1
|
Rijmers J, Sparidans RW, Acda M, Loos NHC, Epeslidou E, Bui V, Lebre MC, Tibben M, Beijnen JH, Schinkel AH. Brain Exposure to the Macrocyclic ALK Inhibitor Zotizalkib is Restricted by ABCB1, and Its Plasma Disposition is Affected by Mouse Carboxylesterase 1c. Mol Pharm 2024; 21:5159-5170. [PMID: 39312722 DOI: 10.1021/acs.molpharmaceut.4c00542] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/25/2024]
Abstract
Zotizalkib (TPX-0131), a fourth-generation macrocyclic anaplastic lymphoma kinase (ALK) inhibitor, is designed to overcome resistance due to secondary ALK mutations in non-small cell lung cancer (NSCLC). We here evaluated the pharmacokinetic roles of the ABCB1 (P-gp/MDR1) and ABCG2 (BCRP) efflux transporters, OATP1 influx transporters and the metabolizing enzymes CES1 and CYP3A in plasma and tissue disposition of zotizalkib after oral administration in relevant mouse models. Zotizalkib was efficiently transported by hABCB1 in vitro. In vivo, a significant ∼9-fold higher brain-to-plasma ratio was observed in Abcb1a/b-/- and Abcb1a/b;Abcg2-/- compared to wild-type mice. No change in brain disposition was observed in Abcg2-/- mice, suggesting that mAbcb1a/b markedly restricts the brain accumulation of zotizalkib. ABCB1-mediated efflux of zotizalkib was completely inhibited by elacridar, a dual ABCB1/ABCG2 inhibitor, increasing brain exposure without any signs of acute CNS-related toxicities. In Oatp1a/b-/- mice, no marked changes in plasma exposure or tissue-to-plasma ratios were observed, indicating that zotizalkib is not a substantial in vivo substrate for mOatp1a/b. Zotizalkib may further be metabolized by CYP3A4 but only noticeably at low plasma concentrations. In Ces1-/- mice, a 2.5-fold lower plasma exposure was seen compared to wild-type, without alterations in tissue distribution. This suggests increased plasma retention of zotizalkib by binding to the abundant mouse plasma Ces1c. Notably, the hepatic expression of human CES1 did not affect zotizalkib plasma exposure or tissue distribution. The obtained pharmacokinetic insights may be useful for the further development and optimization of therapeutic efficacy and safety of zotizalkib and related compact macrocyclic ALK inhibitors.
Collapse
MESH Headings
- Animals
- Mice
- Brain/metabolism
- ATP Binding Cassette Transporter, Subfamily G, Member 2/metabolism
- ATP Binding Cassette Transporter, Subfamily G, Member 2/genetics
- ATP Binding Cassette Transporter, Subfamily G, Member 2/antagonists & inhibitors
- Anaplastic Lymphoma Kinase/antagonists & inhibitors
- Anaplastic Lymphoma Kinase/metabolism
- Anaplastic Lymphoma Kinase/genetics
- ATP Binding Cassette Transporter, Subfamily B/metabolism
- ATP Binding Cassette Transporter, Subfamily B/genetics
- Mice, Knockout
- Male
- Cytochrome P-450 CYP3A/metabolism
- Cytochrome P-450 CYP3A/genetics
- Humans
- Tissue Distribution
- Carboxylic Ester Hydrolases/metabolism
- Carboxylic Ester Hydrolases/genetics
- Carboxylic Ester Hydrolases/antagonists & inhibitors
- Carboxylesterase/metabolism
- Carboxylesterase/antagonists & inhibitors
- Carboxylesterase/genetics
- Administration, Oral
- Organic Anion Transport Protein 1/metabolism
- Organic Anion Transport Protein 1/genetics
- Organic Anion Transport Protein 1/antagonists & inhibitors
- Protein Kinase Inhibitors/pharmacokinetics
- Protein Kinase Inhibitors/pharmacology
- Carcinoma, Non-Small-Cell Lung/drug therapy
- Carcinoma, Non-Small-Cell Lung/metabolism
Collapse
Affiliation(s)
- Jamie Rijmers
- Division of Pharmacology, The Netherlands Cancer Institute, Amsterdam 1066 CX, The Netherlands
| | - Rolf W Sparidans
- Faculty of Science, Department of Pharmaceutical Sciences, Division of Pharmacoepidemiology and Clinical Pharmacology, Utrecht University, Utrecht 3584 CS, The Netherlands
| | - Manon Acda
- Division of Pharmacy and Pharmacology, The Netherlands Cancer Institute, Amsterdam 1066 CX, The Netherlands
| | - Nancy H C Loos
- Division of Pharmacology, The Netherlands Cancer Institute, Amsterdam 1066 CX, The Netherlands
| | - Emmanouela Epeslidou
- Division of Pharmacology, The Netherlands Cancer Institute, Amsterdam 1066 CX, The Netherlands
| | - Viët Bui
- Division of Pharmacology, The Netherlands Cancer Institute, Amsterdam 1066 CX, The Netherlands
| | - Maria C Lebre
- Division of Pharmacology, The Netherlands Cancer Institute, Amsterdam 1066 CX, The Netherlands
| | - Matthijs Tibben
- Division of Pharmacy and Pharmacology, The Netherlands Cancer Institute, Amsterdam 1066 CX, The Netherlands
| | - Jos H Beijnen
- Division of Pharmacology, The Netherlands Cancer Institute, Amsterdam 1066 CX, The Netherlands
- Faculty of Science, Department of Pharmaceutical Sciences, Division of Pharmacoepidemiology and Clinical Pharmacology, Utrecht University, Utrecht 3584 CS, The Netherlands
- Division of Pharmacy and Pharmacology, The Netherlands Cancer Institute, Amsterdam 1066 CX, The Netherlands
| | - Alfred H Schinkel
- Division of Pharmacology, The Netherlands Cancer Institute, Amsterdam 1066 CX, The Netherlands
| |
Collapse
|
2
|
Poei D, Ali S, Ye S, Hsu R. ALK inhibitors in cancer: mechanisms of resistance and therapeutic management strategies. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2024; 7:20. [PMID: 38835344 PMCID: PMC11149099 DOI: 10.20517/cdr.2024.25] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 04/18/2024] [Accepted: 05/08/2024] [Indexed: 06/06/2024]
Abstract
Anaplastic lymphoma kinase (ALK) gene rearrangements have been identified as potent oncogenic drivers in several malignancies, including non-small cell lung cancer (NSCLC). The discovery of ALK inhibition using a tyrosine kinase inhibitor (TKI) has dramatically improved the outcomes of patients with ALK-mutated NSCLC. However, the emergence of intrinsic and acquired resistance inevitably occurs with ALK TKI use. This review describes the molecular mechanisms of ALK TKI resistance and discusses management strategies to overcome therapeutic resistance.
Collapse
Affiliation(s)
- Darin Poei
- Department of Internal Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Sana Ali
- Division of Medical Oncology, University of Southern California Norris Comprehensive Cancer Center, Los Angeles, CA 90033, USA
| | - Shirley Ye
- Department of Internal Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Robert Hsu
- Division of Medical Oncology, University of Southern California Norris Comprehensive Cancer Center, Los Angeles, CA 90033, USA
| |
Collapse
|
3
|
Yu Y, Wang Z, Wang L, Wang Q, Tang R, Xiang S, Deng Q, Hou T, Sun H. Deciphering the Shared and Specific Drug Resistance Mechanisms of Anaplastic Lymphoma Kinase via Binding Free Energy Computation. RESEARCH (WASHINGTON, D.C.) 2023; 6:0170. [PMID: 37342628 PMCID: PMC10278961 DOI: 10.34133/research.0170] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Accepted: 05/25/2023] [Indexed: 06/23/2023]
Abstract
Anaplastic lymphoma kinase (ALK), a tyrosine receptor kinase, has been proven to be associated with the occurrence of numerous malignancies. Although there have been already at least 3 generations of ALK inhibitors approved by FDA or in clinical trials, the occurrence of various mutations seriously attenuates the effectiveness of the drugs. Unfortunately, most of the drug resistance mechanisms still remain obscure. Therefore, it is necessary to reveal the bottom reasons of the drug resistance mechanisms caused by the mutations. In this work, on the basis of verifying the accuracy of 2 main kinds of binding free energy calculation methodologies [end-point method of Molecular Mechanics with Poisson-Boltzmann/Generalized Born and Surface Area (MM/PB(GB)SA) and alchemical method of Thermodynamic Integration (TI)], we performed a systematic analysis on the ALK systems to explore the underlying shared and specific drug resistance mechanisms, covering the one-drug-multiple-mutation and multiple-drug-one-mutation cases. Through conventional molecular dynamics (cMD) simulation in conjunction with MM/PB(GB)SA and umbrella sampling (US) in conjunction with contact network analysis (CNA), the resistance mechanisms of the in-pocket, out-pocket, and multiple-site mutations were revealed. Especially for the out-pocket mutation, a possible transfer chain of the mutation effect was revealed, and the reason why different drugs exhibited various sensitivities to the same mutation was also uncovered. The proposed mechanisms may be prevalent in various drug resistance cases.
Collapse
Affiliation(s)
- Yang Yu
- Department of Medicinal Chemistry,
China Pharmaceutical University, Nanjing 210009, Jiangsu, P. R. China
| | - Zhe Wang
- Innovation Institute for Artificial Intelligence in Medicine ofZhejiang University, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, Zhejiang, P. R. China
| | - Lingling Wang
- Department of Medicinal Chemistry,
China Pharmaceutical University, Nanjing 210009, Jiangsu, P. R. China
| | - Qinghua Wang
- Department of Medicinal Chemistry,
China Pharmaceutical University, Nanjing 210009, Jiangsu, P. R. China
| | - Rongfan Tang
- Department of Medicinal Chemistry,
China Pharmaceutical University, Nanjing 210009, Jiangsu, P. R. China
| | - Sutong Xiang
- Department of Medicinal Chemistry,
China Pharmaceutical University, Nanjing 210009, Jiangsu, P. R. China
| | - Qirui Deng
- Department of Medicinal Chemistry,
China Pharmaceutical University, Nanjing 210009, Jiangsu, P. R. China
| | - Tingjun Hou
- Innovation Institute for Artificial Intelligence in Medicine ofZhejiang University, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, Zhejiang, P. R. China
| | - Huiyong Sun
- Department of Medicinal Chemistry,
China Pharmaceutical University, Nanjing 210009, Jiangsu, P. R. China
| |
Collapse
|
4
|
Ando K, Manabe R, Kishino Y, Kusumoto S, Yamaoka T, Tanaka A, Ohmori T, Sagara H. Comparative Efficacy of ALK Inhibitors for Treatment-Naïve ALK-Positive Advanced Non-Small Cell Lung Cancer with Central Nervous System Metastasis: A Network Meta-Analysis. Int J Mol Sci 2023; 24:2242. [PMID: 36768562 PMCID: PMC9917367 DOI: 10.3390/ijms24032242] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 01/18/2023] [Accepted: 01/20/2023] [Indexed: 01/24/2023] Open
Abstract
Central nervous system (CNS) metastases and acquired resistance complicate the treatment of anaplastic lymphoma kinase (ALK) rearrangement-positive (ALK-p) advanced non-small cell lung cancer (NSCLC). Thus, this review aimed to provide a comprehensive overview of brain metastasis, acquired resistance, and prospects for overcoming these challenges. A network meta-analysis of relevant phase III randomized controlled trials was performed to compare the efficacies of multiple ALK inhibitors by drug and generation in overall patients with ALK-p untreated advanced NSCLC and a subgroup of patients with CNS metastases. The primary endpoint was progression-free survival (PFS). Generation-specific comparison results showed that third-generation ALK inhibitors were significantly more effective than second-generation ALK inhibitors in prolonging the PFS of the subgroup of patients with CNS metastases. Drug-specific comparison results demonstrated that lorlatinib was the most effective in prolonging PFS, followed by brigatinib, alectinib, ensartinib, ceritinib, crizotinib, and chemotherapy. While lorlatinib was superior to brigatinib for PFS in the overall patient population, no significant difference between the two was found in the subgroup of patients with CNS metastases. These results can serve as a foundation for basic, clinical, and translational research and guide clinical oncologists in developing individualized treatment strategies for patients with ALK-p, ALK inhibitor-naive advanced NSCLC.
Collapse
Affiliation(s)
- Koichi Ando
- Division of Respirology and Allergology, Department of Medicine, Showa University School of Medicine, 1-5-8 Hatanodai, Shinagawa-ku, Tokyo 142-8666, Japan
- Division of Internal Medicine, Showa University Dental Hospital Medical Clinic, Senzoku Campus, Showa University, 2-1-1 Kita-senzoku, Ohta-ku, Tokyo 145-8515, Japan
| | - Ryo Manabe
- Division of Respirology and Allergology, Department of Medicine, Showa University School of Medicine, 1-5-8 Hatanodai, Shinagawa-ku, Tokyo 142-8666, Japan
| | - Yasunari Kishino
- Division of Respirology and Allergology, Department of Medicine, Showa University School of Medicine, 1-5-8 Hatanodai, Shinagawa-ku, Tokyo 142-8666, Japan
| | - Sojiro Kusumoto
- Division of Respirology and Allergology, Department of Medicine, Showa University School of Medicine, 1-5-8 Hatanodai, Shinagawa-ku, Tokyo 142-8666, Japan
| | - Toshimitsu Yamaoka
- Division of Respirology and Allergology, Department of Medicine, Showa University School of Medicine, 1-5-8 Hatanodai, Shinagawa-ku, Tokyo 142-8666, Japan
- Advanced Cancer Translational Research Institute, Hatanodai Campus, Showa University, 1-5-8 Hatanodai, Shinagawa-ku, Tokyo 142-8555, Japan
| | - Akihiko Tanaka
- Division of Respirology and Allergology, Department of Medicine, Showa University School of Medicine, 1-5-8 Hatanodai, Shinagawa-ku, Tokyo 142-8666, Japan
| | - Tohru Ohmori
- Division of Respirology and Allergology, Department of Medicine, Showa University School of Medicine, 1-5-8 Hatanodai, Shinagawa-ku, Tokyo 142-8666, Japan
- Department of Medicine, Division of Respiratory Medicine, Tokyo Metropolitan Health and Hospitals Corporation, Ebara Hospital, 4-5-10 Higashiyukigaya, Ohta-ku, Tokyo 145-0065, Japan
| | - Hironori Sagara
- Division of Respirology and Allergology, Department of Medicine, Showa University School of Medicine, 1-5-8 Hatanodai, Shinagawa-ku, Tokyo 142-8666, Japan
| |
Collapse
|
5
|
Faria P, Pacheco C, Moura RP, Sarmento B, Martins C. Multifunctional nanomedicine strategies to manage brain diseases. Drug Deliv Transl Res 2022; 13:1322-1342. [PMID: 36344871 DOI: 10.1007/s13346-022-01256-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/30/2022] [Indexed: 11/09/2022]
Abstract
Brain diseases represent a substantial social and economic burden, currently affecting one in six individuals worldwide. Brain research has been focus of great attention in order to unravel the pathogenesis and complexity of brain diseases at the cellular, molecular, and microenvironmental levels. Due to the intrinsic nature of the brain, the presence of the highly restrictive blood-brain barrier (BBB), and the pathophysiology of most diseases, therapies can hardly be considered successful purely by the administration of one drug to a patient. Apart from improving pharmacokinetic parameters, tailoring biodistribution, and reducing the number of side effects, nanomedicines are able to actively co-target the therapeutics to the brain parenchyma and brain lesions, as well as to achieve the delivery of multiple cargos with therapeutic, diagnostic, and theranostic properties. Among other multivalent effects that can be personalized according to the disease needs, this represents a promising class of novel nanosystems, termed multifunctional nanomedicines. Herein, we review the principal mechanisms of therapeutic resistance of the most prevalent brain diseases, how to overcome this therapeutic resistance through the use of multifunctional nanomedicines that tackle multiple fronts of the disease microenvironment, and the promising therapeutic responses achieved by some of the most cutting-edge multifunctional nanomedicines reported in literature.
Collapse
|
6
|
Anti-Angiogenic Therapy in ALK Rearranged Non-Small Cell Lung Cancer (NSCLC). Int J Mol Sci 2022; 23:ijms23168863. [PMID: 36012123 PMCID: PMC9407780 DOI: 10.3390/ijms23168863] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 08/07/2022] [Accepted: 08/08/2022] [Indexed: 11/17/2022] Open
Abstract
The management of advanced lung cancer has been transformed with the identification of targetable oncogenic driver alterations. This includes anaplastic lymphoma kinase (ALK) gene rearrangements. ALK tyrosine kinase inhibitors (TKI) are established first-line treatment options in advanced ALK rearranged non-small cell lung cancer (NSCLC), with several next-generation ALK TKIs (alectinib, brigatinib, ensartinib and lorlatinib) demonstrating survival benefit compared with the first-generation ALK TKI crizotinib. Still, despite high objective response rates and durable progression-free survival, drug resistance inevitably ensues, and treatment options beyond ALK TKI are predominantly limited to cytotoxic chemotherapy. Anti-angiogenic therapy targeting the vascular endothelial growth factor (VEGF) signaling pathway has shown efficacy in combination with platinum-doublet chemotherapy in advanced NSCLC without a driver alteration, and with EGFR TKI in advanced EGFR mutated NSCLC. The role for anti-angiogenic therapy in ALK rearranged NSCLC, however, remains to be elucidated. This review will discuss the pre-clinical rationale, clinical trial evidence to date, and future directions to evaluate anti-angiogenic therapy in ALK rearranged NSCLC.
Collapse
|
7
|
Murciano-Goroff YR, Harada G, Drilon A. An Ascendant Challenge: Central Nervous System Metastases in ALK+ Lung Cancers. Clin Cancer Res 2022; 28:2477-2479. [PMID: 35394532 PMCID: PMC9197849 DOI: 10.1158/1078-0432.ccr-22-0341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 03/17/2022] [Accepted: 03/24/2022] [Indexed: 12/15/2022]
Abstract
Central nervous system (CNS) metastases constitute a challenge for the design of anaplastic lymphoma kinase (ALK) fusion-positive lung cancer trials. The ASCEND-7 study of ceritinib demonstrates the feasibility of broadening CNS eligibility criteria to include symptomatic brain and leptomeningeal disease and highlights design features that contemporary trials will need to incorporate. See related article by Chow et al., p. 2506.
Collapse
Affiliation(s)
| | - Guilherme Harada
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Alexander Drilon
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY,Weill Cornell Medical College, New York, NY
| |
Collapse
|
8
|
P-glycoprotein Mediates Resistance to the Anaplastic Lymphoma Kinase Inhiitor Ensartinib in Cancer Cells. Cancers (Basel) 2022; 14:cancers14092341. [PMID: 35565470 PMCID: PMC9104801 DOI: 10.3390/cancers14092341] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 04/23/2022] [Accepted: 05/05/2022] [Indexed: 01/27/2023] Open
Abstract
Ensartinib (X-396) is a promising second-generation small-molecule inhibitor of anaplastic lymphoma kinase (ALK) that was developed for the treatment of ALK-positive non-small-cell lung cancer. Preclinical and clinical trial results for ensartinib showed superior efficacy and a favorable safety profile compared to the first-generation ALK inhibitors that have been approved by the U.S. Food and Drug Administration. Although the potential mechanisms of acquired resistance to ensartinib have not been reported, the inevitable emergence of resistance to ensartinib may limit its therapeutic application in cancer. In this work, we investigated the interaction of ensartinib with P-glycoprotein (P-gp) and ABCG2, two ATP-binding cassette (ABC) multidrug efflux transporters that are commonly associated with the development of multidrug resistance in cancer cells. Our results revealed that P-gp overexpression, but not expression of ABCG2, was associated with reduced cancer cell susceptibility to ensartinib. P-gp directly decreased the intracellular accumulation of ensartinib, and consequently reduced apoptosis and cytotoxicity induced by this drug. The cytotoxicity of ensartinib could be significantly reversed by treatment with the P-gp inhibitor tariquidar. In conclusion, we report that ensartinib is a substrate of P-gp, and provide evidence that this transporter plays a role in the development of ensartinib resistance. Further investigation is needed.
Collapse
|
9
|
Song YK, Kim MJ, Kim MS, Lee JH, Chung SJ, Song JS, Chae YJ, Lee KR. Role of the Efflux Transporters Abcb1 and Abcg2 in the Brain Distribution of Olaparib in Mice. Eur J Pharm Sci 2022; 173:106177. [PMID: 35341895 DOI: 10.1016/j.ejps.2022.106177] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 03/18/2022] [Accepted: 03/23/2022] [Indexed: 11/18/2022]
Abstract
Olaparib is a first-in-class poly (ADP-ribose) polymerase oral inhibitor used to treat various tumors. In this study, we clarified the roles of ABCB1/Abcb1 and ABCG2/Abcg2 transporters in restricting olaparib distribution to the brain. Olaparib was efficiently transported by human ABCG2, human ABCB1, and mouse Abcg2 in vitro. In the in vivo disposition study of olaparib using single or combination knockout mice, the systemic exposure of olaparib did not differ significantly between the strains over an 8-h period. However, the brain-to-plasma unbound concentration ratio of olaparib increased 5.6- and 8.1-fold in Abcb1a/1b and Abcb1a/1b;Abcg2 knockout mice, respectively, compared with wild-type mice. The Abcg2 single knockout mice exhibited a similar brain-to-plasma unbound concentration ratio to wild-type mice. Moreover, the brain distribution of olaparib could be modulated by the ABCB1/ABCG2 dual inhibitor elacridar to reach a similar degree of inhibition to Abcb1a/1b-/-. These findings suggest that olaparib is actively transported by both human and mouse ABCB1/Abcb1 and ABCG2/Abcg2; while Abcb1a/1b is a major determinant of olaparib brain penetration in mice, Abcg2 is likely to be a minor contributor. Concomitant treatment with temozolomide slightly increased the brain distribution of olaparib in mouse, but the clinical impact of the interaction was expected to be limited.
Collapse
Affiliation(s)
- Yoo-Kyung Song
- Laboratory Animal Resource Center, Korea Research Institute of Bioscience and Biotechnology, Ochang 28116, Republic of Korea
| | - Min-Ju Kim
- Laboratory Animal Resource Center, Korea Research Institute of Bioscience and Biotechnology, Ochang 28116, Republic of Korea
| | - Min-Soo Kim
- College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea
| | - Ji Hun Lee
- Convergence Drug Research Center, Korea Research Institute of Chemical Technology, Daejeon 34114, Republic of Korea
| | - Suk-Jae Chung
- College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea; Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, Republic of Korea
| | - Jin Sook Song
- Convergence Drug Research Center, Korea Research Institute of Chemical Technology, Daejeon 34114, Republic of Korea
| | - Yoon-Jee Chae
- College of Pharmacy, Woosuk University, Wanju-Gun 55338, Republic of Korea.
| | - Kyeong-Ryoon Lee
- Laboratory Animal Resource Center, Korea Research Institute of Bioscience and Biotechnology, Ochang 28116, Republic of Korea; Department of Bioscience, University of Science and Technology, Daejeon 34113, Republic of Korea.
| |
Collapse
|
10
|
Yin Q, Li P, Wang P, Zhang Z, Liu Q, Sun Z, Li W, Ma L, Wang X. Alectinib Together with Intracranial Therapies Improved Survival Outcomes in Untreated ALK-Positive Patients with Non-Small-Cell Lung Cancer and Symptomatic and Synchronic Brain Metastases: A Retrospective Study. Onco Targets Ther 2022; 14:5533-5542. [PMID: 35002258 PMCID: PMC8722687 DOI: 10.2147/ott.s345439] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Accepted: 12/17/2021] [Indexed: 12/23/2022] Open
Abstract
Purpose The performance of alectinib and crizotinib in untreated anaplastic lymphoma kinase (ALK)-positive patients with non-small-cell lung cancer (NSCLC) and symptomatic and synchronic brain metastases is largely unknown. This retrospective study assessed the effectiveness of alectinib and crizotinib, together with intracranial therapies in a cohort of these patients. Patients and Methods This study included 34 previously untreated ALK-positive NSCLC patients with three or fewer intracranial metastases. Of these patients, 13 received oral alectinib 600 mg twice daily, and 21 received oral crizotinib 250 mg twice daily, until progressive disease, unacceptable toxicity, or death. All intracranial metastases were treated with craniotomy, CyberKnife, or both. Results Median overall progression-free survival (PFS) was 32.8 months (95% CI 24.4–41.2 months) in patients treated with alectinib and 8.0 months (95% CI 7.3–8.7 months) in patients treated with crizotinib. Median PFS of brain lesions was not yet reached with alectinib (95% CI 30.1 months–not estimated) and was 8.5 months (95% CI 7.2–12.3 months) with crizotinib. Median PFS of lung lesions was 38.5 months (95% CI 27.5–49.5 months) with alectinib and 9.2 months (95% CI 7.4–11.0 months) with crizotinib. Median overall survival was not yet reached with alectinib (95% CI 31.0 months–not estimated) and 30.3 months (95% CI 27.3–37.1 months) with crizotinib. Conclusion Compared with crizotinib, alectinib showed superior efficacy and lower toxicity in the treatment of ALK-positive patients with NSCLC and symptomatic and synchronic brain metastases. The inclusion of intracranial therapies such as craniotomy or CyberKnife further improved the brain PFS and overall survival of these patients.
Collapse
Affiliation(s)
- Qiang Yin
- Department of Neurosurgery, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, People's Republic of China
| | - Peng Li
- Department of Neurosurgery, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, People's Republic of China
| | - Peng Wang
- Department of Neurosurgery, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, People's Republic of China
| | - Zhen Zhang
- Department of Neurosurgery, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, People's Republic of China
| | - Qun Liu
- Department of Neurosurgery, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, People's Republic of China
| | - Zengfeng Sun
- Department of Neurosurgery, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, People's Republic of China
| | - Wenliang Li
- Department of Neurosurgery, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, People's Republic of China
| | - Li Ma
- Department of Neurosurgery, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, People's Republic of China
| | - Xiaoguang Wang
- Department of Neurosurgery, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, People's Republic of China
| |
Collapse
|
11
|
Salgia SK, Govindarajan A, Salgia R, Pal SK. ALK-Directed Therapy in Non-NSCLC Malignancies: Are We Ready? JCO Precis Oncol 2022; 5:767-770. [PMID: 34994610 DOI: 10.1200/po.21.00078] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Affiliation(s)
- Sabrina K Salgia
- Department of Medical Oncology and Experimental Therapeutics, City of Hope Comprehensive Cancer Center, Duarte, CA
| | - Ameish Govindarajan
- Department of Medical Oncology and Experimental Therapeutics, City of Hope Comprehensive Cancer Center, Duarte, CA
| | - Ravi Salgia
- Department of Medical Oncology and Experimental Therapeutics, City of Hope Comprehensive Cancer Center, Duarte, CA
| | - Sumanta K Pal
- Department of Medical Oncology and Experimental Therapeutics, City of Hope Comprehensive Cancer Center, Duarte, CA
| |
Collapse
|
12
|
Goebel J, Chmielewski J, Hrycyna CA. The roles of the human ATP-binding cassette transporters P-glycoprotein and ABCG2 in multidrug resistance in cancer and at endogenous sites: future opportunities for structure-based drug design of inhibitors. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2022; 4:784-804. [PMID: 34993424 PMCID: PMC8730335 DOI: 10.20517/cdr.2021.19] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The ATP-binding cassette (ABC) transporters P-glycoprotein (P-gp) and ABCG2 are multidrug transporters that confer drug resistance to numerous anti-cancer therapeutics in cell culture. These findings initially created great excitement in the medical oncology community, as inhibitors of these transporters held the promise of overcoming clinical multidrug resistance in cancer patients. However, clinical trials of P-gp and ABCG2 inhibitors in combination with cancer chemotherapeutics have not been successful due, in part, to flawed clinical trial designs resulting from an incomplete molecular understanding of the multifactorial basis of multidrug resistance (MDR) in the cancers examined. The field was also stymied by the lack of high-resolution structural information for P-gp and ABCG2 for use in the rational structure-based drug design of inhibitors. Recent advances in structural biology have led to numerous structures of both ABCG2 and P-gp that elucidated more clearly the mechanism of transport and the polyspecific nature of their substrate and inhibitor binding sites. These data should prove useful helpful for developing even more potent and specific inhibitors of both transporters. As such, although possible pharmacokinetic interactions would need to be evaluated, these inhibitors may show greater effectiveness in overcoming ABC-dependent multidrug resistance in combination with chemotherapeutics in carefully selected subsets of cancers. Another perhaps even more compelling use of these inhibitors may be in reversibly inhibiting endogenously expressed P-gp and ABCG2, which serve a protective role at various blood-tissue barriers. Inhibition of these transporters at sanctuary sites such as the brain and gut could lead to increased penetration by chemotherapeutics used to treat brain cancers or other brain disorders and increased oral bioavailability of these agents, respectively.
Collapse
Affiliation(s)
- Jason Goebel
- Department of Chemistry, Purdue University West Lafayette, IN 47907, USA
| | - Jean Chmielewski
- Department of Chemistry, Purdue University West Lafayette, IN 47907, USA
| | | |
Collapse
|
13
|
Gristina V, La Mantia M, Iacono F, Galvano A, Russo A, Bazan V. The Emerging Therapeutic Landscape of ALK Inhibitors in Non-Small Cell Lung Cancer. Pharmaceuticals (Basel) 2020; 13:E474. [PMID: 33352844 PMCID: PMC7766858 DOI: 10.3390/ph13120474] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 12/03/2020] [Accepted: 12/16/2020] [Indexed: 12/11/2022] Open
Abstract
The treatment of metastatic non-small cell lung cancer (NSCLC) has undergone a paradigm shift over the last decade. Better molecular characterization of the disease has led to the rapid improvement of personalized medicine and the prompt delivery of targeted therapies to patients with NSCLC. The discovery of the EML4-ALK fusion gene in a limited subset of patients affected by NSCLC and the subsequent clinical development of crizotinib in 2011 has been an impressive milestone in lung cancer research. Unfortunately, acquired resistances regularly develop, hence disease progression occurs. Afterward, modern tyrosine kinase inhibitors (TKIs), such as ceritinib, alectinib, brigatinib, and lorlatinib, have been approved by the Food and Drug Administration (FDA) for the management of anaplastic lymphoma kinase (ALK)-positive NSCLCs. Several compounds are currently under investigation to achieve the optimal strategy of therapy. Additionally, the results of ongoing clinical trials with novel-generation TKI will provide more evidence on the best sequence in the treatment of ALK-positive NSCLC patients. In this review, we provide a comprehensive overview of the state-of-the-art targeted therapy options in ALK-positive NSCLCs. Resistance, potential therapeutic strategies to overcome drug resistance, and future perspectives for this subset of patients are critically analyzed and summarized.
Collapse
Affiliation(s)
- Valerio Gristina
- Department of Surgical, Oncological and Oral Sciences, University of Palermo, 90127 Palermo, Italy; (V.G.); (M.L.M.); (F.I.); (A.G.); (A.R.)
| | - Maria La Mantia
- Department of Surgical, Oncological and Oral Sciences, University of Palermo, 90127 Palermo, Italy; (V.G.); (M.L.M.); (F.I.); (A.G.); (A.R.)
| | - Federica Iacono
- Department of Surgical, Oncological and Oral Sciences, University of Palermo, 90127 Palermo, Italy; (V.G.); (M.L.M.); (F.I.); (A.G.); (A.R.)
| | - Antonio Galvano
- Department of Surgical, Oncological and Oral Sciences, University of Palermo, 90127 Palermo, Italy; (V.G.); (M.L.M.); (F.I.); (A.G.); (A.R.)
| | - Antonio Russo
- Department of Surgical, Oncological and Oral Sciences, University of Palermo, 90127 Palermo, Italy; (V.G.); (M.L.M.); (F.I.); (A.G.); (A.R.)
| | - Viviana Bazan
- Department of Biomedicine, Neuroscience and Advanced Diagnostics (Bi.N.D.), Section of Medical Oncology, University of Palermo, 90127 Palermo, Italy
| |
Collapse
|
14
|
Ogihara T, Mizoi K, Kamioka H, Yano K. Physiological Roles of ERM Proteins and Transcriptional Regulators in Supporting Membrane Expression of Efflux Transporters as Factors of Drug Resistance in Cancer. Cancers (Basel) 2020; 12:E3352. [PMID: 33198344 PMCID: PMC7696277 DOI: 10.3390/cancers12113352] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 11/06/2020] [Accepted: 11/10/2020] [Indexed: 12/23/2022] Open
Abstract
One factor contributing to the malignancy of cancer cells is the acquisition of drug resistance during chemotherapy via increased expression of efflux transporters, such as P-glycoprotein (P-gp), multidrug resistance-associated proteins (MRPs), and breast cancer resistance protein (BCRP). These transporters operate at the cell membrane, and are anchored in place by the scaffold proteins ezrin (Ezr), radixin (Rdx), and moesin (Msn) (ERM proteins), which regulate their functional activity. The identity of the regulatory scaffold protein(s) differs depending upon the transporter, and also upon the tissue in which it is expressed, even for the same transporter. Another factor contributing to malignancy is metastatic ability. Epithelial-mesenchymal transition (EMT) is the first step in the conversion of primary epithelial cells into mesenchymal cells that can be transported to other organs via the blood. The SNAI family of transcriptional regulators triggers EMT, and SNAI expression is used is an indicator of malignancy. Furthermore, EMT has been suggested to be involved in drug resistance, since drug excretion from cancer cells is promoted during EMT. We showed recently that ERM proteins are induced by a member of the SNAI family, Snail. Here, we first review recent progress in research on the relationship between efflux transporters and scaffold proteins, including the question of tissue specificity. In the second part, we review the relationship between ERM scaffold proteins and the transcriptional regulatory factors that induce their expression.
Collapse
Affiliation(s)
- Takuo Ogihara
- Graduate School of Pharmaceutical Sciences, Takasaki University of Health and Welfare, 60, Nakaorui-machi, Takasaki, Gunma 370-0033, Japan;
| | - Kenta Mizoi
- Faculty of Pharmacy, Takasaki University of Health and Welfare, 60, Nakaorui-machi, Takasaki, Gunma 370-0033, Japan; (K.M.); (K.Y.)
| | - Hiroki Kamioka
- Graduate School of Pharmaceutical Sciences, Takasaki University of Health and Welfare, 60, Nakaorui-machi, Takasaki, Gunma 370-0033, Japan;
| | - Kentaro Yano
- Faculty of Pharmacy, Takasaki University of Health and Welfare, 60, Nakaorui-machi, Takasaki, Gunma 370-0033, Japan; (K.M.); (K.Y.)
- Laboratory of Drug Metabolism and Pharmacokinetics, Yokohama University of Pharmacy, 601, Matano-cho, Totsuka-ku, Yokohama, Kanagawa 245-0066, Japan
| |
Collapse
|
15
|
Zhao D, Chen J, Chu M, Long X, Wang J. Pharmacokinetic-Based Drug-Drug Interactions with Anaplastic Lymphoma Kinase Inhibitors: A Review. DRUG DESIGN DEVELOPMENT AND THERAPY 2020; 14:1663-1681. [PMID: 32431491 PMCID: PMC7198400 DOI: 10.2147/dddt.s249098] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Accepted: 04/02/2020] [Indexed: 12/21/2022]
Abstract
Anaplastic lymphoma kinase (ALK) inhibitors are important treatment options for non-small-cell lung cancer (NSCLC), associated with ALK gene rearrangement. Patients with ALK gene rearrangement show sensitivity to and benefit clinically from treatment with ALK tyrosine kinase inhibitors (ALK-TKIs). To date, crizotinib, ceritinib, alectinib, brigatinib, lorlatinib, and entrectinib have received approval from the US Food and Drug Administration and/or the European Medicines Agency for use during the treatment of ALK-gene-rearrangement forms of NSCLC. Although the oral route of administration is convenient and results in good compliance among patients, oral administration can be affected by many factors, such as food, intragastric pH, cytochrome P450 enzymes, transporters, and p-glycoprotein. These factors can result in increased risks for serious adverse events or can lead to reduced therapeutic effects of ALK-TKIs. This review characterizes and summarizes the pharmacokinetic parameters and drug–-drug interactions associated with ALK-TKIs to provide specific recommendations for oncologists and clinical pharmacists when prescribing ALK-TKIs.
Collapse
Affiliation(s)
- Dehua Zhao
- Department of Clinical Pharmacy, The Third Hospital of Mianyang (Sichuan Mental Health Center), Mianyang 621000, People's Republic of China
| | - Jing Chen
- Department of Clinical Pharmacy, The Third Hospital of Mianyang (Sichuan Mental Health Center), Mianyang 621000, People's Republic of China
| | - Mingming Chu
- Department of Clinical Pharmacy, The Second Affiliated Hospital of Army Medical University, Chongqing 400037, People's Republic of China
| | - Xiaoqing Long
- Department of Clinical Pharmacy, The Third Hospital of Mianyang (Sichuan Mental Health Center), Mianyang 621000, People's Republic of China
| | - Jisheng Wang
- Department of Clinical Pharmacy, The Third Hospital of Mianyang (Sichuan Mental Health Center), Mianyang 621000, People's Republic of China
| |
Collapse
|
16
|
Gil-Martins E, Barbosa DJ, Silva V, Remião F, Silva R. Dysfunction of ABC transporters at the blood-brain barrier: Role in neurological disorders. Pharmacol Ther 2020; 213:107554. [PMID: 32320731 DOI: 10.1016/j.pharmthera.2020.107554] [Citation(s) in RCA: 81] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Accepted: 04/07/2020] [Indexed: 12/14/2022]
Abstract
ABC (ATP-binding cassette) transporters represent one of the largest and most diverse superfamily of proteins in living species, playing an important role in many biological processes such as cell homeostasis, cell signaling, drug metabolism and nutrient uptake. Moreover, using the energy generated from ATP hydrolysis, they mediate the efflux of endogenous and exogenous substrates from inside the cells, thereby reducing their intracellular accumulation. At present, 48 ABC transporters have been identified in humans, which were classified into 7 different subfamilies (A to G) according to their phylogenetic analysis. Nevertheless, the most studied members with importance in drug therapeutic efficacy and toxicity include P-glycoprotein (P-gp), a member of the ABCB subfamily, the multidrug-associated proteins (MPRs), members of the ABCC subfamily, and breast cancer resistance protein (BCRP), a member of the ABCG subfamily. They exhibit ubiquitous expression throughout the human body, with a special relevance in barrier tissues like the blood-brain barrier (BBB). At this level, they play a physiological function in tissue protection by reducing or limiting the brain accumulation of neurotoxins. Furthermore, dysfunction of ABC transporters, at expression and/or activity level, has been associated with many neurological diseases, including epilepsy, multiple sclerosis, Alzheimer's disease, and amyotrophic lateral sclerosis. Additionally, these transporters are strikingly associated with the pharmacoresistance to central nervous system (CNS) acting drugs, because they contribute to the decrease in drug bioavailability. This article reviews the signaling pathways that regulate the expression and activity of P-gp, BCRP and MRPs subfamilies of transporters, with particular attention at the BBB level, and their mis-regulation in neurological disorders.
Collapse
Affiliation(s)
- Eva Gil-Martins
- UCIBIO-REQUIMTE, Laboratório de Toxicologia, Departamento de Ciências Biológicas, Faculdade de Farmácia, Universidade do Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal
| | - Daniel José Barbosa
- Instituto de Biologia Molecular e Celular (IBMC), Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, 4200-135 Porto, Portugal.
| | - Vera Silva
- UCIBIO-REQUIMTE, Laboratório de Toxicologia, Departamento de Ciências Biológicas, Faculdade de Farmácia, Universidade do Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal
| | - Fernando Remião
- UCIBIO-REQUIMTE, Laboratório de Toxicologia, Departamento de Ciências Biológicas, Faculdade de Farmácia, Universidade do Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal.
| | - Renata Silva
- UCIBIO-REQUIMTE, Laboratório de Toxicologia, Departamento de Ciências Biológicas, Faculdade de Farmácia, Universidade do Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal.
| |
Collapse
|
17
|
Fujita K, Matsumoto N, Ishida H, Kubota Y, Iwai S, Shibanuma M, Kato Y. Decreased Disposition of Anticancer Drugs Predominantly Eliminated via the Liver in Patients with Renal Failure. Curr Drug Metab 2019; 20:361-376. [PMID: 30947665 PMCID: PMC6700602 DOI: 10.2174/1389200220666190402143125] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Revised: 02/27/2019] [Accepted: 03/11/2019] [Indexed: 12/20/2022]
Abstract
Background: Evidence has revealed that renal impairment can affect the systemic exposure of drugs which are predominantly eliminated via the liver. The modulation of drug-metabolizing enzymes and transporters expressed in the liver and/or small intestine by diverse entities, including uremic toxins, in systemic circulation of patients with severe renal failure is considered as the cause of atypical pharmacokinetics, which sometimes induce undesirable adverse events that are especially critical for drugs with narrow therapeutic window such as anticancer drugs. A dosing strategy for anticancer drugs in these patients needs to be established. Methods: The effects of renal impairment on the systemic exposure and safety of anticancer drugs were summarized. The proposed mechanisms for the alterations in the pharmacokinetics of these anticancer drugs were also discussed. Results: Changes in pharmacokinetics and clinical response were reported in 9 out of 10 cytotoxic anticancer drugs investigated, although available information was limited and sometimes controversial. Systemic exposure of 3 out of 16 tyrosine kinase inhibitors was higher in patients with severe renal failure than that in patients with normal kidney function. An increase in systemic exposure of anticancer drugs in patients with renal impairment is likely to be observed for substrates of OATP1B1, despite the limited evidence. Conclusion: The molecular basis for the effect of uremia on non-renal drug elimination still needed to be clarified with further studies to generate generalizable concepts, which may provide insights into establishing better clinical usage of anticancer drugs, i.e. identifying patients at risk and dose adjustment.
Collapse
Affiliation(s)
- Kenichi Fujita
- Division of Cancer Cell Biology, Department of Pharmaceutical Sciences, Showa University School of Pharmacy, 1-5-8 Hatanodai, Shinagawa-ku, Tokyo 1428555, Japan
| | - Natsumi Matsumoto
- Department of Healthcare and Regulatory Sciences, Showa University School of Pharmacy, 1-5-8, Hatanodai, Shinagawa-ku, Tokyo 1428555, Japan.,Breast and Imaging Center, St. Marianna University School of Medicine, 6-7-2 Manpukuji, Asao-ku, Kawasaki 2158520, Japan
| | - Hiroo Ishida
- Department of Medical Oncology, Showa University School of Medicine, 1-5- 8 Hatanodai, Shinagawa-ku, Tokyo 1428555, Japan
| | - Yutaro Kubota
- Department of Medical Oncology, Showa University School of Medicine, 1-5- 8 Hatanodai, Shinagawa-ku, Tokyo 1428555, Japan
| | - Shinichi Iwai
- Department of Healthcare and Regulatory Sciences, Showa University School of Pharmacy, 1-5-8, Hatanodai, Shinagawa-ku, Tokyo 1428555, Japan
| | - Motoko Shibanuma
- Division of Cancer Cell Biology, Department of Pharmaceutical Sciences, Showa University School of Pharmacy, 1-5-8 Hatanodai, Shinagawa-ku, Tokyo 1428555, Japan
| | - Yukio Kato
- Molecular Pharmacotherapeutics, Faculty of Pharmacy, Kanazawa University, Kakuma-machi, Kanazawa 9201192, Japan
| |
Collapse
|
18
|
Strope JD, Peer CJ, Sissung TM, Hall OM, Huang PA, Harris EM, Gustafson KR, Henrich CJ, Sigano DM, Pauly GT, Schneider JP, Bates SE, Figg WD. Botryllamide G is an ABCG2 inhibitor that improves lapatinib delivery in mouse brain. Cancer Biol Ther 2019; 21:223-230. [PMID: 31709896 PMCID: PMC7012088 DOI: 10.1080/15384047.2019.1683324] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Accepted: 10/06/2019] [Indexed: 01/10/2023] Open
Abstract
Introduction: Transporters comprising the blood-brain barrier complicate delivery of many therapeutics to the central nervous system. The present study ascertained whether the natural product botryllamide G is viable for in vivo inhibition of ABCG2 using lapatinib as a probe for ABCB1 and ABCG2-mediated efflux from the brain. Methods: Wild-type and Mdr1a/Mdr1b (-/-) mice were treated with botryllamide G and lapatinib ("doublet therapy"), and while a separate cohort of wild-type mice was treated with botryllamide, tariquidar and lapatinib ("triplet therapy"). Results: Botryllamide G demonstrates biphasic elimination with a rapid distribution, decreasing below the in vitro IC50 of 6.9 µM within minutes, yet with a relatively slower terminal half-life (4.6 h). In Mdr1a/Mdr1b (-/-) mice, doublet therapy resulted in a significant increase in brain lapatinib AUC at 8 h (2058 h*ng/mL vs 4007 h*ng/mL; P = .031), but not plasma exposure (P = .15). No significant differences were observed after 24 h. Lapatinib brain exposure was greater through 1 h when wild-type mice were administered triplet therapy (298 h*pg/mg vs 120 h*pg/mg; P < .001), but the triplet decreased brain AUC through 24 h vs. mice administered lapatinib alone (2878 h*pg/mg vs 4461hr*ng/mL; P < .001) and did not alter the brain:plasma ratio. Conclusions: In summary, the ABCG2 inhibitor, botryllamide G, increases brain exposure to lapatinib in mice lacking Abcb1, although the combination of botryllamide G and tariquidar increases brain exposure in wild-type mice only briefly (1 h). Additional research is needed to find analogs of this compound that have better pharmacokinetics and pharmacodynamic effects on ABCG2 inhibition.
Collapse
Affiliation(s)
- Jonathan D. Strope
- Molecular Pharmacology Section, National Cancer Institute, Bethesda, MD, USA
| | - Cody J. Peer
- Clinical Pharmacology Program, National Cancer Institute, Bethesda, MD, USA
| | - Tristan M. Sissung
- Clinical Pharmacology Program, National Cancer Institute, Bethesda, MD, USA
| | - O. Morgan Hall
- Clinical Pharmacology Program, National Cancer Institute, Bethesda, MD, USA
| | - Phoebe A. Huang
- Molecular Pharmacology Section, National Cancer Institute, Bethesda, MD, USA
| | - Emily M. Harris
- Molecular Pharmacology Section, National Cancer Institute, Bethesda, MD, USA
| | - Kirk R. Gustafson
- Molecular Targets Program, National Cancer Institute, Frederick, MD, USA
| | - Curtis J. Henrich
- Molecular Targets Program, National Cancer Institute, Frederick, MD, USA
- Basic Research Program, Leidos Biomedical Research Inc., Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Dina M. Sigano
- Chemical Biology Laboratory, National Cancer Institute, Frederick, MD, USA
| | - Gary T. Pauly
- Chemical Biology Laboratory, National Cancer Institute, Frederick, MD, USA
| | - Joel P. Schneider
- Chemical Biology Laboratory, National Cancer Institute, Frederick, MD, USA
| | - Susan E. Bates
- Department of Medicine, Division of Hematology and Oncology, Columbia University, New York, NY, USA
| | - William D. Figg
- Molecular Pharmacology Section, National Cancer Institute, Bethesda, MD, USA
- Clinical Pharmacology Program, National Cancer Institute, Bethesda, MD, USA
| |
Collapse
|
19
|
Bai Z, Liu X, Guan Q, Ding N, Wei Q, Tong B, Zhao M, Zhang W, Ma L. 5-(3,4,5-trimethoxybenzoyl)-4-methyl-2-(p-tolyl) imidazol (BZML) targets tubulin and DNA to induce anticancer activity and overcome multidrug resistance in colorectal cancer cells. Chem Biol Interact 2019; 315:108886. [PMID: 31682804 DOI: 10.1016/j.cbi.2019.108886] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Accepted: 10/27/2019] [Indexed: 12/13/2022]
Abstract
Colorectal cancer (CRC) is one of the most common malignancies, and multidrug resistance (MDR) reduces the efficiency of anticancer drugs. Therefore, the development of novel anticancer drugs that are highly active against CRC with MDR is urgently needed. Our previous study showed that 5-(3,4,5-trimethoxybenzoyl)-4-methyl-2-(p-tolyl) imidazol (BZML) is not a P-glycoprotein (P-gp) substrate and has a potent anticancer effect against paclitaxel -sensitive or -resistant non-small-cell lung cancer (NSCLC) in vitro and in vivo. In the present study, we found that BZML exhibited strong anticancer activity not only in sensitive CRC cells (SW480 and HCT-116 cells) but also in intrinsically drug-resistant CRC cells (Caco2 cells). In addition, by targeting the colchicine binding site, BZML inhibited tubulin polymerization, which induced G2/M phase arrest, and it caused DNA damage by directly targeting DNA or producing ROS. Further, BZML induced apoptosis through the time-dependent ROS-mediated mitochondrial apoptotic pathway in the CRC cells. Additionally, BZML inhibited P-gp-mediated drug efflux and enhanced the inhibition of the cell growth that had been induced by paclitaxel or doxorubicin in Caco2 cells. In summary, BZML is a multi-targeted anticancer drug that targets tubulin and DNA, and the mechanisms underlying its potent anticancer activity involve disrupting microtubule assembly, causing DNA damage, inducing cell cycle arrest and eventually activating the ROS-mediated mitochondrial apoptotic pathway in SW480, HCT-116 and Caco2 cells. Therefore, the novel compound BZML is a promising anticancer drug that has tremendous potential for CRC treatment, especially for the treatment of drug-resistant CRC.
Collapse
Affiliation(s)
- Zhaoshi Bai
- Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & the Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, 210009, Jiangsu, China.
| | - Xiaolin Liu
- Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & the Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, 210009, Jiangsu, China
| | - Qi Guan
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang, 110016, China
| | - Nianyang Ding
- Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & the Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, 210009, Jiangsu, China
| | - Qing Wei
- Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & the Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, 210009, Jiangsu, China
| | - Bending Tong
- Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & the Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, 210009, Jiangsu, China
| | - Min Zhao
- Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & the Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, 210009, Jiangsu, China
| | - Weige Zhang
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang, 110016, China
| | - Lingman Ma
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, 210009, Jiangsu, China
| |
Collapse
|
20
|
Pinsolle J, McLeer-Florin A, Giaj Levra M, de Fraipont F, Emprou C, Gobbini E, Toffart AC. Translating Systems Medicine Into Clinical Practice: Examples From Pulmonary Medicine With Genetic Disorders, Infections, Inflammations, Cancer Genesis, and Treatment Implication of Molecular Alterations in Non-small-cell Lung Cancers and Personalized Medicine. Front Med (Lausanne) 2019; 6:233. [PMID: 31737634 PMCID: PMC6828737 DOI: 10.3389/fmed.2019.00233] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Accepted: 10/03/2019] [Indexed: 12/30/2022] Open
Abstract
Non-small-cell lung cancers (NSCLC) represent 85% of all lung cancers, with adenocarcinoma as the most common subtype. Since the 2000's, the discovery of molecular alterations including epidermal growth factor receptor (EGFR) mutations and anaplastic lymphoma kinase (ALK) rearrangements together with the development of specific tyrosine kinase inhibitors (TKIs) has facilitated the development of personalized medicine in the management of this disease. This review focuses on the biology of molecular alterations in NSCLC as well as the diagnostic tools and therapeutic alternatives available for each targetable alteration. Rapid and sensitive methods are essential to detect gene alterations, using tumor tissue biopsies or liquid biopsies. Massive parallel sequencing or Next Generation Sequencing (NGS) allows to simultaneously analyze numerous genes from relatively low amounts of DNA. The detection of oncogenic fusions can be conducted using fluorescence in situ hybridization, reverse-transcription polymerase chain reaction, immunohistochemistry, or NGS. EGFR mutations, ALK and ROS1 rearrangements, MET (MET proto-oncogenereceptor tyrosine kinase), BRAF (B-Raf proto-oncogen serine/threonine kinase), NTRK (neurotrophic tropomyosin receptor kinase), and RET (ret proto-oncogene) alterations are described with their respective TKIs, either already authorized or still in development. We have herein paid particular attention to the mechanisms of resistance to EGFR and ALK-TKI. As a wealth of diagnostic tools and personalized treatments are currently under development, a close collaboration between molecular biologists, pathologists, and oncologists is crucial.
Collapse
Affiliation(s)
- Julian Pinsolle
- Department of Pneumology, CHU Grenoble Alpes, Grenoble, France
- Medicine Faculty, Université Grenoble Alpes, Grenoble, France
| | - Anne McLeer-Florin
- Medicine Faculty, Université Grenoble Alpes, Grenoble, France
- Departement of Pathological Anatomy and Cytology, Pôle de Biologie et Pathologie, CHU Grenoble Alpes, Grenoble, France
- UGA/INSERM U1209/CNRS 5309-Institute for Advanced Biosciences - Université Grenoble Alpes, Grenoble, France
| | - Matteo Giaj Levra
- Department of Pneumology, CHU Grenoble Alpes, Grenoble, France
- Department of Biochemistry, Molecular Biology and Environmental Toxicology, CHU Grenoble Alpes, Grenoble, France
| | - Florence de Fraipont
- UGA/INSERM U1209/CNRS 5309-Institute for Advanced Biosciences - Université Grenoble Alpes, Grenoble, France
- Department of Biochemistry, Molecular Biology and Environmental Toxicology, CHU Grenoble Alpes, Grenoble, France
| | - Camille Emprou
- Medicine Faculty, Université Grenoble Alpes, Grenoble, France
- Departement of Pathological Anatomy and Cytology, Pôle de Biologie et Pathologie, CHU Grenoble Alpes, Grenoble, France
| | - Elisa Gobbini
- Department of Pneumology, CHU Grenoble Alpes, Grenoble, France
- Cancer Research Center Lyon, Centre Léon Bérard, Lyon, France
| | - Anne-Claire Toffart
- Department of Pneumology, CHU Grenoble Alpes, Grenoble, France
- Medicine Faculty, Université Grenoble Alpes, Grenoble, France
- UGA/INSERM U1209/CNRS 5309-Institute for Advanced Biosciences - Université Grenoble Alpes, Grenoble, France
| |
Collapse
|
21
|
Huang SH, Huang ACC, Wang CC, Chang WC, Liu CY, Pavlidis S, Ko HW, Chung FT, Hsu PC, Guo YK, Kuo CHS, Yang CT. Front-line treatment of ceritinib improves efficacy over crizotinib for Asian patients with anaplastic lymphoma kinase fusion NSCLC: The role of systemic progression control. Thorac Cancer 2019; 10:2274-2281. [PMID: 31613427 PMCID: PMC6885427 DOI: 10.1111/1759-7714.13221] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Accepted: 09/26/2019] [Indexed: 12/25/2022] Open
Abstract
Background Approximately 3%–5% of lung adenocarcinoma is driven by anaplastic lymphoma kinase (ALK) fusion oncogene, whose activity can be suppressed by multiple ALK inhibitors. Crizotinib and ceritinib have demonstrated superior efficacy to platinum‐based chemotherapy as front‐line treatment for patients with ALK‐positive advanced non‐small cell lung cancer (NSCLC). However, the direct comparison between them in the front‐line setting remains lacking. Methods A total of 48 patients with ALK‐positive, previously untreated advanced NSCLC, who received crizotinib and ceritinib as front‐line treatment were retrospectively investigated. The efficacy and pattern of disease progression were analyzed. Results Patients receiving ceritinib treatment were significantly younger than those receiving crizotinib treatment (52.0 vs. 63.0, P = 0.016). The median progression‐free survival (PFS) was significantly longer with ceritinib than with crizotinib treatment (32.3 vs. 12.9 months; log‐rank P = 0.020); the hazard ratio for disease progression or death, 0.27 (95% CI, 0.08–0.90; P = 0.033). An objective response was noted in all patients in the ceritinib group and in 23 patients in the crizotinib group (74.2%; 95% CI, 59.0 to 88.5). The rate of systemic progression was significantly lower over time with ceritinib treatment compared to crizotinib treatment (cause‐specific hazard ratio, 0.21; 95% CI 0.06–0.73; P = 0.014). Serious adverse events were noted in one (2.9%) patient showing elevated liver function in the crizotinib group and three (23.1%) patients showing diarrhea in the ceritinib group. Dose reduction was needed in five out of 13 (38.5%) patients receiving ceritinib treatment. Conclusion Ceritinib showed higher efficacy associated with a better control of systemic progression compared to crizotinib for the front‐line treatment of ALK‐positive advanced NSCLCs.
Collapse
Affiliation(s)
- Shih-Hao Huang
- Division of Thoracic Oncology, Department of Thoracic Medicine, Chang Gung Memorial Hospital, Chang Gung University, College of Medicine, Taipei, Taiwan
| | - Allen Chung-Cheng Huang
- Division of Thoracic Oncology, Department of Thoracic Medicine, Chang Gung Memorial Hospital, Chang Gung University, College of Medicine, Taipei, Taiwan
| | - Chin-Chou Wang
- Division of Pulmonary & Critical Care Medicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan
| | - Wen-Chen Chang
- Department of Medical Oncology, Chang Gung Memorial Hospital, Chang Gung University, Taipei, Taiwan
| | - Chien-Ying Liu
- Division of Thoracic Oncology, Department of Thoracic Medicine, Chang Gung Memorial Hospital, Chang Gung University, College of Medicine, Taipei, Taiwan
| | - Stelios Pavlidis
- Data Science Institute, Department of Computing, Imperial College London, London, UK
| | - Ho-Wen Ko
- Division of Thoracic Oncology, Department of Thoracic Medicine, Chang Gung Memorial Hospital, Chang Gung University, College of Medicine, Taipei, Taiwan
| | - Fu-Tsai Chung
- Division of Thoracic Oncology, Department of Thoracic Medicine, Chang Gung Memorial Hospital, Chang Gung University, College of Medicine, Taipei, Taiwan
| | - Ping-Chih Hsu
- Division of Thoracic Oncology, Department of Thoracic Medicine, Chang Gung Memorial Hospital, Chang Gung University, College of Medicine, Taipei, Taiwan
| | - Yi-Ke Guo
- Data Science Institute, Department of Computing, Imperial College London, London, UK
| | - Chih-Hsi Scott Kuo
- Division of Thoracic Oncology, Department of Thoracic Medicine, Chang Gung Memorial Hospital, Chang Gung University, College of Medicine, Taipei, Taiwan.,Data Science Institute, Department of Computing, Imperial College London, London, UK
| | - Cheng-Ta Yang
- Division of Thoracic Oncology, Department of Thoracic Medicine, Chang Gung Memorial Hospital, Chang Gung University, College of Medicine, Taipei, Taiwan
| |
Collapse
|
22
|
Ceritinib-Induced Regression of an Insulin-Like Growth Factor-Driven Neuroepithelial Brain Tumor. Int J Mol Sci 2019; 20:ijms20174267. [PMID: 31480400 PMCID: PMC6747232 DOI: 10.3390/ijms20174267] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Revised: 08/15/2019] [Accepted: 08/28/2019] [Indexed: 12/25/2022] Open
Abstract
The insulin-like growth factor (IGF) pathway plays an important role in several brain tumor entities. However, the lack of inhibitors crossing the blood–brain barrier remains a significant obstacle for clinical translation. Here, we targeted the IGF pathway using ceritinib, an off-target inhibitor of the IGF1 receptor (IGF1R) and insulin receptor (INSR), in a pediatric patient with an unclassified brain tumor and a notch receptor 1 (NOTCH1) germline mutation. Pathway analysis of the tumor revealed activation of the sonic hedgehog (SHH), the wingless and integrated-1 (WNT), the IGF, and the Notch pathway. The proliferation of the patient tumor cells (225ZL) was inhibited by arsenic trioxide (ATO), which is an inhibitor of the SHH pathway, by linsitinib, which is an inhibitor of IGF1R and INSR, and by ceritinib. 225ZL expressed INSR but not IGF1R at the protein level, and ceritinib blocked the phosphorylation of INSR. Our first personalized treatment included ATO, but because of side effects, we switched to ceritinib. After 46 days, we achieved a concentration of 1.70 µM of ceritinib in the plasma, and after 58 days, MRI confirmed that there was a response to the treatment. Ceritinib accumulated in the tumor at a concentration of 2.72 µM. Our data suggest ceritinib as a promising drug for the treatment of IGF-driven brain tumors.
Collapse
|
23
|
Radaram B, Pisaneschi F, Rao Y, Yang P, Piwnica-Worms D, Alauddin MM. Novel derivatives of anaplastic lymphoma kinase inhibitors: Synthesis, radiolabeling, and preliminary biological studies of fluoroethyl analogues of crizotinib, alectinib, and ceritinib. Eur J Med Chem 2019; 182:111571. [PMID: 31425908 DOI: 10.1016/j.ejmech.2019.111571] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Revised: 07/26/2019] [Accepted: 07/28/2019] [Indexed: 12/13/2022]
Abstract
Anaplastic lymphoma kinase (ALK), an oncogenic receptor tyrosine kinase, is a therapeutic target in various cancers, including non-small cell lung cancer. Although several ALK inhibitors, including crizotinib, ceritinib, and alectinib, are approved for cancer treatment, their long-term benefit is often limited by the cancer's acquisition of resistance owing to secondary point mutations in ALK. Importantly, some ALK inhibitors cannot cross the blood-brain barrier (BBB) and thus have little or no efficacy against brain metastases. The introduction of a lipophilic moiety, such as a fluoroethyl group may improve the drug's BBB penetration. Herein, we report the synthesis of fluoroethyl analogues of crizotinib 1, alectinib 4, and ceritinib 9, and their radiolabeling with 18F for pharmacokinetic studies. The fluoroethyl derivatives and their radioactive analogues were obtained in good yields with high purity and good molar activity. A cytotoxicity screen in ALK-expressing H2228 lung cancer cells showed that the analogues had up to nanomolar potency and the addition of the fluorinated moiety had minimal impact overall on the potency of the original drugs. Positron emission tomography in healthy mice showed that the analogues had enhanced BBB penetration, suggesting that they have therapeutic potential against central nervous system metastases.
Collapse
Affiliation(s)
- Bhasker Radaram
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Federica Pisaneschi
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Yi Rao
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Ping Yang
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - David Piwnica-Worms
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA.
| | - Mian M Alauddin
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA.
| |
Collapse
|
24
|
Lowrence RC, Subramaniapillai SG, Ulaganathan V, Nagarajan S. Tackling drug resistance with efflux pump inhibitors: from bacteria to cancerous cells. Crit Rev Microbiol 2019; 45:334-353. [PMID: 31248314 DOI: 10.1080/1040841x.2019.1607248] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Drug resistance is a serious concern in a clinical setting jeopardizing treatment for both infectious agents and cancers alike. The wide-spread emergence of multi-drug resistant (MDR) phenotypes from bacteria to cancerous cells necessitates the need to target resistance mechanisms and prevent the emergence of resistant mutants. Drug efflux seems to be one of the preferred approaches embraced by both microbial and mammalian cells alike, to thwart the action of chemotherapeutic agents thereby leading to a drug resistant phenotype. Relative to microbes, which predominantly employs proton motive force (PMF) powered, Major Facilitator Superfamily (MFS)/Resistance Nodulation and Division (RND) classes of efflux pumps to efflux drugs, cancerous cells preferentially use ATP fuelled ATP binding cassette (ABC) transporters to extrude chemotherapeutic agents. The prevalence, evolutionary characteristics and overlapping functions of ABC transporters have been highlighted in this review. Additionally, we outline the role of ABC pumps in conferring MDR phenotype to both bacteria and cancerous cells and underscore the importance of efflux pump inhibitors (EPI) to mitigate drug resistance. Based on the literature reports and analysis, we reason out feasibility of employing bacteria as a tool to screen for EPI's targeting ABC pumps of cancerous cells.
Collapse
Affiliation(s)
- Rene Christena Lowrence
- a Department of Molecular Biology and Biotechnology, University of Sheffield , Sheffield , UK
| | | | | | - Saisubramanian Nagarajan
- c Department of Biotechnology, School of Chemical and Biotechnology, SASTRA Deemed to be University , Thanjavur , India
| |
Collapse
|
25
|
IGF1R Is a Potential New Therapeutic Target for HGNET-BCOR Brain Tumor Patients. Int J Mol Sci 2019; 20:ijms20123027. [PMID: 31234291 PMCID: PMC6627083 DOI: 10.3390/ijms20123027] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Revised: 05/28/2019] [Accepted: 06/14/2019] [Indexed: 12/24/2022] Open
Abstract
(1) Background: The high-grade neuroepithelial tumor of the central nervous system with BCOR alteration (HGNET-BCOR) is a highly malignant tumor. Preclinical models and molecular targets are urgently required for this cancer. Previous data suggest a potential role of insulin-like growth factor (IGF) signaling in HGNET-BCOR. (2) Methods: The primary HGNET-BCOR cells PhKh1 were characterized by western blot, copy number variation, and methylation analysis and by electron microscopy. The expression of IGF2 and IGF1R was assessed by qRT-PCR. The effect of chemotherapeutics and IGF1R inhibitors on PhKh1 proliferation was tested. The phosphorylation of IGF1R and downstream molecules was assessed by western blot. (3) Results: Phkh1 cells showed a DNA methylation profile compatible with the DNA methylation class "HGNET-BCOR" and morphologic features of cellular cannibalism. IGF2 and IGF1R were highly expressed by three HGNET-BCOR tumor samples and PhKh1 cells. PhKh1 cells were particularly sensitive to vincristine, vinblastine, actinomycin D (IC50 < 10 nM for all drugs), and ceritinib (IC50 = 310 nM). Ceritinib was able to abrogate the proliferation of PhKh1 cells and blocked the phosphorylation of IGF1R and AKT. (4) Conclusion: IGF1R is as an attractive target for the development of new therapy protocols for HGNET-BCOR patients, which may include ceritinib and vinblastine.
Collapse
|
26
|
Chukwueke UN, Brastianos PK. Precision Medical Approaches to the Diagnoses and Management of Brain Metastases. Curr Treat Options Oncol 2019; 20:49. [PMID: 31062107 DOI: 10.1007/s11864-019-0649-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
OPINION STATEMENT Brain metastases represent a common and devastating complication of cancer with survival on the order of a few months in most patients. Melanoma, breast cancer, and lung cancer remain the primary disease histologies with the highest rates of metastatic spread to the brain. The incidence of brain metastases has continued to rise, likely explained by multiple factors. Improvement in progression-free survival in systemic cancer is likely attributable to advances in medical therapy, earlier supportive and symptomatic care, and improved precision around diagnosis and detection. In this context, longer survival and improved extracranial control disease has likely contributed to the increased development of metastatic spread intracranially. The foundation of management remains systemic therapy, as well as a combination of surgery and radiation therapy. In the era of targeted therapies, specific agents have demonstrated improved CNS penetration, however with varying degrees of durable responses. Most patients develop resistance to targeted agents, limiting their duration of use for patients. In this era of personalized medicine, the role of genomic characterization in cancer has been critical in the field of brain metastases, as alterations unique to both the brain metastases and its systemic predecessor have been identified, potentially offering new avenues for therapy.
Collapse
Affiliation(s)
- Ugonma N Chukwueke
- Center for Neuro-Oncology, Department of Medical Oncology, Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, MA, 02215, USA. .,Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, 450 Brookline Avenue, Boston, MA, 02215, USA.
| | - Priscilla K Brastianos
- Department of Medical Oncology, Division of Neuro-Oncology, Massachusetts General Hospital, 55 Fruit Street, Boston, MA, 02114, USA.,Department of Medicine, Harvard Medical School, 55 Fruit Street, Boston, MA, 02114, USA
| |
Collapse
|
27
|
Liu L, Liu X. Contributions of Drug Transporters to Blood-Brain Barriers. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1141:407-466. [PMID: 31571171 DOI: 10.1007/978-981-13-7647-4_9] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Blood-brain interfaces comprise the cerebral microvessel endothelium forming the blood-brain barrier (BBB) and the epithelium of the choroid plexuses forming the blood-cerebrospinal fluid barrier (BCSFB). Their main functions are to impede free diffusion between brain fluids and blood; to provide transport processes for essential nutrients, ions, and metabolic waste products; and to regulate the homeostasis of central nervous system (CNS), all of which are attributed to absent fenestrations, high expression of tight junction proteins at cell-cell contacts, and expression of multiple transporters, receptors, and enzymes. Existence of BBB is an important reason that systemic drug administration is not suitable for the treatment of CNS diseases. Some diseases, such epilepsy, Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis, and diabetes, alter BBB function via affecting tight junction proteins or altering expression and function of these transporters. This chapter will illustrate function of BBB, expression of transporters, as well as their alterations under disease status.
Collapse
Affiliation(s)
- Li Liu
- China Pharmaceutical University, Nanjing, China
| | - Xiaodong Liu
- China Pharmaceutical University, Nanjing, China.
| |
Collapse
|
28
|
P-glycoprotein and breast cancer resistance protein restrict brigatinib brain accumulation and toxicity, and, alongside CYP3A, limit its oral availability. Pharmacol Res 2018; 137:47-55. [PMID: 30253203 DOI: 10.1016/j.phrs.2018.09.020] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2018] [Revised: 09/14/2018] [Accepted: 09/20/2018] [Indexed: 01/13/2023]
Abstract
Brigatinib is an FDA-approved oral anaplastic lymphoma kinase (ALK) inhibitor for treatment of metastatic non-small cell lung cancer (NSCLC). Using genetically modified mouse models, we investigated the roles of the multidrug efflux transporters ABCB1 and ABCG2, and the multispecific drug-metabolizing enzyme CYP3 A in plasma pharmacokinetics and tissue distribution of brigatinib. In vitro, brigatinib was exceptionally well transported by human ABCB1 and mouse Abcg2, and efficiently by human ABCG2. Following oral brigatinib administration (10 mg/kg), brain accumulation was dramatically increased in Abcb1a/1b-/- (19.3-fold) and Abcb1a/1b;Abcg2-/-(41.8-fold), but not in single Abcg2-/- mice compared to wild-type mice. Brigatinib testis accumulation showed qualitatively similar behavior. mAbcb1a/1b and mAbcg2 together restricted systemic exposure of brigatinib: with both systems absent oral availability increased 1.9-fold. Coadministration of elacridar, an ABCB1/ABCG2 inhibitor, caused a pronounced increase (36-fold) in brain-to-plasma ratios of brigatinib, approaching the levels seen in Abcb1a/1b;Abcg2-/- mice. Unexpectedly, lethal toxicity of oral brigatinib was observed in mice with genetic knockout or pharmacological inhibition of mAbcb1a/1b and mAbcg2, indicating a pronounced protective role for these transporters. In Cyp3a-/- mice, brigatinib plasma exposure increased 1.3-fold, and was subsequently 1.8-fold reduced by transgenic overexpression of human CYP3 A4 in liver and intestine. The relative tissue distribution of brigatinib, however, remained unaltered. ABCB1 and ABCG2 thus limit brain accumulation, toxicity, and systemic exposure of brigatinib, whereas CYP3 A also markedly restricts its oral availability. Unexpected toxicities should therefore be carefully monitored when brigatinib is coadministered with ABCB1/ABCG2 inhibitors in patients. Collectively, these insights may support the clinical application of brigatinib.
Collapse
|
29
|
Efficacy of ALK inhibitors on NSCLC brain metastases: A systematic review and pooled analysis of 21 studies. PLoS One 2018; 13:e0201425. [PMID: 30052658 PMCID: PMC6063430 DOI: 10.1371/journal.pone.0201425] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2018] [Accepted: 07/16/2018] [Indexed: 12/21/2022] Open
Abstract
Background Patients with anaplastic lymphoma kinase rearranged (ALK+) non-small cell lung cancer (NSCLC) have a higher risk of developing brain metastases (BMs) than patients with other NSCLC sub-types. ALK inhibitors have activity in BMs due to ALK+ NSCLC. We performed a systematic review of the literature with the aim of assessing the efficacy of ALK inhibitors on BMs. Material and methods A systematic search of the literature was performed using the databases Pubmed, EMBASE, Web of Science, The Cochrane Library, and SCOPUS. Relevant publications reporting activity of ALK inhibitors in NSCLC BMs were retrieved. Data were pooled using the number of events/number of evaluable patients according to fixed or random effect models. Intracranial tumour response was assessed through overall response rate (ORR), disease control rate (DCR: ORR + stable disease rate), median progression-free survival (PFS), and overall survival (OS). The primary endpoint was intracranial overall response rate (IC ORR). Results A total of 1,016 patients with BMs from 21 studies were analysed. In patients receiving ALK inhibitors in the first line setting, the pooled IC ORR was 39.17% (95%CI 13.1–65.2%), while the pooled IC ORR observed in further lines was 44.2% (95%CI 33.3–55.1%). Intracranial disease control rate (IC DCR) was 70.3% and 78.2% in naïve and pre-treated patients, respectively. Patients who had not received brain radiation attained an IC ORR of 49.0%. Conclusions Based on these data, ALK inhibitors are effective in both naive and pre-treated patients with similar IC ORR and IC DCR, irrespective of the line of therapy.
Collapse
|
30
|
Abstract
Leptomeningeal metastasis (LM) results from dissemination of cancer cells to both the leptomeninges (pia and arachnoid) and cerebrospinal fluid (CSF) compartment. Breast cancer, lung cancer, and melanoma are the most common solid tumors that cause LM. Recent approval of more active anticancer therapies has resulted in improvement in survival that is partly responsible for an increased incidence of LM. Neurologic deficits, once manifest, are mostly irreversible, and often have a significant impact on patient quality of life. LM-directed therapy is based on symptom palliation, circumscribed use of neurosurgery, limited field radiotherapy, intra-CSF and systemic therapies. Novel methods of detecting LM include detection of CSF circulating tumor cells and tumor cell-free DNA. A recent international guideline for a standardization of response assessment in LM may improve cross-trial comparisons as well as within-trial evaluation of treatment. An increasing number of retrospective studies suggest that molecular-targeted therapy, such as EGFR and ALK inhibitors in lung cancer, trastuzumab in HER2+ breast cancer, and BRAF inhibitors in melanoma, may be effective as part of the multidisciplinary management of LM. Prospective randomized trials with standardized response assessment are needed to further validate these preliminary findings.
Collapse
|
31
|
Li W, Sparidans RW, Wang Y, Lebre MC, Wagenaar E, Beijnen JH, Schinkel AH. P-glycoprotein (MDR1/ABCB1) restricts brain accumulation and cytochrome P450-3A (CYP3A) limits oral availability of the novel ALK/ROS1 inhibitor lorlatinib. Int J Cancer 2018; 143:2029-2038. [PMID: 29744867 DOI: 10.1002/ijc.31582] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2018] [Revised: 04/07/2018] [Accepted: 04/20/2018] [Indexed: 01/09/2023]
Abstract
Lorlatinib (PF-06463922) is a promising oral anaplastic lymphoma kinase (ALK) and ROS1 inhibitor currently in Phase III clinical trials for treatment of non-small-cell lung cancer (NSCLC) containing an ALK rearrangement. With therapy-resistant brain metastases a major concern in NSCLC, lorlatinib was designed to have high membrane and blood-brain barrier permeability. We investigated the roles of the multidrug efflux transporters ABCB1 and ABCG2, and the multispecific drug-metabolizing enzyme CYP3A in plasma pharmacokinetics and tissue distribution of lorlatinib using genetically modified mouse strains. In vitro, human ABCB1 and mouse Abcg2 modestly transported lorlatinib. Following oral lorlatinib administration (at 10 mg/kg), brain accumulation of lorlatinib, while relatively high in wild-type mice, was still fourfold increased in Abcb1a/1b-/- and Abcb1a/1b;Abcg2-/- mice, but not in single Abcg2-/- mice. Lorlatinib plasma levels were not altered. Oral coadministration of the ABCB1/ABCG2 inhibitor elacridar increased the brain accumulation of lorlatinib in wild-type mice fourfold, that is, to the same level as in Abcb1a/1b;Abcg2-/- mice, without altering plasma exposure. Similar results were obtained for lorlatinib testis accumulation. In Cyp3a-/- mice, the plasma exposure of lorlatinib was increased 1.3-fold, but was then twofold reduced upon transgenic overexpression of human CYP3A4 in liver and intestine, whereas relative tissue distribution of lorlatinib remained unaltered. Our data indicate that lorlatinib brain accumulation is substantially limited by P-glycoprotein/ABCB1 in the blood-brain barrier, but this can be effectively reversed by elacridar coadministration. Moreover, oral availability of lorlatinib is markedly restricted by CYP3A4 activity. These insights may be used in optimizing the therapeutic application of lorlatinib.
Collapse
Affiliation(s)
- Wenlong Li
- Division of Pharmacology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX, Amsterdam, The Netherlands
| | - Rolf W Sparidans
- Faculty of Science, Department of Pharmaceutical Sciences, Division of Pharmacoepidemiology & Clinical Pharmacology, Utrecht University, Universiteitsweg 99, 3584 CG, Utrecht, The Netherlands
| | - Yaogeng Wang
- Division of Pharmacology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX, Amsterdam, The Netherlands
| | - Maria C Lebre
- Division of Pharmacology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX, Amsterdam, The Netherlands
| | - Els Wagenaar
- Division of Pharmacology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX, Amsterdam, The Netherlands
| | - Jos H Beijnen
- Division of Pharmacology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX, Amsterdam, The Netherlands.,Faculty of Science, Department of Pharmaceutical Sciences, Division of Pharmacoepidemiology & Clinical Pharmacology, Utrecht University, Universiteitsweg 99, 3584 CG, Utrecht, The Netherlands.,Department of Pharmacy & Pharmacology, The Netherlands Cancer Institute/Slotervaart Hospital, Plesmanlaan 121, 1066 CX, Amsterdam, The Netherlands
| | - Alfred H Schinkel
- Division of Pharmacology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX, Amsterdam, The Netherlands
| |
Collapse
|
32
|
Robey RW, Pluchino KM, Hall MD, Fojo AT, Bates SE, Gottesman MM. Revisiting the role of ABC transporters in multidrug-resistant cancer. Nat Rev Cancer 2018; 18:452-464. [PMID: 29643473 PMCID: PMC6622180 DOI: 10.1038/s41568-018-0005-8] [Citation(s) in RCA: 1145] [Impact Index Per Article: 190.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Most patients who die of cancer have disseminated disease that has become resistant to multiple therapeutic modalities. Ample evidence suggests that the expression of ATP-binding cassette (ABC) transporters, especially the multidrug resistance protein 1 (MDR1, also known as P-glycoprotein or P-gp), which is encoded by ABC subfamily B member 1 (ABCB1), can confer resistance to cytotoxic and targeted chemotherapy. However, the development of MDR1 as a therapeutic target has been unsuccessful. At the time of its discovery, appropriate tools for the characterization and clinical development of MDR1 as a therapeutic target were lacking. Thirty years after the initial cloning and characterization of MDR1 and the implication of two additional ABC transporters, the multidrug resistance-associated protein 1 (MRP1; encoded by ABCC1)), and ABCG2, in multidrug resistance, interest in investigating these transporters as therapeutic targets has waned. However, with the emergence of new data and advanced techniques, we propose to re-evaluate whether these transporters play a clinical role in multidrug resistance. With this Opinion article, we present recent evidence indicating that it is time to revisit the investigation into the role of ABC transporters in efficient drug delivery in various cancer types and at the blood-brain barrier.
Collapse
Affiliation(s)
- Robert W Robey
- Laboratory of Cell Biology, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Kristen M Pluchino
- Laboratory of Cell Biology, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Matthew D Hall
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, USA
| | - Antonio T Fojo
- Division of Hematology/Oncology, Department of Medicine, Columbia University/New York Presbyterian Hospital, Manhattan, NY, USA
- James J. Peters VA Medical Center, Bronx, NY, USA
| | - Susan E Bates
- Division of Hematology/Oncology, Department of Medicine, Columbia University/New York Presbyterian Hospital, Manhattan, NY, USA
- James J. Peters VA Medical Center, Bronx, NY, USA
| | - Michael M Gottesman
- Laboratory of Cell Biology, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
33
|
Clinical Pharmacokinetics of Anaplastic Lymphoma Kinase Inhibitors in Non-Small-Cell Lung Cancer. Clin Pharmacokinet 2018; 58:403-420. [DOI: 10.1007/s40262-018-0689-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
34
|
Liao BC, Lin CC, Yang JCH. Treating brain metastases in non-small cell lung cancer patients: what have we learnt from pharmaceutical recent clinical trials? Expert Opin Pharmacother 2018; 19:851-864. [PMID: 29726292 DOI: 10.1080/14656566.2018.1472765] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
INTRODUCTION Brain metastases (BMs) develop in up to 40% of patients with non-small cell lung cancer (NSCLC). In many recent practice-changing clinical trials, patients with BM were included; however, only few trials reported intracranial efficacies in either post hoc or pre-planned analysis. Clinically meaningful intracranial efficacy data of novel agents have not been completely disclosed. AREAS COVERED The authors performed a systemic review of recent pharmaceutical clinical trials, mainly pivotal or practice-changing trials. Some of the prospective clinical trials focused on patients with NSCLC and BM. The authors collected and compared intracranial efficacy reports of chemotherapy, epidermal growth factor receptor tyrosine kinase inhibitors (EGFR-TKIs), ALK inhibitors, and immune checkpoint inhibitors. EXPERT OPINION Many clinical trials, especially those on 'brain-active' EGFR-TKIs and ALK inhibitors, have robust reports of intracranial efficacies either as post hoc or pre-planned analysis. Physicians should interpret this data with caution and apply the results to patients accordingly. For the design of future clinical trials, enrolling patients with only BM, incorporating novel risk classifications, pre-planning intracranial efficacy endpoints, reporting prior local brain therapies, and applying novel response evaluation criteria are emerging trends in this area.
Collapse
Affiliation(s)
- Bin-Chi Liao
- a Department of Oncology , National Taiwan University Hospital , Taipei , Taiwan.,b National Taiwan University Cancer Center, College of Medicine, National Taiwan University , Taipei , Taiwan.,c Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University , Taipei , Taiwan
| | - Chia-Chi Lin
- a Department of Oncology , National Taiwan University Hospital , Taipei , Taiwan.,c Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University , Taipei , Taiwan
| | - James Chih-Hsin Yang
- a Department of Oncology , National Taiwan University Hospital , Taipei , Taiwan.,b National Taiwan University Cancer Center, College of Medicine, National Taiwan University , Taipei , Taiwan.,c Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University , Taipei , Taiwan.,d Graduate Institute of Oncology, College of Medicine, National Taiwan University , Taipei , Taiwan
| |
Collapse
|
35
|
Zhang Y, Gong W, Wang Y, Liu Y, Li C. Exploring movement and energy in human P-glycoprotein conformational rearrangement. J Biomol Struct Dyn 2018; 37:1104-1119. [PMID: 29620438 DOI: 10.1080/07391102.2018.1461133] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Human P-glycoprotein (P-gp), a kind of ATP-Binding Cassette transporter, can export a diverse variety of anti-cancer drugs out of the tumor cell. Its overexpression is one of the main reasons for the multidrug resistance (MDR) of tumor cells. It has been confirmed that during the substrate transport process, P-gp experiences a large-scale structural rearrangement from the inward- to outward-facing states. However, the mechanism of how the nucleotide-binding domains (NBDs) control the transmembrane domains (TMDs) to open towards the periplasm in the outward-facing state has not yet been fully characterized. Herein, targeted molecular dynamics simulations were performed to explore the conformational rearrangement of human P-gp. The results show that the allosteric process proceeds in a coupled way, and first the transition is driven by the NBDs, and then transmitted to the cytoplasmic parts of TMDs, finally to the periplasmic parts. The trajectories show that besides the translational motions, the NBDs undergo a rotation movement, which mainly occurs in xy plane and ensures the formation of the correct ATP-binding pockets. The analyses on the interaction energies between the six structure segments (cICLs) from the TMDs and NBDs reveal that their subtle energy differences play an important role in causing the periplasmic parts of the transmembrane helices to separate from each other in the established directions and in appropriate amplitudes. This conclusion can explain the two experimental phenomena about human P-gp in some extent. These studies have provided a detailed exploration into human P-gp rearrangement process and given an energy insight into the TMD reorientation during P-gp transition.
Collapse
Affiliation(s)
- Yue Zhang
- a College of Life Science and Bioengineering , Beijing University of Technology , Beijing , 100124 , China
| | - Weikang Gong
- a College of Life Science and Bioengineering , Beijing University of Technology , Beijing , 100124 , China
| | - Yan Wang
- b Key Laboratory of Molecular Biophysics of the Ministry of Education, School of Life Science and Technology , Huazhong University of Science and Technology , Wuhan , Hubei , 430074 , China
| | - Yang Liu
- a College of Life Science and Bioengineering , Beijing University of Technology , Beijing , 100124 , China
| | - Chunhua Li
- a College of Life Science and Bioengineering , Beijing University of Technology , Beijing , 100124 , China
| |
Collapse
|
36
|
Pellerino A, Bertero L, Rudà R, Soffietti R. Neoplastic meningitis in solid tumors: from diagnosis to personalized treatments. Ther Adv Neurol Disord 2018. [PMID: 29535794 PMCID: PMC5844521 DOI: 10.1177/1756286418759618] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Neoplastic meningitis (NM) is a devastating complication of solid tumors with poor outcome. Some randomized clinical trials have been conducted with heterogeneous inclusion criteria, diagnostic parameters, response evaluation and primary endpoints. Recently, the Leptomeningeal Assessment in Neuro-Oncology (LANO) Group and the European Society for Medical Oncology/European Association for Neuro-Oncology have proposed some recommendations in order to provide diagnostic criteria and response evaluation scores for NM. The aim of these guidelines is to integrate the neurological examination with magnetic resonance imaging and cerebrospinal fluid findings as well as to provide a framework for use in clinical trials. However, this composite assessment needs further validation. Since intrathecal therapy represents a treatment with limited efficacy in NM, many studies have been conducted on systemic therapies, including target therapies, with some encouraging results in terms of disease control. In this review, we have analyzed the clinical aspects and the most recent diagnostic tools and therapeutic options in NM.
Collapse
Affiliation(s)
- Alessia Pellerino
- Department of Neuro-Oncology, University and City of Health and Science Hospital, Via Cherasco 15, Turin, 10126 Italy
| | - Luca Bertero
- Section of Pathology, Department of Medical Sciences, University of Turin, Turin, Italy
| | - Roberta Rudà
- Department of Neuro-Oncology, University and City of Health and Science Hospital, Turin, Italy
| | - Riccardo Soffietti
- Department of Neuro-Oncology, University and City of Health and Science Hospital, Turin, Italy
| |
Collapse
|
37
|
Probable drug-drug interaction between erlotinib and amiodarone causes severe neurotoxicity in a patient with advanced lung cancer. Anticancer Drugs 2018; 29:380-383. [PMID: 29420339 DOI: 10.1097/cad.0000000000000600] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Drug-drug interactions (DDIs) are of great concern in the treatment of cancer, especially when target therapies, such as tyrosine kinase inhibitors, are being used. Here, we report a case of probable DDI between erlotinib and amiodarone leading to severe neurotoxicity. Amiodarone inhibits P-glycoprotein (P-gp), for which erlotinib is a substrate. P-gp is an important drug transporter that is involved in limiting the blood-brain barrier penetration of erlotinib. Clinicians should be aware of emerging data characterizing the effect of the P-gp transport system on drug exposure and its potential for DDI.
Collapse
|
38
|
Kabraji S, Ni J, Lin NU, Xie S, Winer EP, Zhao JJ. Drug Resistance in HER2-Positive Breast Cancer Brain Metastases: Blame the Barrier or the Brain? Clin Cancer Res 2018; 24:1795-1804. [PMID: 29437794 DOI: 10.1158/1078-0432.ccr-17-3351] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2017] [Revised: 01/06/2018] [Accepted: 02/01/2018] [Indexed: 12/11/2022]
Abstract
The brain is the most common site of first metastasis for patients with HER2-positive breast cancer treated with HER2-targeting drugs. However, the development of effective therapies for breast cancer brain metastases (BCBM) is limited by an incomplete understanding of the mechanisms governing drug sensitivity in the central nervous system. Pharmacodynamic data from patients and in vivo models suggest that inadequate drug penetration across the "blood-tumor" barrier is not the whole story. Using HER2-positive BCBMs as a case study, we highlight recent data from orthotopic brain metastasis models that implicate brain-specific drug resistance mechanisms in BCBMs and suggest a translational research paradigm to guide drug development for treatment of BCBMs. Clin Cancer Res; 24(8); 1795-804. ©2018 AACR.
Collapse
Affiliation(s)
- Sheheryar Kabraji
- Department of Medical Oncology, Dana Farber Cancer Institute, Boston, Massachusetts. .,Department of Cancer Biology, Dana Farber Cancer Institute, Boston, Massachusetts
| | - Jing Ni
- Department of Cancer Biology, Dana Farber Cancer Institute, Boston, Massachusetts.,Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts
| | - Nancy U Lin
- Department of Medical Oncology, Dana Farber Cancer Institute, Boston, Massachusetts
| | - Shaozhen Xie
- Department of Cancer Biology, Dana Farber Cancer Institute, Boston, Massachusetts.,Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts
| | - Eric P Winer
- Department of Medical Oncology, Dana Farber Cancer Institute, Boston, Massachusetts
| | - Jean J Zhao
- Department of Cancer Biology, Dana Farber Cancer Institute, Boston, Massachusetts. .,Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
39
|
Berghoff AS, Preusser M. Role of the blood-brain barrier in metastatic disease of the central nervous system. HANDBOOK OF CLINICAL NEUROLOGY 2018; 149:57-66. [PMID: 29307361 DOI: 10.1016/b978-0-12-811161-1.00004-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Systemic therapy is an important backbone in the multimodal treatment approach of brain metastases. However, the blood-brain barrier or, more correctly, the blood-tumor barrier, as the properties of tumor-associated vessels differ from the physiologic state, potentially limits the passage of systemic drugs. Indeed, several preclinical and clinical investigations showed that the distribution of drugs is very heterogeneous within a given brain metastasis, despite the contrast enhancement in magnetic resonance imaging. Brain metastases may show lower intratumoral concentrations of some drugs as compared to extracranial tumor sites, resulting in mixed responses. Therefore, a more profound understanding of the role of the blood-brain/blood-tumor barrier is needed to effectively formulate clinical trial approaches on systemic therapy options in patients with brain metastases.
Collapse
Affiliation(s)
- Anna S Berghoff
- Clinical Division of Oncology, Department of Medicine and CNS Tumors Unit, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Matthias Preusser
- Clinical Division of Oncology, Department of Medicine and CNS Tumors Unit, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
40
|
Abstract
Ceritinib (Zykadia™) is an oral, selective inhibitor of the anaplastic lymphoma kinase (ALK), a receptor tyrosine kinase which, after genetic rearrangement, acts as an oncogenic driver in a proportion of non-small cell lung cancers (NSCLCs). The drug is approved in several countries worldwide for the treatment of patients with ALK-positive, advanced NSCLC who have previously received the first-generation ALK inhibitor crizotinib (indication details may vary by country). Approval was based on its clinical benefit in this setting in the phase I and II trials known as ASCEND-1 and -2. Across these noncomparative studies, 36-56 % of patients achieved a response with ceritinib (at the recommended dosage of 750 mg once daily) and the responses were durable, lasting up to a median of 10 months. Patients survived free from progression for a median of up to 7 months and had a median overall survival of up to 17 months. Moreover, efficacy outcomes in patients with brain metastases were generally consistent with those of the overall study populations. Ceritinib has an acceptable tolerability profile, with gastrointestinal issues, fatigue and liver test abnormalities being the most common adverse reactions. Thus, ceritinib is a valuable treatment option for patients with ALK-positive advanced NSCLC who have already received crizotinib therapy.
Collapse
|
41
|
BCRP/ABCG2 and high-alert medications: Biochemical, pharmacokinetic, pharmacogenetic, and clinical implications. Biochem Pharmacol 2017; 147:201-210. [PMID: 29031817 DOI: 10.1016/j.bcp.2017.10.004] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Accepted: 10/11/2017] [Indexed: 01/14/2023]
Abstract
The human breast cancer resistance protein (BCRP/ABCG2) is an ATP-binding cassette efflux transporter that uses ATP hydrolysis to expel xenobiotics from cells, including anti-cancer medications. It is expressed in the gastrointestinal tract, liver, kidney, and brain endothelium. Thus, ABCG2 functions as a tissue barrier to drug transport that strongly influences the pharmacokinetics of substrate medications. Genetic polymorphisms of ABCG2 are closely related to inter-individual variations in therapeutic performance. The common single nucleotide polymorphism c.421C>A, p.Q141K reduces cell surface expression of ABCG2 protein, resulting in lower efflux of substrates. Consequently, a higher plasma concentration of substrate is observed in patients carrying an ABCG2 c.421C>A allele. Detailed pharmacokinetic analyses have revealed that altered intestinal absorption is responsible for the distinct pharmacokinetics of ABCG2 substrates in genetic carriers of the ABCG2 c.421C>A polymorphism. Recent studies have focused on the high-alert medications among ABCG2 substrates (defined as those with high risk of adverse events), such as tyrosine kinase inhibitors (TKIs) and direct oral anti-coagulants (DOACs). For these high-alert medications, inter-individual variation may be closely related to the severity of side effects. In addition, ethnic differences in the frequency of ABCG2 c.421C>A have been reported, with markedly higher frequency in East Asian (∼30-60%) than Caucasian and African-American populations (∼5-10%). Therefore, ABCG2 polymorphisms must be considered not only in the drug development phase, but also in clinical practice. In the present review, we provide an update of basic and clinical knowledge on genetic polymorphisms of ABCG2.
Collapse
|
42
|
Tomono T, Yano K, Ogihara T. Snail-Induced Epithelial-to-Mesenchymal Transition Enhances P-gp-Mediated Multidrug Resistance in HCC827 Cells. J Pharm Sci 2017; 106:2642-2649. [DOI: 10.1016/j.xphs.2017.03.011] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Revised: 03/03/2017] [Accepted: 03/09/2017] [Indexed: 12/12/2022]
|
43
|
Santarpia M, Daffinà MG, D’Aveni A, Marabello G, Liguori A, Giovannetti E, Karachaliou N, Gonzalez Cao M, Rosell R, Altavilla G. Spotlight on ceritinib in the treatment of ALK+ NSCLC: design, development and place in therapy. Drug Des Devel Ther 2017; 11:2047-2063. [PMID: 28740365 PMCID: PMC5503498 DOI: 10.2147/dddt.s113500] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The identification of echinoderm microtubule-associated protein-like 4 (EML4) and anaplastic lymphoma kinase (ALK) fusion gene in non-small cell lung cancer (NSCLC) has radically changed the treatment of a subset of patients harboring this oncogenic driver. Crizotinib was the first ALK tyrosine kinase inhibitor to receive fast approval and is currently indicated as the first-line therapy for advanced, ALK-positive NSCLC patients. However, despite crizotinib's efficacy, patients almost invariably progress, with the central nervous system being one of the most common sites of relapse. Different mechanisms of acquired resistance have been identified, including secondary ALK mutations, ALK copy number alterations and activation of bypass tracks. Different highly potent and brain-penetrant next-generation ALK inhibitors have been developed and tested in NSCLC patients with ALK rearrangements. Ceritinib, a structurally distinct and selective ALK inhibitor, showed 20 times higher potency than crizotinib in inhibiting ALK and had activity against the most common crizotinib-resistant mutations, including L1196M and G1269A, in preclinical models. In Phase I and II studies, ceritinib demonstrated pronounced activity in both crizotinib-naïve and crizotinib-refractory patients, with responses observed regardless of the presence of ALK resistance mutations. Ceritinib was the first ALK inhibitor to be approved for the treatment of crizotinib-refractory, ALK-rearranged NSCLC, and recent results from a Phase III study have demonstrated superior efficacy compared to standard chemotherapy in the first- and second-line setting. We provide an extensive overview of ceritinib from the design of the compound through preclinical data until efficacy and toxicity results from Phase I-III clinical studies. We review the molecular alterations associated with resistance to ceritinib and highlight the importance of obtaining tumor biopsy at progression to tailor therapy based upon the underlying resistance mechanism. We finally provide an outlook on novel rational therapeutic combinations.
Collapse
Affiliation(s)
- Mariacarmela Santarpia
- Medical Oncology Unit, Department of Human Pathology “G. Barresi”, University of Messina, Messina, Italy
| | - Maria Grazia Daffinà
- Medical Oncology Unit, Department of Human Pathology “G. Barresi”, University of Messina, Messina, Italy
| | - Alessandro D’Aveni
- Medical Oncology Unit, Department of Human Pathology “G. Barresi”, University of Messina, Messina, Italy
| | - Grazia Marabello
- Medical Oncology Unit, Department of Human Pathology “G. Barresi”, University of Messina, Messina, Italy
| | - Alessia Liguori
- Medical Oncology Unit, Department of Human Pathology “G. Barresi”, University of Messina, Messina, Italy
| | - Elisa Giovannetti
- Department of Medical Oncology, VU University Medical Center, Amsterdam, The Netherlands
- Department of Nanoscience and Nanotechnologies, CNR-Nano, Institute of Nanoscience and Nanotechnology
- Cancer Pharmacology Lab, AIRC Start-Up Unit, University of Pisa, Pisa, Italy
| | - Niki Karachaliou
- Institute of Oncology Rosell (IOR), University Hospital Sagrat Cor
| | - Maria Gonzalez Cao
- Oncology Department, Institute of Oncology Rosell (IOR), Quirón-Dexeus University Institute, Barcelona
| | - Rafael Rosell
- Cancer Biology and Precision Medicine Program, Germans Trias i Pujol Research Institute
- Catalan Institute of Oncology, Germans Trias i Pujol University Hospital, Badalona, Spain
| | - Giuseppe Altavilla
- Medical Oncology Unit, Department of Human Pathology “G. Barresi”, University of Messina, Messina, Italy
| |
Collapse
|
44
|
Liao MZ, Gao C, Shireman LM, Phillips B, Risler LJ, Neradugomma NK, Choudhari P, Prasad B, Shen DD, Mao Q. P-gp/ABCB1 exerts differential impacts on brain and fetal exposure to norbuprenorphine. Pharmacol Res 2017; 119:61-71. [PMID: 28111265 PMCID: PMC5392442 DOI: 10.1016/j.phrs.2017.01.018] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2016] [Revised: 01/10/2017] [Accepted: 01/16/2017] [Indexed: 01/11/2023]
Abstract
Norbuprenorphine is the major active metabolite of buprenorphine which is commonly used to treat opiate addiction during pregnancy. Norbuprenorphine produces marked respiratory depression and was 10 times more potent than buprenorphine. Therefore, it is important to understand the mechanism that controls fetal exposure to norbuprenorphine, as exposure to this compound may pose a significant risk to the developing fetus. P-gp/ABCB1 and BCRP/ABCG2 are two major efflux transporters regulating tissue distribution of drugs. Previous studies have shown that norbuprenorphine, but not buprenorphine, is a P-gp substrate. In this study, we systematically examined and compared the roles of P-gp and BCRP in determining maternal brain and fetal distribution of norbuprenorphine using transporter knockout mouse models. We administered 1mg/kg norbuprenorphine by retro-orbital injection to pregnant FVB wild-type, Abcb1a-/-/1b-/-, and Abcb1a-/-/1b-/-/Abcg2-/- mice on gestation day 15. The fetal AUC of norbuprenorphine was ∼64% of the maternal plasma AUC in wild-type mice, suggesting substantial fetal exposure to norbuprenorphine. The maternal plasma AUCs of norbuprenorphine in Abcb1a-/-/1b-/- and Abcb1a-/-/1b-/-/Abcg2-/- mice were ∼2 times greater than that in wild-type mice. Fetal AUCs in Abcb1a-/-/1b-/- and Abcb1a-/-/1b-/-/Abcg2-/- mice were also increased compared to wild-type mice; however, the fetal-to-maternal plasma AUC ratio remained relatively unchanged by the knockout of Abcb1a/1b or Abcb1a/1b/Abcg2. In contrast, the maternal brain-to-maternal plasma AUC ratio in Abcb1a-/-/1b-/- or Abcb1a-/-/1b-/-/Abcg2-/- mice was increased ∼30-fold compared to wild-type mice. Protein quantification by LC-MS/MS proteomics revealed significantly higher amounts of P-gp protein in the wild-type mice brain than that in the placenta. These results indicate that fetal exposure to norbuprenorphine is substantial and that P-gp has a minor impact on fetal exposure to norbuprenorphine, but plays a significant role in restricting its brain distribution. The differential impacts of P-gp on norbuprenorphine distribution into the brain and fetus are likely, at least in part, due to the differences in amounts of P-gp protein expressed in the blood-brain and blood-placental barriers. BCRP is not as important as P-gp in determining both the systemic and tissue exposure to norbuprenorphine. Finally, fetal AUCs of the metabolite norbuprenorphine-β-d-glucuronide were 3-7 times greater than maternal plasma AUCs, while the maternal brain AUCs were <50% of maternal plasma AUCs, suggesting that a reversible pool of conjugated metabolite in the fetus may contribute to the high fetal exposure to norbuprenorphine.
Collapse
MESH Headings
- ATP Binding Cassette Transporter, Subfamily B, Member 1/analysis
- ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics
- ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism
- ATP Binding Cassette Transporter, Subfamily G, Member 2/analysis
- ATP Binding Cassette Transporter, Subfamily G, Member 2/genetics
- ATP Binding Cassette Transporter, Subfamily G, Member 2/metabolism
- Animals
- Blood-Brain Barrier/metabolism
- Brain/metabolism
- Buprenorphine/administration & dosage
- Buprenorphine/analogs & derivatives
- Buprenorphine/metabolism
- Buprenorphine/pharmacokinetics
- Female
- Gene Knockout Techniques
- Maternal Exposure
- Maternal-Fetal Exchange
- Mice
- Mice, Knockout
- Narcotic Antagonists/administration & dosage
- Narcotic Antagonists/metabolism
- Narcotic Antagonists/pharmacokinetics
- Pregnancy
- Tissue Distribution
Collapse
Affiliation(s)
- Michael Z Liao
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, WA 98195, USA
| | - Chunying Gao
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, WA 98195, USA
| | - Laura M Shireman
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, WA 98195, USA
| | - Brian Phillips
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, WA 98195, USA
| | - Linda J Risler
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, WA 98195, USA
| | - Naveen K Neradugomma
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, WA 98195, USA
| | - Prachi Choudhari
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, WA 98195, USA
| | - Bhagwat Prasad
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, WA 98195, USA
| | - Danny D Shen
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, WA 98195, USA
| | - Qingcheng Mao
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, WA 98195, USA.
| |
Collapse
|
45
|
Breast cancer resistance protein (BCRP/ABCG2) and P-glycoprotein (P-gp/ABCB1) transport afatinib and restrict its oral availability and brain accumulation. Pharmacol Res 2017; 120:43-50. [PMID: 28288939 DOI: 10.1016/j.phrs.2017.01.035] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Revised: 12/22/2016] [Accepted: 01/28/2017] [Indexed: 12/29/2022]
Abstract
Afatinib is a highly selective, irreversible inhibitor of EGFR and HER-2. It is orally administered for the treatment of patients with EGFR mutation-positive types of metastatic NSCLC. We investigated whether afatinib is a substrate for the multidrug efflux transporters ABCB1 and ABCG2 and whether these transporters influence oral availability and brain and other tissue accumulation of afatinib. We used in vitro transport assays to assess human (h)ABCB1-, hABCG2- or murine (m)Abcg2-mediated transport of afatinib. To study the single and combined roles of Abcg2 and Abcb1a/1b in oral afatinib disposition, we used appropriate knockout mouse strains. Afatinib was transported well by hABCB1, hABCG2 and mAbcg2 in vitro. Upon oral administration of afatinib, Abcg2-/-, Abcb1a/1b-/- and Abcb1a/1b-/-;Abcg2-/- mice displayed a 4.2-, 2.4- and 7-fold increased afatinib plasma AUC0-24 compared with wild-type mice. Abcg2-deficient strains also displayed decreased afatinib plasma clearance. At 2h, relative brain accumulation of afatinib was not significantly altered in the single knockout strains, but 23.8-fold increased in Abcb1a/1b-/-;Abcg2-/- mice compared to wild-type mice. Abcg2 and Abcb1a/1b restrict oral availability and brain accumulation of afatinib. Inhibition of these transporters may therefore be of clinical importance for patients with brain (micro)metastases positioned behind an intact blood-brain barrier.
Collapse
|
46
|
Systematic evaluation of multifunctional paclitaxel-loaded polymeric mixed micelles as a potential anticancer remedy to overcome multidrug resistance. Acta Biomater 2017; 50:381-395. [PMID: 27956367 DOI: 10.1016/j.actbio.2016.12.021] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Revised: 10/20/2016] [Accepted: 12/07/2016] [Indexed: 01/13/2023]
Abstract
Multidrug resistance (MDR) of tumor cells is becoming the main reason for the failure of chemotherapy and P-glycoprotein (P-gp) mediated drug efflux has demonstrated to be the key factor for MDR. To address this issue, a novel pH-responsive mixed micelles drug delivery system composed of dextran-g-poly(lactide-co-glycolide)-g-histidine (HDP) and folate acid-D-α-tocopheryl polyethylene glycol 2000 (FA-TPGS2K) copolymers has been designed for the delivery of antitumor agent, paclitaxel (PTX) via FA-receptor mediated cell endocytosis, into PTX-resistant breast cancer MCF-7 cells (MCF-7/PTX). PTX-loaded FA-TPGS2K/HDP mixed micelles were characterized to have a small size distribution, high loading content and excellent pH-responsive drug release profiles. Compared with HDP micelles, FA-TPGS2K/HDP mixed micelles showed a higher cytotoxicity against MCF-7 and MCF-7/PTX cells due to the synergistic effect of FA-receptor mediated cell endocytosis, pH-responsive drug release and TPGS mediated P-gp inhibition. P-gp expression level, ATP content and mitochondrial membrane potential change have been measured, the results indicated blank FA-TPGS2K/HDP mixed micelles could inhibit the P-gp activity by reducing the mitochondrial membrane potential and depleting ATP content but not down-regulating the P-gp expression. In vivo antitumor activities demonstrated FA-TPGS2K/HDP mixed micelles could reach higher antitumor activity compared with HDP micelles for MCF-7/PTX tumor cells. Histological assay also indicated that FA-TPGS2K/HDP mixed micelles showed strongly apoptosis inducing effect, anti-proliferation effect and anti-angiogenesis effect. All these evidences demonstrated this pH-sensitive FA-TPGS2K/HDP micelle-based drug delivery system is a promising approach for overcoming MDR. STATEMENT OF SIGNIFICANCE In this work, a novel FA-TPGS2K copolymer has been synthesized and used it to construct mixed micelles with HDP copolymer to overcome MDR effect. Furthermore, a series in vitro and in vivo evaluations have been made, which supported enough evidences for the efficient delivery of antitumor drug to MDR cells.
Collapse
|
47
|
Lin JJ, Riely GJ, Shaw AT. Targeting ALK: Precision Medicine Takes on Drug Resistance. Cancer Discov 2017; 7:137-155. [PMID: 28122866 PMCID: PMC5296241 DOI: 10.1158/2159-8290.cd-16-1123] [Citation(s) in RCA: 363] [Impact Index Per Article: 51.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2016] [Revised: 12/13/2016] [Accepted: 12/14/2016] [Indexed: 12/14/2022]
Abstract
Anaplastic lymphoma kinase (ALK) is a validated molecular target in several ALK-rearranged malignancies, including non-small cell lung cancer. However, the clinical benefit of targeting ALK using tyrosine kinase inhibitors (TKI) is almost universally limited by the emergence of drug resistance. Diverse mechanisms of resistance to ALK TKIs have now been discovered, and these basic mechanisms are informing the development of novel therapeutic strategies to overcome resistance in the clinic. In this review, we summarize the current successes and challenges of targeting ALK. SIGNIFICANCE Effective long-term treatment of ALK-rearranged cancers requires a mechanistic understanding of resistance to ALK TKIs so that rational therapies can be selected to combat resistance. This review underscores the importance of serial biopsies in capturing the dynamic therapeutic vulnerabilities within a patient's tumor and offers a perspective into the complexity of on-target and off-target ALK TKI resistance mechanisms. Therapeutic strategies that can successfully overcome, and potentially prevent, these resistance mechanisms will have the greatest impact on patient outcome. Cancer Discov; 7(2); 137-55. ©2017 AACR.
Collapse
Affiliation(s)
- Jessica J Lin
- Massachusetts General Hospital Cancer Center, Boston, Massachusetts
| | - Gregory J Riely
- Memorial Sloan Kettering Cancer Center, Weill Cornell Medical College, New York, New York
| | - Alice T Shaw
- Massachusetts General Hospital Cancer Center, Boston, Massachusetts.
| |
Collapse
|
48
|
Chamberlain MC, Baik CS, Gadi VK, Bhatia S, Chow LQM. Systemic therapy of brain metastases: non-small cell lung cancer, breast cancer, and melanoma. Neuro Oncol 2017; 19:i1-i24. [PMID: 28031389 PMCID: PMC5193029 DOI: 10.1093/neuonc/now197] [Citation(s) in RCA: 146] [Impact Index Per Article: 20.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Brain metastases (BM) occur frequently in many cancers, particularly non-small cell lung cancer (NSCLC), breast cancer, and melanoma. The development of BM is associated with poor prognosis and has an adverse impact on survival and quality of life. Commonly used therapies for BM such as surgery or radiotherapy are associated with only modest benefits. However, recent advances in systemic therapy of many cancers have generated considerable interest in exploration of those therapies for treatment of intracranial metastases.This review discusses the epidemiology of BM from the aforementioned primary tumors and the challenges of using systemic therapies for metastatic disease located within the central nervous system. Cumulative data from several retrospective and small prospective studies suggest that molecularly targeted systemic therapies may be an effective option for the treatment of BM from NSCLC, breast cancer, and melanoma, either as monotherapy or in conjunction with other therapies. Larger prospective studies are warranted to further characterize the efficacy and safety profiles of these targeted agents for the treatment of BM.
Collapse
Affiliation(s)
- Marc C Chamberlain
- Seattle Cancer Center Alliance, Seattle, Washington (M.C.C., C.S.B., V.K.G., S.B., L.Q.M.C.); Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington (C.S.B., V.K.G., L.Q.M.C.); Departments of Neurology and Neurological Surgery, University of Washington, Seattle, Washington (M.C.C.); Division of Medical Oncology, University of Washington, Seattle, Washington (C.S.B., V.K.G., S.B., L.Q.M.C)
| | - Christina S Baik
- Seattle Cancer Center Alliance, Seattle, Washington (M.C.C., C.S.B., V.K.G., S.B., L.Q.M.C.); Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington (C.S.B., V.K.G., L.Q.M.C.); Departments of Neurology and Neurological Surgery, University of Washington, Seattle, Washington (M.C.C.); Division of Medical Oncology, University of Washington, Seattle, Washington (C.S.B., V.K.G., S.B., L.Q.M.C)
| | - Vijayakrishna K Gadi
- Seattle Cancer Center Alliance, Seattle, Washington (M.C.C., C.S.B., V.K.G., S.B., L.Q.M.C.); Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington (C.S.B., V.K.G., L.Q.M.C.); Departments of Neurology and Neurological Surgery, University of Washington, Seattle, Washington (M.C.C.); Division of Medical Oncology, University of Washington, Seattle, Washington (C.S.B., V.K.G., S.B., L.Q.M.C)
| | - Shailender Bhatia
- Seattle Cancer Center Alliance, Seattle, Washington (M.C.C., C.S.B., V.K.G., S.B., L.Q.M.C.); Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington (C.S.B., V.K.G., L.Q.M.C.); Departments of Neurology and Neurological Surgery, University of Washington, Seattle, Washington (M.C.C.); Division of Medical Oncology, University of Washington, Seattle, Washington (C.S.B., V.K.G., S.B., L.Q.M.C)
| | - Laura Q M Chow
- Seattle Cancer Center Alliance, Seattle, Washington (M.C.C., C.S.B., V.K.G., S.B., L.Q.M.C.); Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington (C.S.B., V.K.G., L.Q.M.C.); Departments of Neurology and Neurological Surgery, University of Washington, Seattle, Washington (M.C.C.); Division of Medical Oncology, University of Washington, Seattle, Washington (C.S.B., V.K.G., S.B., L.Q.M.C)
| |
Collapse
|
49
|
Gadgeel SM, Shaw AT, Govindan R, Gandhi L, Socinski MA, Camidge DR, De Petris L, Kim DW, Chiappori A, Moro-Sibilot DL, Duruisseaux M, Crino L, De Pas T, Dansin E, Tessmer A, Yang JCH, Han JY, Bordogna W, Golding S, Zeaiter A, Ou SHI. Pooled Analysis of CNS Response to Alectinib in Two Studies of Pretreated Patients With ALK-Positive Non-Small-Cell Lung Cancer. J Clin Oncol 2016; 34:4079-4085. [PMID: 27863201 PMCID: PMC7845943 DOI: 10.1200/jco.2016.68.4639] [Citation(s) in RCA: 142] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Purpose Alectinib has shown activity in the CNS in phase I and II studies. To further evaluate this activity, we pooled efficacy and safety data from two single-arm phase II studies (NP28761 and NP28673; ClinicalTrials.gov identifiers: NCT01871805 and NCT01801111, respectively) in patients with ALK-positive non-small-cell lung cancer (NSCLC). Patients and Methods Both studies included patients with ALK-positive NSCLC who had previously received crizotinib; all patients received alectinib 600 mg twice per day. The primary end point in both studies was independent review committee (IRC)-assessed objective response rate (ORR; by Response Evaluation Criteria in Solid Tumors [RECIST] version 1.1). Additional end points (all by IRC) included CNS ORR (CORR), CNS disease control rate (CDCR), and CNS duration of response (CDOR). Results One hundred thirty-six patients had baseline CNS metastases (60% of the overall study populations); 50 patients (37%) had measurable CNS disease at baseline. Ninety-five patients (70%) had prior CNS radiotherapy; 55 patients completed the CNS radiotherapy more than 6 months before starting alectinib. Median follow-up time was 12.4 months (range, 0.9 to 19.7 months). For patients with baseline measurable CNS disease, IRC CORR was 64.0% (95% CI, 49.2% to 77.1%), CDCR was 90.0% (95% CI, 78.2% to 96.7%), and median CDOR was 10.8 months (95% CI, 7.6 to 14.1 months). For patients with measurable and/or nonmeasurable baseline CNS disease, IRC CORR was 42.6% (95% CI, 34.2% to 51.4%), CDCR was 85.3% (95% CI, 78.2% to 90.8%), and median CDOR was 11.1 months (95% CI, 10.3 months to not evaluable). CORR was 35.8% (95% CI, 26.2% to 46.3%) for patients with prior radiotherapy (n = 95) and 58.5% (95% CI, 42.1% to 73.7%) for patients without prior radiotherapy (n = 41). As previously reported, alectinib was well tolerated, regardless of baseline CNS disease. Conclusion Alectinib showed good efficacy against CNS metastases, in addition to systemic activity, in crizotinib-refractory ALK-positive NSCLC.
Collapse
Affiliation(s)
- Shirish M. Gadgeel
- Shirish M. Gadgeel, Karmanos Cancer Institute, Wayne State University, Detroit, MI; Alice T. Shaw, Massachusetts General Hospital; Leena Gandhi, Dana-Farber Cancer Institute, Boston, MA; Ramaswamy Govindan, Washington University School of Medicine, St Louis, MO; Mark A. Socinski, University of Pittsburgh, Pittsburgh, PA; D. Ross Camidge, University of Colorado Cancer Center, Denver, CO; Alberto Chiappori, Thoracic Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL; Sai-Hong Ignatius Ou, University of California at Irvine, Orange, CA; Luigi De Petris, Karolinska Institutet, Stockholm, Sweden; Dong-Wan Kim, Seoul National University Hospital, Seoul; Ji-Youn Han, Lung Cancer Centre, National Cancer Centre, Goyang, South Korea; Denis L. Moro-Sibilot, Service de Pneumologie; Michael Duruisseaux, Centre Hospitalier Universitaire de Grenoble, Grenoble; Eric Dansin, Centre Oscar Lambret, Lille, France; Lucio Crino, Santa Maria della Misericordia Hospital, Perugia; Tommaso De Pas, European Institute of Oncology, Milan, Italy; Antje Tessmer, Evangelische Lungenklinik Berlin, Berlin, Germany; James Chih-Hsin Yang, Graduate Institute of Oncology and Cancer Research Centre, National Taiwan University, Taipei, Taiwan; and Walter Bordogna, Sophie Golding, and Ali Zeaiter, F. Hoffmann-La Roche, Basel, Switzerland
| | - Alice T. Shaw
- Shirish M. Gadgeel, Karmanos Cancer Institute, Wayne State University, Detroit, MI; Alice T. Shaw, Massachusetts General Hospital; Leena Gandhi, Dana-Farber Cancer Institute, Boston, MA; Ramaswamy Govindan, Washington University School of Medicine, St Louis, MO; Mark A. Socinski, University of Pittsburgh, Pittsburgh, PA; D. Ross Camidge, University of Colorado Cancer Center, Denver, CO; Alberto Chiappori, Thoracic Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL; Sai-Hong Ignatius Ou, University of California at Irvine, Orange, CA; Luigi De Petris, Karolinska Institutet, Stockholm, Sweden; Dong-Wan Kim, Seoul National University Hospital, Seoul; Ji-Youn Han, Lung Cancer Centre, National Cancer Centre, Goyang, South Korea; Denis L. Moro-Sibilot, Service de Pneumologie; Michael Duruisseaux, Centre Hospitalier Universitaire de Grenoble, Grenoble; Eric Dansin, Centre Oscar Lambret, Lille, France; Lucio Crino, Santa Maria della Misericordia Hospital, Perugia; Tommaso De Pas, European Institute of Oncology, Milan, Italy; Antje Tessmer, Evangelische Lungenklinik Berlin, Berlin, Germany; James Chih-Hsin Yang, Graduate Institute of Oncology and Cancer Research Centre, National Taiwan University, Taipei, Taiwan; and Walter Bordogna, Sophie Golding, and Ali Zeaiter, F. Hoffmann-La Roche, Basel, Switzerland
| | - Ramaswamy Govindan
- Shirish M. Gadgeel, Karmanos Cancer Institute, Wayne State University, Detroit, MI; Alice T. Shaw, Massachusetts General Hospital; Leena Gandhi, Dana-Farber Cancer Institute, Boston, MA; Ramaswamy Govindan, Washington University School of Medicine, St Louis, MO; Mark A. Socinski, University of Pittsburgh, Pittsburgh, PA; D. Ross Camidge, University of Colorado Cancer Center, Denver, CO; Alberto Chiappori, Thoracic Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL; Sai-Hong Ignatius Ou, University of California at Irvine, Orange, CA; Luigi De Petris, Karolinska Institutet, Stockholm, Sweden; Dong-Wan Kim, Seoul National University Hospital, Seoul; Ji-Youn Han, Lung Cancer Centre, National Cancer Centre, Goyang, South Korea; Denis L. Moro-Sibilot, Service de Pneumologie; Michael Duruisseaux, Centre Hospitalier Universitaire de Grenoble, Grenoble; Eric Dansin, Centre Oscar Lambret, Lille, France; Lucio Crino, Santa Maria della Misericordia Hospital, Perugia; Tommaso De Pas, European Institute of Oncology, Milan, Italy; Antje Tessmer, Evangelische Lungenklinik Berlin, Berlin, Germany; James Chih-Hsin Yang, Graduate Institute of Oncology and Cancer Research Centre, National Taiwan University, Taipei, Taiwan; and Walter Bordogna, Sophie Golding, and Ali Zeaiter, F. Hoffmann-La Roche, Basel, Switzerland
| | - Leena Gandhi
- Shirish M. Gadgeel, Karmanos Cancer Institute, Wayne State University, Detroit, MI; Alice T. Shaw, Massachusetts General Hospital; Leena Gandhi, Dana-Farber Cancer Institute, Boston, MA; Ramaswamy Govindan, Washington University School of Medicine, St Louis, MO; Mark A. Socinski, University of Pittsburgh, Pittsburgh, PA; D. Ross Camidge, University of Colorado Cancer Center, Denver, CO; Alberto Chiappori, Thoracic Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL; Sai-Hong Ignatius Ou, University of California at Irvine, Orange, CA; Luigi De Petris, Karolinska Institutet, Stockholm, Sweden; Dong-Wan Kim, Seoul National University Hospital, Seoul; Ji-Youn Han, Lung Cancer Centre, National Cancer Centre, Goyang, South Korea; Denis L. Moro-Sibilot, Service de Pneumologie; Michael Duruisseaux, Centre Hospitalier Universitaire de Grenoble, Grenoble; Eric Dansin, Centre Oscar Lambret, Lille, France; Lucio Crino, Santa Maria della Misericordia Hospital, Perugia; Tommaso De Pas, European Institute of Oncology, Milan, Italy; Antje Tessmer, Evangelische Lungenklinik Berlin, Berlin, Germany; James Chih-Hsin Yang, Graduate Institute of Oncology and Cancer Research Centre, National Taiwan University, Taipei, Taiwan; and Walter Bordogna, Sophie Golding, and Ali Zeaiter, F. Hoffmann-La Roche, Basel, Switzerland
| | - Mark A. Socinski
- Shirish M. Gadgeel, Karmanos Cancer Institute, Wayne State University, Detroit, MI; Alice T. Shaw, Massachusetts General Hospital; Leena Gandhi, Dana-Farber Cancer Institute, Boston, MA; Ramaswamy Govindan, Washington University School of Medicine, St Louis, MO; Mark A. Socinski, University of Pittsburgh, Pittsburgh, PA; D. Ross Camidge, University of Colorado Cancer Center, Denver, CO; Alberto Chiappori, Thoracic Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL; Sai-Hong Ignatius Ou, University of California at Irvine, Orange, CA; Luigi De Petris, Karolinska Institutet, Stockholm, Sweden; Dong-Wan Kim, Seoul National University Hospital, Seoul; Ji-Youn Han, Lung Cancer Centre, National Cancer Centre, Goyang, South Korea; Denis L. Moro-Sibilot, Service de Pneumologie; Michael Duruisseaux, Centre Hospitalier Universitaire de Grenoble, Grenoble; Eric Dansin, Centre Oscar Lambret, Lille, France; Lucio Crino, Santa Maria della Misericordia Hospital, Perugia; Tommaso De Pas, European Institute of Oncology, Milan, Italy; Antje Tessmer, Evangelische Lungenklinik Berlin, Berlin, Germany; James Chih-Hsin Yang, Graduate Institute of Oncology and Cancer Research Centre, National Taiwan University, Taipei, Taiwan; and Walter Bordogna, Sophie Golding, and Ali Zeaiter, F. Hoffmann-La Roche, Basel, Switzerland
| | - D. Ross Camidge
- Shirish M. Gadgeel, Karmanos Cancer Institute, Wayne State University, Detroit, MI; Alice T. Shaw, Massachusetts General Hospital; Leena Gandhi, Dana-Farber Cancer Institute, Boston, MA; Ramaswamy Govindan, Washington University School of Medicine, St Louis, MO; Mark A. Socinski, University of Pittsburgh, Pittsburgh, PA; D. Ross Camidge, University of Colorado Cancer Center, Denver, CO; Alberto Chiappori, Thoracic Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL; Sai-Hong Ignatius Ou, University of California at Irvine, Orange, CA; Luigi De Petris, Karolinska Institutet, Stockholm, Sweden; Dong-Wan Kim, Seoul National University Hospital, Seoul; Ji-Youn Han, Lung Cancer Centre, National Cancer Centre, Goyang, South Korea; Denis L. Moro-Sibilot, Service de Pneumologie; Michael Duruisseaux, Centre Hospitalier Universitaire de Grenoble, Grenoble; Eric Dansin, Centre Oscar Lambret, Lille, France; Lucio Crino, Santa Maria della Misericordia Hospital, Perugia; Tommaso De Pas, European Institute of Oncology, Milan, Italy; Antje Tessmer, Evangelische Lungenklinik Berlin, Berlin, Germany; James Chih-Hsin Yang, Graduate Institute of Oncology and Cancer Research Centre, National Taiwan University, Taipei, Taiwan; and Walter Bordogna, Sophie Golding, and Ali Zeaiter, F. Hoffmann-La Roche, Basel, Switzerland
| | - Luigi De Petris
- Shirish M. Gadgeel, Karmanos Cancer Institute, Wayne State University, Detroit, MI; Alice T. Shaw, Massachusetts General Hospital; Leena Gandhi, Dana-Farber Cancer Institute, Boston, MA; Ramaswamy Govindan, Washington University School of Medicine, St Louis, MO; Mark A. Socinski, University of Pittsburgh, Pittsburgh, PA; D. Ross Camidge, University of Colorado Cancer Center, Denver, CO; Alberto Chiappori, Thoracic Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL; Sai-Hong Ignatius Ou, University of California at Irvine, Orange, CA; Luigi De Petris, Karolinska Institutet, Stockholm, Sweden; Dong-Wan Kim, Seoul National University Hospital, Seoul; Ji-Youn Han, Lung Cancer Centre, National Cancer Centre, Goyang, South Korea; Denis L. Moro-Sibilot, Service de Pneumologie; Michael Duruisseaux, Centre Hospitalier Universitaire de Grenoble, Grenoble; Eric Dansin, Centre Oscar Lambret, Lille, France; Lucio Crino, Santa Maria della Misericordia Hospital, Perugia; Tommaso De Pas, European Institute of Oncology, Milan, Italy; Antje Tessmer, Evangelische Lungenklinik Berlin, Berlin, Germany; James Chih-Hsin Yang, Graduate Institute of Oncology and Cancer Research Centre, National Taiwan University, Taipei, Taiwan; and Walter Bordogna, Sophie Golding, and Ali Zeaiter, F. Hoffmann-La Roche, Basel, Switzerland
| | - Dong-Wan Kim
- Shirish M. Gadgeel, Karmanos Cancer Institute, Wayne State University, Detroit, MI; Alice T. Shaw, Massachusetts General Hospital; Leena Gandhi, Dana-Farber Cancer Institute, Boston, MA; Ramaswamy Govindan, Washington University School of Medicine, St Louis, MO; Mark A. Socinski, University of Pittsburgh, Pittsburgh, PA; D. Ross Camidge, University of Colorado Cancer Center, Denver, CO; Alberto Chiappori, Thoracic Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL; Sai-Hong Ignatius Ou, University of California at Irvine, Orange, CA; Luigi De Petris, Karolinska Institutet, Stockholm, Sweden; Dong-Wan Kim, Seoul National University Hospital, Seoul; Ji-Youn Han, Lung Cancer Centre, National Cancer Centre, Goyang, South Korea; Denis L. Moro-Sibilot, Service de Pneumologie; Michael Duruisseaux, Centre Hospitalier Universitaire de Grenoble, Grenoble; Eric Dansin, Centre Oscar Lambret, Lille, France; Lucio Crino, Santa Maria della Misericordia Hospital, Perugia; Tommaso De Pas, European Institute of Oncology, Milan, Italy; Antje Tessmer, Evangelische Lungenklinik Berlin, Berlin, Germany; James Chih-Hsin Yang, Graduate Institute of Oncology and Cancer Research Centre, National Taiwan University, Taipei, Taiwan; and Walter Bordogna, Sophie Golding, and Ali Zeaiter, F. Hoffmann-La Roche, Basel, Switzerland
| | - Alberto Chiappori
- Shirish M. Gadgeel, Karmanos Cancer Institute, Wayne State University, Detroit, MI; Alice T. Shaw, Massachusetts General Hospital; Leena Gandhi, Dana-Farber Cancer Institute, Boston, MA; Ramaswamy Govindan, Washington University School of Medicine, St Louis, MO; Mark A. Socinski, University of Pittsburgh, Pittsburgh, PA; D. Ross Camidge, University of Colorado Cancer Center, Denver, CO; Alberto Chiappori, Thoracic Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL; Sai-Hong Ignatius Ou, University of California at Irvine, Orange, CA; Luigi De Petris, Karolinska Institutet, Stockholm, Sweden; Dong-Wan Kim, Seoul National University Hospital, Seoul; Ji-Youn Han, Lung Cancer Centre, National Cancer Centre, Goyang, South Korea; Denis L. Moro-Sibilot, Service de Pneumologie; Michael Duruisseaux, Centre Hospitalier Universitaire de Grenoble, Grenoble; Eric Dansin, Centre Oscar Lambret, Lille, France; Lucio Crino, Santa Maria della Misericordia Hospital, Perugia; Tommaso De Pas, European Institute of Oncology, Milan, Italy; Antje Tessmer, Evangelische Lungenklinik Berlin, Berlin, Germany; James Chih-Hsin Yang, Graduate Institute of Oncology and Cancer Research Centre, National Taiwan University, Taipei, Taiwan; and Walter Bordogna, Sophie Golding, and Ali Zeaiter, F. Hoffmann-La Roche, Basel, Switzerland
| | - Denis L. Moro-Sibilot
- Shirish M. Gadgeel, Karmanos Cancer Institute, Wayne State University, Detroit, MI; Alice T. Shaw, Massachusetts General Hospital; Leena Gandhi, Dana-Farber Cancer Institute, Boston, MA; Ramaswamy Govindan, Washington University School of Medicine, St Louis, MO; Mark A. Socinski, University of Pittsburgh, Pittsburgh, PA; D. Ross Camidge, University of Colorado Cancer Center, Denver, CO; Alberto Chiappori, Thoracic Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL; Sai-Hong Ignatius Ou, University of California at Irvine, Orange, CA; Luigi De Petris, Karolinska Institutet, Stockholm, Sweden; Dong-Wan Kim, Seoul National University Hospital, Seoul; Ji-Youn Han, Lung Cancer Centre, National Cancer Centre, Goyang, South Korea; Denis L. Moro-Sibilot, Service de Pneumologie; Michael Duruisseaux, Centre Hospitalier Universitaire de Grenoble, Grenoble; Eric Dansin, Centre Oscar Lambret, Lille, France; Lucio Crino, Santa Maria della Misericordia Hospital, Perugia; Tommaso De Pas, European Institute of Oncology, Milan, Italy; Antje Tessmer, Evangelische Lungenklinik Berlin, Berlin, Germany; James Chih-Hsin Yang, Graduate Institute of Oncology and Cancer Research Centre, National Taiwan University, Taipei, Taiwan; and Walter Bordogna, Sophie Golding, and Ali Zeaiter, F. Hoffmann-La Roche, Basel, Switzerland
| | - Michael Duruisseaux
- Shirish M. Gadgeel, Karmanos Cancer Institute, Wayne State University, Detroit, MI; Alice T. Shaw, Massachusetts General Hospital; Leena Gandhi, Dana-Farber Cancer Institute, Boston, MA; Ramaswamy Govindan, Washington University School of Medicine, St Louis, MO; Mark A. Socinski, University of Pittsburgh, Pittsburgh, PA; D. Ross Camidge, University of Colorado Cancer Center, Denver, CO; Alberto Chiappori, Thoracic Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL; Sai-Hong Ignatius Ou, University of California at Irvine, Orange, CA; Luigi De Petris, Karolinska Institutet, Stockholm, Sweden; Dong-Wan Kim, Seoul National University Hospital, Seoul; Ji-Youn Han, Lung Cancer Centre, National Cancer Centre, Goyang, South Korea; Denis L. Moro-Sibilot, Service de Pneumologie; Michael Duruisseaux, Centre Hospitalier Universitaire de Grenoble, Grenoble; Eric Dansin, Centre Oscar Lambret, Lille, France; Lucio Crino, Santa Maria della Misericordia Hospital, Perugia; Tommaso De Pas, European Institute of Oncology, Milan, Italy; Antje Tessmer, Evangelische Lungenklinik Berlin, Berlin, Germany; James Chih-Hsin Yang, Graduate Institute of Oncology and Cancer Research Centre, National Taiwan University, Taipei, Taiwan; and Walter Bordogna, Sophie Golding, and Ali Zeaiter, F. Hoffmann-La Roche, Basel, Switzerland
| | - Lucio Crino
- Shirish M. Gadgeel, Karmanos Cancer Institute, Wayne State University, Detroit, MI; Alice T. Shaw, Massachusetts General Hospital; Leena Gandhi, Dana-Farber Cancer Institute, Boston, MA; Ramaswamy Govindan, Washington University School of Medicine, St Louis, MO; Mark A. Socinski, University of Pittsburgh, Pittsburgh, PA; D. Ross Camidge, University of Colorado Cancer Center, Denver, CO; Alberto Chiappori, Thoracic Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL; Sai-Hong Ignatius Ou, University of California at Irvine, Orange, CA; Luigi De Petris, Karolinska Institutet, Stockholm, Sweden; Dong-Wan Kim, Seoul National University Hospital, Seoul; Ji-Youn Han, Lung Cancer Centre, National Cancer Centre, Goyang, South Korea; Denis L. Moro-Sibilot, Service de Pneumologie; Michael Duruisseaux, Centre Hospitalier Universitaire de Grenoble, Grenoble; Eric Dansin, Centre Oscar Lambret, Lille, France; Lucio Crino, Santa Maria della Misericordia Hospital, Perugia; Tommaso De Pas, European Institute of Oncology, Milan, Italy; Antje Tessmer, Evangelische Lungenklinik Berlin, Berlin, Germany; James Chih-Hsin Yang, Graduate Institute of Oncology and Cancer Research Centre, National Taiwan University, Taipei, Taiwan; and Walter Bordogna, Sophie Golding, and Ali Zeaiter, F. Hoffmann-La Roche, Basel, Switzerland
| | - Tommaso De Pas
- Shirish M. Gadgeel, Karmanos Cancer Institute, Wayne State University, Detroit, MI; Alice T. Shaw, Massachusetts General Hospital; Leena Gandhi, Dana-Farber Cancer Institute, Boston, MA; Ramaswamy Govindan, Washington University School of Medicine, St Louis, MO; Mark A. Socinski, University of Pittsburgh, Pittsburgh, PA; D. Ross Camidge, University of Colorado Cancer Center, Denver, CO; Alberto Chiappori, Thoracic Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL; Sai-Hong Ignatius Ou, University of California at Irvine, Orange, CA; Luigi De Petris, Karolinska Institutet, Stockholm, Sweden; Dong-Wan Kim, Seoul National University Hospital, Seoul; Ji-Youn Han, Lung Cancer Centre, National Cancer Centre, Goyang, South Korea; Denis L. Moro-Sibilot, Service de Pneumologie; Michael Duruisseaux, Centre Hospitalier Universitaire de Grenoble, Grenoble; Eric Dansin, Centre Oscar Lambret, Lille, France; Lucio Crino, Santa Maria della Misericordia Hospital, Perugia; Tommaso De Pas, European Institute of Oncology, Milan, Italy; Antje Tessmer, Evangelische Lungenklinik Berlin, Berlin, Germany; James Chih-Hsin Yang, Graduate Institute of Oncology and Cancer Research Centre, National Taiwan University, Taipei, Taiwan; and Walter Bordogna, Sophie Golding, and Ali Zeaiter, F. Hoffmann-La Roche, Basel, Switzerland
| | - Eric Dansin
- Shirish M. Gadgeel, Karmanos Cancer Institute, Wayne State University, Detroit, MI; Alice T. Shaw, Massachusetts General Hospital; Leena Gandhi, Dana-Farber Cancer Institute, Boston, MA; Ramaswamy Govindan, Washington University School of Medicine, St Louis, MO; Mark A. Socinski, University of Pittsburgh, Pittsburgh, PA; D. Ross Camidge, University of Colorado Cancer Center, Denver, CO; Alberto Chiappori, Thoracic Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL; Sai-Hong Ignatius Ou, University of California at Irvine, Orange, CA; Luigi De Petris, Karolinska Institutet, Stockholm, Sweden; Dong-Wan Kim, Seoul National University Hospital, Seoul; Ji-Youn Han, Lung Cancer Centre, National Cancer Centre, Goyang, South Korea; Denis L. Moro-Sibilot, Service de Pneumologie; Michael Duruisseaux, Centre Hospitalier Universitaire de Grenoble, Grenoble; Eric Dansin, Centre Oscar Lambret, Lille, France; Lucio Crino, Santa Maria della Misericordia Hospital, Perugia; Tommaso De Pas, European Institute of Oncology, Milan, Italy; Antje Tessmer, Evangelische Lungenklinik Berlin, Berlin, Germany; James Chih-Hsin Yang, Graduate Institute of Oncology and Cancer Research Centre, National Taiwan University, Taipei, Taiwan; and Walter Bordogna, Sophie Golding, and Ali Zeaiter, F. Hoffmann-La Roche, Basel, Switzerland
| | - Antje Tessmer
- Shirish M. Gadgeel, Karmanos Cancer Institute, Wayne State University, Detroit, MI; Alice T. Shaw, Massachusetts General Hospital; Leena Gandhi, Dana-Farber Cancer Institute, Boston, MA; Ramaswamy Govindan, Washington University School of Medicine, St Louis, MO; Mark A. Socinski, University of Pittsburgh, Pittsburgh, PA; D. Ross Camidge, University of Colorado Cancer Center, Denver, CO; Alberto Chiappori, Thoracic Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL; Sai-Hong Ignatius Ou, University of California at Irvine, Orange, CA; Luigi De Petris, Karolinska Institutet, Stockholm, Sweden; Dong-Wan Kim, Seoul National University Hospital, Seoul; Ji-Youn Han, Lung Cancer Centre, National Cancer Centre, Goyang, South Korea; Denis L. Moro-Sibilot, Service de Pneumologie; Michael Duruisseaux, Centre Hospitalier Universitaire de Grenoble, Grenoble; Eric Dansin, Centre Oscar Lambret, Lille, France; Lucio Crino, Santa Maria della Misericordia Hospital, Perugia; Tommaso De Pas, European Institute of Oncology, Milan, Italy; Antje Tessmer, Evangelische Lungenklinik Berlin, Berlin, Germany; James Chih-Hsin Yang, Graduate Institute of Oncology and Cancer Research Centre, National Taiwan University, Taipei, Taiwan; and Walter Bordogna, Sophie Golding, and Ali Zeaiter, F. Hoffmann-La Roche, Basel, Switzerland
| | - James Chih-Hsin Yang
- Shirish M. Gadgeel, Karmanos Cancer Institute, Wayne State University, Detroit, MI; Alice T. Shaw, Massachusetts General Hospital; Leena Gandhi, Dana-Farber Cancer Institute, Boston, MA; Ramaswamy Govindan, Washington University School of Medicine, St Louis, MO; Mark A. Socinski, University of Pittsburgh, Pittsburgh, PA; D. Ross Camidge, University of Colorado Cancer Center, Denver, CO; Alberto Chiappori, Thoracic Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL; Sai-Hong Ignatius Ou, University of California at Irvine, Orange, CA; Luigi De Petris, Karolinska Institutet, Stockholm, Sweden; Dong-Wan Kim, Seoul National University Hospital, Seoul; Ji-Youn Han, Lung Cancer Centre, National Cancer Centre, Goyang, South Korea; Denis L. Moro-Sibilot, Service de Pneumologie; Michael Duruisseaux, Centre Hospitalier Universitaire de Grenoble, Grenoble; Eric Dansin, Centre Oscar Lambret, Lille, France; Lucio Crino, Santa Maria della Misericordia Hospital, Perugia; Tommaso De Pas, European Institute of Oncology, Milan, Italy; Antje Tessmer, Evangelische Lungenklinik Berlin, Berlin, Germany; James Chih-Hsin Yang, Graduate Institute of Oncology and Cancer Research Centre, National Taiwan University, Taipei, Taiwan; and Walter Bordogna, Sophie Golding, and Ali Zeaiter, F. Hoffmann-La Roche, Basel, Switzerland
| | - Ji-Youn Han
- Shirish M. Gadgeel, Karmanos Cancer Institute, Wayne State University, Detroit, MI; Alice T. Shaw, Massachusetts General Hospital; Leena Gandhi, Dana-Farber Cancer Institute, Boston, MA; Ramaswamy Govindan, Washington University School of Medicine, St Louis, MO; Mark A. Socinski, University of Pittsburgh, Pittsburgh, PA; D. Ross Camidge, University of Colorado Cancer Center, Denver, CO; Alberto Chiappori, Thoracic Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL; Sai-Hong Ignatius Ou, University of California at Irvine, Orange, CA; Luigi De Petris, Karolinska Institutet, Stockholm, Sweden; Dong-Wan Kim, Seoul National University Hospital, Seoul; Ji-Youn Han, Lung Cancer Centre, National Cancer Centre, Goyang, South Korea; Denis L. Moro-Sibilot, Service de Pneumologie; Michael Duruisseaux, Centre Hospitalier Universitaire de Grenoble, Grenoble; Eric Dansin, Centre Oscar Lambret, Lille, France; Lucio Crino, Santa Maria della Misericordia Hospital, Perugia; Tommaso De Pas, European Institute of Oncology, Milan, Italy; Antje Tessmer, Evangelische Lungenklinik Berlin, Berlin, Germany; James Chih-Hsin Yang, Graduate Institute of Oncology and Cancer Research Centre, National Taiwan University, Taipei, Taiwan; and Walter Bordogna, Sophie Golding, and Ali Zeaiter, F. Hoffmann-La Roche, Basel, Switzerland
| | - Walter Bordogna
- Shirish M. Gadgeel, Karmanos Cancer Institute, Wayne State University, Detroit, MI; Alice T. Shaw, Massachusetts General Hospital; Leena Gandhi, Dana-Farber Cancer Institute, Boston, MA; Ramaswamy Govindan, Washington University School of Medicine, St Louis, MO; Mark A. Socinski, University of Pittsburgh, Pittsburgh, PA; D. Ross Camidge, University of Colorado Cancer Center, Denver, CO; Alberto Chiappori, Thoracic Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL; Sai-Hong Ignatius Ou, University of California at Irvine, Orange, CA; Luigi De Petris, Karolinska Institutet, Stockholm, Sweden; Dong-Wan Kim, Seoul National University Hospital, Seoul; Ji-Youn Han, Lung Cancer Centre, National Cancer Centre, Goyang, South Korea; Denis L. Moro-Sibilot, Service de Pneumologie; Michael Duruisseaux, Centre Hospitalier Universitaire de Grenoble, Grenoble; Eric Dansin, Centre Oscar Lambret, Lille, France; Lucio Crino, Santa Maria della Misericordia Hospital, Perugia; Tommaso De Pas, European Institute of Oncology, Milan, Italy; Antje Tessmer, Evangelische Lungenklinik Berlin, Berlin, Germany; James Chih-Hsin Yang, Graduate Institute of Oncology and Cancer Research Centre, National Taiwan University, Taipei, Taiwan; and Walter Bordogna, Sophie Golding, and Ali Zeaiter, F. Hoffmann-La Roche, Basel, Switzerland
| | - Sophie Golding
- Shirish M. Gadgeel, Karmanos Cancer Institute, Wayne State University, Detroit, MI; Alice T. Shaw, Massachusetts General Hospital; Leena Gandhi, Dana-Farber Cancer Institute, Boston, MA; Ramaswamy Govindan, Washington University School of Medicine, St Louis, MO; Mark A. Socinski, University of Pittsburgh, Pittsburgh, PA; D. Ross Camidge, University of Colorado Cancer Center, Denver, CO; Alberto Chiappori, Thoracic Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL; Sai-Hong Ignatius Ou, University of California at Irvine, Orange, CA; Luigi De Petris, Karolinska Institutet, Stockholm, Sweden; Dong-Wan Kim, Seoul National University Hospital, Seoul; Ji-Youn Han, Lung Cancer Centre, National Cancer Centre, Goyang, South Korea; Denis L. Moro-Sibilot, Service de Pneumologie; Michael Duruisseaux, Centre Hospitalier Universitaire de Grenoble, Grenoble; Eric Dansin, Centre Oscar Lambret, Lille, France; Lucio Crino, Santa Maria della Misericordia Hospital, Perugia; Tommaso De Pas, European Institute of Oncology, Milan, Italy; Antje Tessmer, Evangelische Lungenklinik Berlin, Berlin, Germany; James Chih-Hsin Yang, Graduate Institute of Oncology and Cancer Research Centre, National Taiwan University, Taipei, Taiwan; and Walter Bordogna, Sophie Golding, and Ali Zeaiter, F. Hoffmann-La Roche, Basel, Switzerland
| | - Ali Zeaiter
- Shirish M. Gadgeel, Karmanos Cancer Institute, Wayne State University, Detroit, MI; Alice T. Shaw, Massachusetts General Hospital; Leena Gandhi, Dana-Farber Cancer Institute, Boston, MA; Ramaswamy Govindan, Washington University School of Medicine, St Louis, MO; Mark A. Socinski, University of Pittsburgh, Pittsburgh, PA; D. Ross Camidge, University of Colorado Cancer Center, Denver, CO; Alberto Chiappori, Thoracic Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL; Sai-Hong Ignatius Ou, University of California at Irvine, Orange, CA; Luigi De Petris, Karolinska Institutet, Stockholm, Sweden; Dong-Wan Kim, Seoul National University Hospital, Seoul; Ji-Youn Han, Lung Cancer Centre, National Cancer Centre, Goyang, South Korea; Denis L. Moro-Sibilot, Service de Pneumologie; Michael Duruisseaux, Centre Hospitalier Universitaire de Grenoble, Grenoble; Eric Dansin, Centre Oscar Lambret, Lille, France; Lucio Crino, Santa Maria della Misericordia Hospital, Perugia; Tommaso De Pas, European Institute of Oncology, Milan, Italy; Antje Tessmer, Evangelische Lungenklinik Berlin, Berlin, Germany; James Chih-Hsin Yang, Graduate Institute of Oncology and Cancer Research Centre, National Taiwan University, Taipei, Taiwan; and Walter Bordogna, Sophie Golding, and Ali Zeaiter, F. Hoffmann-La Roche, Basel, Switzerland
| | - Sai-Hong Ignatius Ou
- Shirish M. Gadgeel, Karmanos Cancer Institute, Wayne State University, Detroit, MI; Alice T. Shaw, Massachusetts General Hospital; Leena Gandhi, Dana-Farber Cancer Institute, Boston, MA; Ramaswamy Govindan, Washington University School of Medicine, St Louis, MO; Mark A. Socinski, University of Pittsburgh, Pittsburgh, PA; D. Ross Camidge, University of Colorado Cancer Center, Denver, CO; Alberto Chiappori, Thoracic Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL; Sai-Hong Ignatius Ou, University of California at Irvine, Orange, CA; Luigi De Petris, Karolinska Institutet, Stockholm, Sweden; Dong-Wan Kim, Seoul National University Hospital, Seoul; Ji-Youn Han, Lung Cancer Centre, National Cancer Centre, Goyang, South Korea; Denis L. Moro-Sibilot, Service de Pneumologie; Michael Duruisseaux, Centre Hospitalier Universitaire de Grenoble, Grenoble; Eric Dansin, Centre Oscar Lambret, Lille, France; Lucio Crino, Santa Maria della Misericordia Hospital, Perugia; Tommaso De Pas, European Institute of Oncology, Milan, Italy; Antje Tessmer, Evangelische Lungenklinik Berlin, Berlin, Germany; James Chih-Hsin Yang, Graduate Institute of Oncology and Cancer Research Centre, National Taiwan University, Taipei, Taiwan; and Walter Bordogna, Sophie Golding, and Ali Zeaiter, F. Hoffmann-La Roche, Basel, Switzerland
| |
Collapse
|
50
|
Genova C, Rijavec E, Biello F, Rossi G, Barletta G, Dal Bello MG, Vanni I, Coco S, Alama A, Grossi F. New systemic strategies for overcoming resistance to targeted therapies in non-small cell lung cancer. Expert Opin Pharmacother 2016; 18:19-33. [DOI: 10.1080/14656566.2016.1261109] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Affiliation(s)
- Carlo Genova
- Lung Cancer Unit, San Martino Hospital – National Institute for Cancer Research, Genova, Italy
| | - Erika Rijavec
- Lung Cancer Unit, San Martino Hospital – National Institute for Cancer Research, Genova, Italy
| | - Federica Biello
- Lung Cancer Unit, San Martino Hospital – National Institute for Cancer Research, Genova, Italy
| | - Giovanni Rossi
- Lung Cancer Unit, San Martino Hospital – National Institute for Cancer Research, Genova, Italy
| | - Giulia Barletta
- Lung Cancer Unit, San Martino Hospital – National Institute for Cancer Research, Genova, Italy
| | | | - Irene Vanni
- Lung Cancer Unit, San Martino Hospital – National Institute for Cancer Research, Genova, Italy
| | - Simona Coco
- Lung Cancer Unit, San Martino Hospital – National Institute for Cancer Research, Genova, Italy
| | - Angela Alama
- Lung Cancer Unit, San Martino Hospital – National Institute for Cancer Research, Genova, Italy
| | - Francesco Grossi
- Lung Cancer Unit, San Martino Hospital – National Institute for Cancer Research, Genova, Italy
| |
Collapse
|