Ono, Hirotsugu Kenmotsu, Tateaki Naito, Toshiaki Takahashi Japanese Journal of Clinical Oncology.2020; 50(5): 617. CrossRef
Tumor mutation burden and checkpoint immunotherapy markers in primary and metastatic synovial sarcoma Mai He, Brooj Abro, Madhurima Kaushal, Ling Chen, Tiffany Chen, Mercia Gondim, Weisi Yan, Julie Neidich, Louis P. Dehner, John D. Pfeifer Human Pathology.2020; 100: 15. CrossRef
Clinicopathological characteristics of primary lung nuclear protein in testis carcinoma: A single‐institute experience of 10 cases Yoon Ah Cho, Yoon‐La Choi, Inwoo Hwang, Kyungjong Lee, Jong Ho Cho, Joungho Han Thoracic Cancer.2020; 11(11): 3205. CrossRef
Impact of EGFR mutation on the clinical efficacy of PD-1 inhibitors in patients with pulmonary adenocarcinoma Jang Ho Cho, Hyun Ae Jung, Se-Hoon Lee, Jin Seok Ahn, Myung-Ju Ahn, Keunchil Park, Jong-Mu Sun Journal of Cancer Research and Clinical Oncology.2019; 145(5): 1341. CrossRef
The canonical TGF-β/Smad signalling pathway is involved in PD-L1-induced primary resistance to EGFR-TKIs in EGFR-mutant non-small-cell lung cancer Yang Zhang, Yuanyuan Zeng, Ting Liu, Wenwen Du, Jianjie Zhu, Zeyi Liu, Jian-an Huang Respiratory Research.2019;[Epub] CrossRef
Association with PD-L1 Expression and Clinicopathological Features in 1000 Lung Cancers: A Large Single-Institution Study of Surgically Resected Lung Cancers with a High Prevalence of EGFR Mutation Lee, Kim, Sung, Lee, Han, Kim, Choi International Journal of Molecular Sciences.2019; 20(19): 4794. CrossRef
Clinical and Molecular Predictors of PD-L1 Expression in Non–Small-Cell Lung Cancer: Systematic Review and Meta-analysis Fausto Petrelli, Mariangela Maltese, Gianluca Tomasello, Barbara Conti, Karen Borgonovo, Mary Cabiddu, Mara Ghilardi, Michele Ghidini, Rodolfo Passalacqua, Sandro Barni, Matteo Brighenti Clinical Lung Cancer.2018; 19(4): 315. CrossRef
Status of programmed death-ligand 1 expression in sarcomas Hyung Kyu Park, Mingi Kim, Minjung Sung, Seung Eun Lee, Yu Jin Kim, Yoon-La Choi Journal of Translational Medicine.2018;[Epub] CrossRef
Association between PD-L1 expression and driver gene status in non-small-cell lung cancer: a meta-analysis Bo Lan, Chengxi Ma, Chengyan Zhang, Shoujie Chai, Pingli Wang, Liren Ding, Kai Wang Oncotarget.2018; 9(7): 7684. CrossRef
Hippo effector YAP directly regulates the expression of PD-L1 transcripts in EGFR-TKI-resistant lung adenocarcinoma Byung Soo Lee, Dong Il Park, Da Hye Lee, Jeong Eun Lee, Min-kyung Yeo, Yeon Hee Park, Dae Sik Lim, Wonyoung Choi, Da Hye Lee, Geon Yoo, Han-byul Kim, Dahyun Kang, Jae Young Moon, Sung Soo Jung, Ju Ock Kim, Sang Yeon Cho, Hee Sun Park, Chaeuk Chung Biochemical and Biophysical Research Communications.2017; 491(2): 493. CrossRef
Purpose The purpose of this study is to determine the maximum tolerated dose (MTD), safety, pharmacokinetics, and recommended phase II dose of an oral drug composed of paclitaxel and HM30181A, which is an inhibitor of P-glycoprotein, in patients with advanced cancers. Materials and Methods Patients with advanced solid tumors received standard therapy were given the study drug at escalating doses, using a 3+3 design. The study drug was orally administered on days 1, 8, and 15, with a 28-day cycle of administration. The dose of paclitaxel was escalated from 60 to 420 mg/m2, and the dose of HM30181A was escalated from 30-210 mg/m2. Results A total of twenty-four patients were enrolled. Only one patient experienced a doselimiting toxicity—a grade 3 neutropenia that persisted for more than 2 weeks, at 240 mg/m2 of paclitaxel. MTD was not reached. The maximum plasma concentration was obtained at a dose level of 300 mg/m2 and the area under the curve of plasma concentration- time from 0 to the most recent plasma concentration measurement of paclitaxel was reached at a dose level of 420 mg/m2. The absorption of paclitaxel tends to be limited at doses that exceed 300 mg/m2. The effective plasma concentration of paclitaxel was achieved at a dose of 120 mg/m2. Responses of 23 patients were evaluated; 8 (34.8%) had stable disease and 15 (65.2%) had progressive disease. Conclusion The study drug appears to be well tolerated, and the effective plasma concentration of paclitaxel was achieved. The recommended phase II dose for oral paclitaxel is 300 mg/m2.
Citations
Citations to this article as recorded by
Screening of photosensitizers-ATP binding cassette (ABC) transporter interactions in vitro Shruti Vig, Payal Srivastava, Idrisa Rahman, Renee Jaranson, Anika Dasgupta, Robert Perttilä, Petteri Uusimaa, Huang-Chiao Huang Cancer Drug Resistance.2024;[Epub] CrossRef
Pharmacokinetic modulation of substrate drugs via the inhibition of drug-metabolizing enzymes and transporters using pharmaceutical excipients Min-Koo Choi, Jihoon Lee, Im-Sook Song Journal of Pharmaceutical Investigation.2023; 53(1): 1. CrossRef
Redox-responsive drug-inhibitor conjugate encapsulated in DSPE-PEG2k micelles for overcoming multidrug resistance to chemotherapy Penghui Wang, Yuling Wang, Xuelin Xia, Wei Huang, Deyue Yan Biomaterials Science.2023; 11(12): 4335. CrossRef
Oral paclitaxel and encequidar in patients with breast cancer: a pharmacokinetic, safety, and antitumor activity study Ming-Shen Dai, Ta-Chung Chao, Chang-Fang Chiu, Yen-Shen Lu, Her-Shyong Shiah, Christopher G. C. A. Jackson, Noelyn Hung, Jianguo Zhi, David L. Cutler, Rudolf Kwan, Douglas Kramer, Wing-Kai Chan, Albert Qin, Kuan-Chiao Tseng, Cheung Tak Hung, Tsu-Yi Chao Therapeutic Advances in Medical Oncology.2023;[Epub] CrossRef
Population pharmacokinetics for oral paclitaxel in patients with advanced/metastatic solid tumors Jimmy He, Christopher G. C. A. Jackson, Sanjeev Deva, Tak Hung, Katriona Clarke, Eva Segelov, Tsu‐Yi Chao, Ming‐Shen Dai, Hsien‐Tang Yeh, Wen Wee Ma, Douglas Kramer, Wing‐Kai Chan, Rudolf Kwan, David Cutler, Jay Zhi CPT: Pharmacometrics & Systems Pharmacology.2022; 11(7): 867. CrossRef
A phase Ib study of Oraxol (oral paclitaxel and encequidar) in patients with advanced malignancies Wen Wee Ma, Jenny J. Li, Nilofer S. Azad, Elaine T. Lam, Jennifer R. Diamond, Grace K. Dy, Mateusz Opyrchal, Jay Zhi, Douglas Kramer, Wing-Kai Chan, David Cutler, Rudolf Kwan, Alex A. Adjei, Antonio Jimeno Cancer Chemotherapy and Pharmacology.2022; 90(1): 7. CrossRef
Carbon nano-onion-mediated dual targeting of P-selectin and P-glycoprotein to overcome cancer drug resistance Hai Wang, Yutong Liang, Yue Yin, Jie Zhang, Wen Su, Alisa M. White, Bin Jiang, Jiangsheng Xu, Yuntian Zhang, Samantha Stewart, Xiongbin Lu, Xiaoming He Nature Communications.2021;[Epub] CrossRef
The Intravenous to Oral Switch of Taxanes: Strategies and Current Clinical Developments Marit A C Vermunt, Andries M Bergman, Eric van der Putten, Jos H Beijnen Future Oncology.2021; 17(11): 1379. CrossRef
Itraconazole synergistically increases therapeutic effect of paclitaxel and 99mTc-MIBI accumulation, as a probe of P-gp activity, in HT-29 tumor-bearing nude mice Mahdi Ghadi, Seyed Jalal Hosseinimehr, Fereshteh Talebpour Amiri, Alireza Mardanshahi, Zohreh Noaparast European Journal of Pharmacology.2021; : 173892. CrossRef
Discovery of Encequidar, First-in-Class Intestine Specific P-glycoprotein Inhibitor Michael P. Smolinski, Sameer Urgaonkar, Laura Pitzonka, Murray Cutler, GwanSun Lee, Kwee Hyun Suh, Johnson Y. N. Lau Journal of Medicinal Chemistry.2021; 64(7): 3677. CrossRef
Oral paclitaxel with encequidar compared to intravenous paclitaxel in patients with advanced cancer: A randomised crossover pharmacokinetic study Christopher G. C. A. Jackson, Tak Hung, Eva Segelov, Paula Barlow, Hans Prenen, Blair McLaren, Noelyn Anne Hung, Katriona Clarke, Tsu‐Yi Chao, Ming‐Shen Dai, Hsien‐Tang Yeh, David L. Cutler, Douglas Kramer, Jimmy He, Jay Zhi, Wing‐Kai Chan, Rudolf Kwan, S British Journal of Clinical Pharmacology.2021; 87(12): 4670. CrossRef
Further enhanced dissolution and oral bioavailability of docetaxel by coamorphization with a natural P-gp inhibitor myricetin Yuanfeng Wei, Shengyan Zhou, Tianyun Hao, Jianjun Zhang, Yuan Gao, Shuai Qian European Journal of Pharmaceutical Sciences.2019; 129: 21. CrossRef
DHP23002 as a next generation oral paclitaxel formulation for pancreatic cancer therapy Eunseo Jang, Minhee Son, Junhee Jang, In-Hyun Lee, Sol Kim, Taejun Kwon, Yong-hyun Jeon, Woo-Suk Koh, Kil-Soo Kim, Sang Kyoon Kim, Sumitra Deb PLOS ONE.2019; 14(11): e0225095. CrossRef
Evading P-glycoprotein mediated-efflux chemoresistance using Solid Lipid Nanoparticles Marco C. Cavaco, Carolina Pereira, Bruna Kreutzer, Luis F. Gouveia, Beatriz Silva-Lima, Alexandra M. Brito, Mafalda Videira European Journal of Pharmaceutics and Biopharmaceutics.2017; 110: 76. CrossRef
Effect of HM30181 mesylate salt-loaded microcapsules on the oral absorption of paclitaxel as a novel P-glycoprotein inhibitor Jin Cheul Kim, Kyeong Soo Kim, Dong Shik Kim, Sung Giu Jin, Dong Wuk Kim, Yong Il Kim, Jae-Hyun Park, Jong Oh Kim, Chul Soon Yong, Yu Seok Youn, Jong Soo Woo, Han-Gon Choi International Journal of Pharmaceutics.2016; 506(1-2): 93. CrossRef
Direct in vivo evidence on the mechanism by which nanoparticles facilitate the absorption of a water insoluble, P-gp substrate Ramesh Soundararajan, Kenji Sasaki, Lisa Godfrey, Uchechukwu Odunze, Nancy Fereira, Andreas Schätzlein, Ijeoma Uchegbu International Journal of Pharmaceutics.2016; 514(1): 121. CrossRef