1
|
Liu B, Lu Y, Lin R, Xu J, Shang Z, Hou X, Shao X, Pan Z, Yu T, Feng W. Preclinical studies of the falnidamol as a highly potent and specific active ABCB1 transporter inhibitor. BMC Cancer 2025; 25:24. [PMID: 39773145 PMCID: PMC11707883 DOI: 10.1186/s12885-024-13371-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2024] [Accepted: 12/19/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND ABCB1 overexpression is a key factor in causing multidrug resistance (MDR). As a result, it is crucial to discover effective medications against ABCB1 to overcome MDR. Falnidamol, a tyrosine kinase inhibitor (TKI) targeting the epidermal growth factor receptor (EGFR), is currently in phase 1 clinical trials for the treatment of solid tumors. In this work, we assessed whether falnidamol could act as an inhibitor of ABCB1 to reverse ABCB1-mediated MDR. METHODS The reversal effect of falnidamol on MDR was assessed by MTT, colony formation, 3D microsphere, and xenograft model assays. The protein expression or cellular localization was tested by western blot and immunofluorescence analysis. The intracellular doxorubicin accumulation and efflux were assessed by flow cytometry. The ATPase activity of ABCB1 was detected by a microplate reader. The interaction between falnidamol and ABCB1 was evaluated by docking analysis and cellular thermal shift assay. RESULTS Our data showed that falnidamol specifically reversed ABCB1-mediated MDR but not ABCG2-mediated MDR in vitro and in vivo. Mechanistic studies suggested falnidamol had no effect on ABCB1 expression or cellular localization, nor on the AKT or ERK pathways. Further studies found that falnidamol reduced ABCB1's efflux function, resulting in enhanced intracellular agent accumulation and thus overcoming MDR. ATPase assay showed that falnidamol suppressed the ABCB1 ATPase activity. Furthermore, docking analysis and cellular thermal shift assay indicated that falnidamol bound directly to the drug-binding site of ABCB1 transporter. CONCLUSION The present study proves that falnidamol acts as a highly potent and specific active ABCB1 transporter inhibitor, and can reverse ABCB1-mediated MDR, implying that combining falnidamol with ABCB1 substrate chemotherapeutic agents has the potential to overcome ABCB1-mediated MDR.
Collapse
Affiliation(s)
- Baojie Liu
- School of Life Science and Technology, Shandong Second Medical University, Weifang, Shandong, 261053, P.R. China
| | - Yongzheng Lu
- School of Life Science and Technology, Shandong Second Medical University, Weifang, Shandong, 261053, P.R. China
| | - Ruihui Lin
- School of Life Science and Technology, Shandong Second Medical University, Weifang, Shandong, 261053, P.R. China
| | - Junbao Xu
- Shandong Public Health Clinical Center, Shandong University, Jinan, Shandong, 250102, P.R. China
| | - Zilin Shang
- School of Life Science and Technology, Shandong Second Medical University, Weifang, Shandong, 261053, P.R. China
| | - Xinyu Hou
- School of Life Science and Technology, Shandong Second Medical University, Weifang, Shandong, 261053, P.R. China
| | - Xulong Shao
- School of Life Science and Technology, Shandong Second Medical University, Weifang, Shandong, 261053, P.R. China
| | - Zhifang Pan
- School of Life Science and Technology, Shandong Second Medical University, Weifang, Shandong, 261053, P.R. China.
| | - Tao Yu
- School of Life Science and Technology, Shandong Second Medical University, Weifang, Shandong, 261053, P.R. China.
- School of Medicine and Pharmacy, Ocean University of China, Qingdao, Shandong, 266003, China.
| | - Weiguo Feng
- School of Life Science and Technology, Shandong Second Medical University, Weifang, Shandong, 261053, P.R. China.
| |
Collapse
|
2
|
Nazari S, Poustforoosh A, Paul PR, Kukreti R, Tavakkoli M, Saso L, Firuzi O, Moosavi F. c-MET tyrosine kinase inhibitors reverse drug resistance mediated by the ATP-binding cassette transporter B1 (ABCB1) in cancer cells. 3 Biotech 2025; 15:2. [PMID: 39650809 PMCID: PMC11618280 DOI: 10.1007/s13205-024-04162-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2024] [Accepted: 11/10/2024] [Indexed: 12/11/2024] Open
Abstract
This study investigated the potential of MET kinase inhibitors, cabozantinib, crizotinib, and PHA665752, in reversing multidrug resistance (MDR) mediated by ABCB1 in cancer cells. The accumulation of the fluorescent probe, Rhodamine 123, was assessed using flow cytometry and fluorescence microscopy in MDR MES-SA/DX5 and parental cells. The growth inhibitory activity of MET inhibitors as monotherapies and in combination with chemotherapeutic drugs was evaluated by MTT assay. CalcuSyn software was used to analyze the combination index (CI) as an index of drug-drug interaction in combination treatments. Results showed that at concentrations of 5, and 25 μM, c-MET inhibitors significantly increased Rhodamine 123 accumulation in MDR cells, with ratios up to 17.8 compared to control cells, while exhibiting no effect in parental cells. Additionally, the combination of c-MET inhibitors with the chemotherapeutic agent doxorubicin synergistically enhanced cytotoxicity in MDR cells, as evidenced by combination index (CI) values of 0.54 ± 0.08, 0.69 ± 0.1, and 0.85 ± 0.07 for cabozantinib, crizotinib, and PHA665752, respectively. While all three c-MET inhibitors stimulated ABCB1 ATPase activity in different manners at certain concentrations, PHA-665752 suppressed it at high concentration. In silico analysis also suggested that the transmembrane domains (TMD) of ABCB1 transporters could be considered potential target for these agents. Our results suggest that c-MET inhibitors can serve as promising MDR reversal agents in ABCB1-medicated drug-resistant cancer cells.
Collapse
Affiliation(s)
- Somayeh Nazari
- Medicinal and Natural Products Chemistry Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Alireza Poustforoosh
- Medicinal and Natural Products Chemistry Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Priyanka Rani Paul
- Genomics and Molecular Medicine Unit, Institute of Genomics and Integrative Biology (IGIB), Council of Scientific and Industrial Research (CSIR), Mall Road, Delhi, 110007 India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002 India
| | - Ritushree Kukreti
- Genomics and Molecular Medicine Unit, Institute of Genomics and Integrative Biology (IGIB), Council of Scientific and Industrial Research (CSIR), Mall Road, Delhi, 110007 India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002 India
| | - Marjan Tavakkoli
- Medicinal and Natural Products Chemistry Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Luciano Saso
- Department of Physiology and Pharmacology “Vittorio Erspamer”, Sapienza University of Rome, P. le Aldo Moro 5, 00185 Rome, Italy
| | - Omidreza Firuzi
- Medicinal and Natural Products Chemistry Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Fatemeh Moosavi
- Medicinal and Natural Products Chemistry Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
3
|
Song X, Zhou Z, Elmezayen A, Wu R, Yu C, Gao B, Minna JD, Westover KD, Zeh HJ, Kroemer G, Heasley LE, Kang R, Tang D. SRC kinase drives multidrug resistance induced by KRAS-G12C inhibition. SCIENCE ADVANCES 2024; 10:eadq4274. [PMID: 39661665 PMCID: PMC11633746 DOI: 10.1126/sciadv.adq4274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Accepted: 11/04/2024] [Indexed: 12/13/2024]
Abstract
Direct targeting of the KRAS-G12C-mutant protein using covalent inhibitors (G12Ci) acts on human non-small cell lung cancer (NSCLC). However, drug resistance is an emerging concern in this approach. Here, we show that MRTX849, a covalent inhibitor targeting the KRAS-G12C mutation, leads to the reactivation of the mitogen-activated protein kinase signaling pathway in MRTX849-resistant NSCLC and pancreatic ductal adenocarcinoma. A genome-wide CRISPR screen revealed that the adenosine triphosphate binding cassette transporter ABCC1 mediates MRTX849 resistance. Functional studies demonstrated that the transcription factor JUN drives ABCC1 expression, resulting in multidrug resistance. An unbiased drug screen identified the tyrosine kinase inhibitor dasatinib that potentiates MRTX849 efficacy by inhibiting SRC-dependent JUN activation, avoiding multidrug resistance and tumor suppression in vitro as well as in suitable preclinical mouse models and patient-derived organoids. SRC inhibitors (DGY-06-116, dasatinib, and bosutinib) also exhibit synergistic effects with MRTX849 in eliminating various tumor cell lines carrying KRAS-G12C mutations. Thus, SRC inhibitors amplify the therapeutic utility of G12Ci.
Collapse
Affiliation(s)
- Xinxin Song
- Department of Surgery, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Zhuan Zhou
- Department of Surgery, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Ammar Elmezayen
- Departments of Biochemistry and Radiation Oncology, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Runliu Wu
- Department of Surgery, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Chunhua Yu
- Department of Surgery, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Boning Gao
- Hamon Center for Therapeutic Oncology Research, Department of Pharmacology, Department of Internal Medicine, Harold C. Simmons Comprehensive Cancer Center, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - John D. Minna
- Hamon Center for Therapeutic Oncology Research, Department of Pharmacology, Department of Internal Medicine, Harold C. Simmons Comprehensive Cancer Center, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Kenneth D. Westover
- Departments of Biochemistry and Radiation Oncology, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Herbert J. Zeh
- Department of Surgery, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Guido Kroemer
- Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université de Paris, Sorbonne Université, INSERM U1138, Institut Universitaire de France, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France
- Pôle de Biologie, Hôpital Européen Georges Pompidou, AP-HP, Paris, France
| | - Lynn E. Heasley
- Department of Craniofacial Biology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
- Eastern Colorado VA Healthcare System, Rocky Mountain Regional VA Medical Center, Aurora, CO 80045, USA
| | - Rui Kang
- Department of Surgery, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Daolin Tang
- Department of Surgery, UT Southwestern Medical Center, Dallas, TX 75390, USA
| |
Collapse
|
4
|
Akbarzadeh MA, Vaez-Gharamaleki Y, Hosseini MS. Mechanisms of resistance to histone deacetylase inhibitors in acute leukemia. Ther Adv Hematol 2024; 15:20406207241306553. [PMID: 39669704 PMCID: PMC11635846 DOI: 10.1177/20406207241306553] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2024] Open
Affiliation(s)
- Mohammad Amin Akbarzadeh
- Research Center for Evidence-Based Medicine, Iranian EBM Center: A JBI Center of Excellence, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Yosra Vaez-Gharamaleki
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad-Salar Hosseini
- Research Center for Integrative Medicine in Aging, Aging Research Institute, Tabriz University of Medical Sciences, Golgasht Street, Tabriz 51666, Iran
- Research Center for Evidence-Based Medicine, Iranian EBM Center: A JBI Center of Excellence, Tabriz University of Medical Sciences, Tabriz, Iran
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
5
|
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
|
6
|
Zhang M, Zeng X, She M, Dong X, Chen J, Xiong Q, Qiu G, Yang S, Li X, Ren G. FRAX486, a PAK inhibitor, overcomes ABCB1-mediated multidrug resistance in breast cancer cells. Braz J Med Biol Res 2024; 57:e13357. [PMID: 38958364 PMCID: PMC11221864 DOI: 10.1590/1414-431x2024e13357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Accepted: 05/06/2024] [Indexed: 07/04/2024] Open
Abstract
The overexpression of P-glycoprotein (P-gp/ABCB1) is a leading cause of multidrug resistance (MDR). Hence, it is crucial to discover effective pharmaceuticals that counteract ABCB1-mediated multidrug resistance. FRAX486 is a p21-activated kinase (PAK) inhibitor. The objective of this study was to investigate whether FRAX486 can reverse ABCB1-mediated multidrug resistance, while also exploring its mechanism of action. The CCK8 assay demonstrated that FRAX486 significantly reversed ABCB1-mediated multidrug resistance. Furthermore, western blotting and immunofluorescence experiments revealed that FRAX486 had no impact on expression level and intracellular localization of ABCB1. Notably, FRAX486 was found to enhance intracellular drug accumulation and reduce efflux, resulting in the reversal of multidrug resistance. Docking analysis also indicated a strong affinity between FRAX486 and ABCB1. This study highlights the ability of FRAX486 to reverse ABCB1-mediated multidrug resistance and provides valuable insights for its clinical application.
Collapse
Affiliation(s)
- Meng Zhang
- Department of Thyroid and Breast Surgery, Shenzhen Hospital of Southern Medical University, Shenzhen, Guangdong, China
| | - Xiaoqi Zeng
- Department of Thyroid and Breast Surgery, Shenzhen Hospital of Southern Medical University, Shenzhen, Guangdong, China
| | - Meiling She
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Xingduo Dong
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, New York, USA
| | - Jun Chen
- Department of Thyroid and Breast Surgery, Shenzhen Hospital of Southern Medical University, Shenzhen, Guangdong, China
| | - Qingquan Xiong
- Department of Thyroid and Breast Surgery, Shenzhen Hospital of Southern Medical University, Shenzhen, Guangdong, China
| | - Guobin Qiu
- Department of Thyroid and Breast Surgery, Shenzhen Hospital of Southern Medical University, Shenzhen, Guangdong, China
| | - Shuyi Yang
- Department of Thyroid and Breast Surgery, Shenzhen Hospital of Southern Medical University, Shenzhen, Guangdong, China
| | - Xiangqi Li
- Department of Breast Surgery, The Second Affiliated Hospital of Shandong First Medical University, Tai'an, Shandong, China
| | - Guanghui Ren
- Department of Thyroid and Breast Surgery, Shenzhen Hospital of Southern Medical University, Shenzhen, Guangdong, China
| |
Collapse
|
7
|
Dong XD, Lu Q, Li YD, Cai CY, Teng QX, Lei ZN, Wei ZH, Yin F, Zeng L, Chen ZS. RN486, a Bruton's Tyrosine Kinase inhibitor, antagonizes multidrug resistance in ABCG2-overexpressing cancer cells. J Transl Int Med 2024; 12:288-298. [PMID: 39081282 PMCID: PMC11284896 DOI: 10.2478/jtim-2024-0011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/02/2024] Open
Abstract
Background and Objectives Overcoming ATP-binding cassette subfamily G member 2 (ABCG2)-mediated multidrug resistance (MDR) has attracted the attention of scientists because one of the critical factors resulting in MDR in cancer is the overexpression of ABCG2. RN486, a Bruton's Tyrosine Kinase (BTK) inhibitor, was discovered to potentially reverse ABCB1-mediated MDR. However, there is still uncertainty about whether RN486 has a reversal off-target impact on ABCG2-mediated MDR. Methods MTT assay was used to detect the reversal effect of RN486 on ABCG2-overexpressing cancer cells. The ABCG2 expression level and subcellular localization were examined by Western blotting and immunofluorescence. Drug accumulation and eflux assay and ATPase assay were performed to analyze the ABCG2 transporter function and ATPase activity. Molecular modeling predicted the binding between RN486 and ABCG2 protein. Results Non-toxic concentrations of RN486 remarkably increased the sensitivity of ABCG2-overexpressing cancer cells to conventional anticancer drugs mitoxantrone and topotecan. The reversal mechanistic studies showed that RN486 elevated the drug accumulation because of reducing the eflux of ABCG2 substrate drug in ABCG2-overexpressing cancer cells. In addition, the inhibitory efect of RN486 on ABCG2-associated ATPase activity was also verified. Molecular docking study implied a strong binding afinity between RN486 and ABCG2 transporter. Meanwhile, the ABCG2 subcellular localization was not altered by the treatment of RN486, but the expression level of ABCG2 was down-regulated. Conclusions Our studies propose that RN486 can antagonize ABCG2-mediated MDR in cancer cells via down-regulating the expression level of ABCG2 protein, reducing ATPase activity of ABCG2 transporter, and inhibiting the transporting function. RN486 could be potentially used in conjunction with chemotherapy to alleviate MDR mediated by ABCG2 in cancer.
Collapse
Affiliation(s)
- Xing-Duo Dong
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY 11439, USA
| | - Qisi Lu
- Department of Hematology, Foresea Life Insurance Guangzhou General Hospital, Guangzhou515500, Guangdong Province, China
| | - Yi-Dong Li
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY 11439, USA
| | - Chao-Yun Cai
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY 11439, USA
| | - Qiu-Xu Teng
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY 11439, USA
| | - Zi-Ning Lei
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY 11439, USA
- Guangdong Provincial Key Laboratory of Digestive Cancer Research, Digestive Diseases Center, Biobank, Precision Medicine Center, Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen518107, Guangdong Province, China
| | - Zeng-Hui Wei
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY 11439, USA
| | - Fan Yin
- Department of Statistics, University of California at Irvine, Irvine, CA 92697, USA
| | - Leli Zeng
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY 11439, USA
- Guangdong Provincial Key Laboratory of Digestive Cancer Research, Digestive Diseases Center, Biobank, Precision Medicine Center, Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen518107, Guangdong Province, China
| | - Zhe-Sheng Chen
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY 11439, USA
| |
Collapse
|
8
|
Ash LJ, Busia-Bourdain O, Okpattah D, Kamel A, Liberchuk A, Wolfe AL. KRAS: Biology, Inhibition, and Mechanisms of Inhibitor Resistance. Curr Oncol 2024; 31:2024-2046. [PMID: 38668053 PMCID: PMC11049385 DOI: 10.3390/curroncol31040150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 03/29/2024] [Accepted: 04/01/2024] [Indexed: 04/28/2024] Open
Abstract
KRAS is a small GTPase that is among the most commonly mutated oncogenes in cancer. Here, we discuss KRAS biology, therapeutic avenues to target it, and mechanisms of resistance that tumors employ in response to KRAS inhibition. Several strategies are under investigation for inhibiting oncogenic KRAS, including small molecule compounds targeting specific KRAS mutations, pan-KRAS inhibitors, PROTACs, siRNAs, PNAs, and mutant KRAS-specific immunostimulatory strategies. A central challenge to therapeutic effectiveness is the frequent development of resistance to these treatments. Direct resistance mechanisms can involve KRAS mutations that reduce drug efficacy or copy number alterations that increase the expression of mutant KRAS. Indirect resistance mechanisms arise from mutations that can rescue mutant KRAS-dependent cells either by reactivating the same signaling or via alternative pathways. Further, non-mutational forms of resistance can take the form of epigenetic marks, transcriptional reprogramming, or alterations within the tumor microenvironment. As the possible strategies to inhibit KRAS expand, understanding the nuances of resistance mechanisms is paramount to the development of both enhanced therapeutics and innovative drug combinations.
Collapse
Affiliation(s)
- Leonard J. Ash
- Department of Biological Sciences, Hunter College, City University of New York, New York, NY 10065, USA
- Molecular, Cellular, and Developmental Biology Subprogram of the Biology Ph.D. Program, Graduate Center, City University of New York, New York, NY 10031, USA
| | - Ottavia Busia-Bourdain
- Department of Biological Sciences, Hunter College, City University of New York, New York, NY 10065, USA
| | - Daniel Okpattah
- Biochemistry Ph.D. Program, Graduate Center, City University of New York, New York, NY 10031, USA
| | - Avrosina Kamel
- Department of Biological Sciences, Hunter College, City University of New York, New York, NY 10065, USA
- Macaulay Honors College, Hunter College, City University of New York, New York, NY 10065, USA
| | - Ariel Liberchuk
- Department of Biological Sciences, Hunter College, City University of New York, New York, NY 10065, USA
- Macaulay Honors College, Hunter College, City University of New York, New York, NY 10065, USA
| | - Andrew L. Wolfe
- Department of Biological Sciences, Hunter College, City University of New York, New York, NY 10065, USA
- Molecular, Cellular, and Developmental Biology Subprogram of the Biology Ph.D. Program, Graduate Center, City University of New York, New York, NY 10031, USA
- Biochemistry Ph.D. Program, Graduate Center, City University of New York, New York, NY 10031, USA
- Department of Pharmacology, Weill Cornell Medicine, New York, NY 10021, USA
| |
Collapse
|
9
|
Zhang M, Huang MN, Dong XD, Cui QB, Yan Y, She ML, Feng WG, Zhao XS, Wang DT. Overexpression of ABCB1 confers resistance to FLT3 inhibitor FN-1501 in cancer cells: in vitro and in vivo characterization. Am J Cancer Res 2023; 13:6026-6037. [PMID: 38187048 PMCID: PMC10767331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Accepted: 11/22/2023] [Indexed: 01/09/2024] Open
Abstract
FN-1501 is a potent FLT3 inhibitor with antitumor activity. A phase 1 trial of FN-1501 monotherapy in patients with advanced solid tumors and R/R AML is in progress. Since one of the primary causes of multidrug resistance (MDR) is the overexpression of ATP-binding cassette superfamily B member 1 (ABCB1), the objective of this study was to investigate the potential relationship between FN-1501 and the ABCB1 transporter. We found ABCB1 overexpressing-cancer cells conferred FN-1501 resistance, which could be reversed by an ABCB1 inhibitor. Molecular docking study revealed that FN-1501 docked the ligand binding site with an affinity score of -9.77 kcal/mol, denoting a strong interaction between FN-1501 and ABCB1. Additionally, the ABCB1 ATPase assay indicated that FN-1501 could significantly stimulate ABCB1 ATPase activity. Furthermore, we observed a similar trend of ABCB1-facilated FN-1501 resistance in tumor-bearing mice model. In sum, we demonstrate that FN-1501 is a substrate of ABCB1 transporter from both in vivo and in vitro studies. Therefore, our findings provide new insight on the mechanism of chemoresistance due to ABCB1 overexpression.
Collapse
Affiliation(s)
- Meng Zhang
- Department of Thyroid and Breast Surgery, Shenzhen Hospital of Southern Medical UniversityNo. 1333 Xinhu Road, Baoan, Shenzhen 510000, Guangdong, China
| | - Min-Na Huang
- Dalton Cardiovascular Research Center, Department of Medical Pharmacology and Physiology, University of Missouri, School of Medicine134 Research Park Dr, Columbia, MO 65211, USA
| | - Xing-Duo Dong
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John’s UniversityQueens, NY 11439, USA
| | - Qing-Bin Cui
- Department of Cancer Biology, University of Toledo College of Medicine and Life SciencesToledo, OH 43606, USA
| | - Yan Yan
- Department of Traditional Chinese Medicine, Shenzhen Hospital of Southern Medical UniversityNo. 1333 Xinhu Road, Baoan, Shenzhen 510000, Guangdong, China
| | - Mei-Ling She
- Department of Traditional Chinese Medicine, Shenzhen Hospital of Southern Medical UniversityNo. 1333 Xinhu Road, Baoan, Shenzhen 510000, Guangdong, China
| | - Wei-Guo Feng
- School of Life Science and Technology, Weifang Medical UniversityNo. 7166 Baotong Street, Weicheng, Weifang 261053, Shandong, China
| | - Xiao-Shan Zhao
- School of Traditional Chinese Medicine, Southern Medical UniversityNo. 1023 Satai South Road, Baiyun, Guangzhou 510000, Guangdong, China
| | - Dong-Tao Wang
- Department of Traditional Chinese Medicine, Shenzhen Hospital of Southern Medical UniversityNo. 1333 Xinhu Road, Baoan, Shenzhen 510000, Guangdong, China
- School of Traditional Chinese Medicine, Southern Medical UniversityNo. 1023 Satai South Road, Baiyun, Guangzhou 510000, Guangdong, China
| |
Collapse
|
10
|
Ye Q, Zhou X, Ren H, Han F, Lin R, Li J. An overview of the past decade of bufalin in the treatment of refractory and drug-resistant cancers: current status, challenges, and future perspectives. Front Pharmacol 2023; 14:1274336. [PMID: 37860119 PMCID: PMC10582727 DOI: 10.3389/fphar.2023.1274336] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Accepted: 09/25/2023] [Indexed: 10/21/2023] Open
Abstract
Profound progress has been made in cancer treatment in the past three decades. However, drug resistance remains prevalent and a critical challenge. Drug resistance can be attributed to oncogenes mutations, activated defensive mechanisms, ATP-bind cassette transporters overexpression, cancer stem cells, etc. Chinese traditional medicine toad venom has been used for centuries for different diseases, including resistant cancers. Bufalin is one of the bufadienolides in toad venom that has been extensively studied for its potential in refractory and drug-resistant cancer treatments in vitro and in vivo. In this work, we would like to critically review the progress made in the past decade (2013-2022) of bufalin in overcoming drug resistance in cancers. Generally, bufalin shows high potential in killing certain refractory and resistant cancer cells via multiple mechanisms. More importantly, bufalin can work as a chemo-sensitizer that enhances the sensitivity of certain conventional and targeted therapies at low concentrations. In addition, the development of bufalin derivatives was also briefly summarized and discussed. We also analyzed the obstacles and challenges and provided possible solutions for future perspectives. We hope that the collective information may help evoke more effort for more in-depth studies and evaluation of bufalin in both lab and possible clinical trials.
Collapse
Affiliation(s)
- Qingmei Ye
- Hainan General Hospital & Hainan Affiliated Hospital of Hainan Medical University, Haikou, Hainan, China
- Key Laboratory of Tropical Medicinal Resource Chemistry of Ministry of Education, Key Laboratory of Tropical Medicinal Plant Chemistry of Hainan Province, College of Chemistry and Chemical Engineering, Hainan Normal University, Haikou, Hainan, China
| | - Xin Zhou
- The Fifth People’s Hospital of Hainan Province & Affiliated Dermatology Hospital of Hainan Medical University, Haikou, Hainan, China
| | - Han Ren
- Hainan General Hospital & Hainan Affiliated Hospital of Hainan Medical University, Haikou, Hainan, China
| | - Fangxuan Han
- Hainan General Hospital & Hainan Affiliated Hospital of Hainan Medical University, Haikou, Hainan, China
| | - Rong Lin
- Hubei Province Key Laboratory of Traditional Chinese Medicine Resource and Chemistry, Department of Pharmacy, Hubei University of Chinese Medicine, Wuhan, Hubei, China
| | - Juan Li
- Hubei Province Key Laboratory of Traditional Chinese Medicine Resource and Chemistry, Department of Pharmacy, Hubei University of Chinese Medicine, Wuhan, Hubei, China
| |
Collapse
|
11
|
Ghosh S, Fan F, Powell RT, Roszik J, Park Y, Stephan C, Sebastian M, Tan L, Sorokin AV, Lorenzi PL, Kopetz S, Ellis LM, Bhattacharya R. Vincristine Enhances the Efficacy of MEK Inhibitors in Preclinical Models of KRAS-mutant Colorectal Cancer. Mol Cancer Ther 2023; 22:962-975. [PMID: 37310170 PMCID: PMC11991686 DOI: 10.1158/1535-7163.mct-23-0110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 05/03/2023] [Accepted: 06/09/2023] [Indexed: 06/14/2023]
Abstract
Mutations in KRAS are found in more than 50% of tumors from patients with metastatic colorectal cancer (mCRC). However, direct targeting of most KRAS mutations is difficult; even the recently developed KRASG12C inhibitors failed to show significant benefit in patients with mCRC. Single agents targeting mitogen-activated protein kinase kinase (MEK), a downstream mediator of RAS, have also been ineffective in colorectal cancer. To identify drugs that can enhance the efficacy of MEK inhibitors, we performed unbiased high-throughput screening using colorectal cancer spheroids. We used trametinib as the anchor drug and examined combinations of trametinib with the NCI-approved Oncology Library version 5. The initial screen, and following focused validation screens, identified vincristine as being strongly synergistic with trametinib. In vitro, the combination strongly inhibited cell growth, reduced clonogenic survival, and enhanced apoptosis compared with monotherapies in multiple KRAS-mutant colorectal cancer cell lines. Furthermore, this combination significantly inhibited tumor growth, reduced cell proliferation, and increased apoptosis in multiple KRAS-mutant patient-derived xenograft mouse models. In vivo studies using drug doses that reflect clinically achievable doses demonstrated that the combination was well tolerated by mice. We further determined that the mechanism underlying the synergistic effect of the combination was due to enhanced intracellular accumulation of vincristine associated with MEK inhibition. The combination also significantly decreased p-mTOR levels in vitro, indicating that it inhibits both RAS-RAF-MEK and PI3K-AKT-mTOR survival pathways. Our data thus provide strong evidence that the combination of trametinib and vincristine represents a novel therapeutic option to be studied in clinical trials for patients with KRAS-mutant mCRC. SIGNIFICANCE Our unbiased preclinical studies have identified vincristine as an effective combination partner for the MEK inhibitor trametinib and provide a novel therapeutic option to be studied in patients with KRAS-mutant colorectal cancer.
Collapse
Affiliation(s)
- Susmita Ghosh
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center
| | - Fan Fan
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center
| | - Reid T. Powell
- Center for Translational Cancer Research, Texas A&M College of Medicine
| | - Jason Roszik
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center
| | - Yongsung Park
- Center for Translational Cancer Research, Texas A&M College of Medicine
| | - Clifford Stephan
- Center for Translational Cancer Research, Texas A&M College of Medicine
| | - Manu Sebastian
- Department of Veterinary Medicine & Surgery, The University of Texas MD Anderson Cancer Center
| | - Lin Tan
- Department of Metabolomics Core Facility-Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center
| | - Alexey V. Sorokin
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center
| | - Philip L. Lorenzi
- Department of Metabolomics Core Facility-Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center
| | - Scott Kopetz
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center
| | - Lee M. Ellis
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center
| | - Rajat Bhattacharya
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center
| |
Collapse
|
12
|
Lei Z, Tian Q, Teng Q, Wurpel JND, Zeng L, Pan Y, Chen Z. Understanding and targeting resistance mechanisms in cancer. MedComm (Beijing) 2023; 4:e265. [PMID: 37229486 PMCID: PMC10203373 DOI: 10.1002/mco2.265] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 03/05/2023] [Accepted: 03/23/2023] [Indexed: 05/27/2023] Open
Abstract
Resistance to cancer therapies has been a commonly observed phenomenon in clinical practice, which is one of the major causes of treatment failure and poor patient survival. The reduced responsiveness of cancer cells is a multifaceted phenomenon that can arise from genetic, epigenetic, and microenvironmental factors. Various mechanisms have been discovered and extensively studied, including drug inactivation, reduced intracellular drug accumulation by reduced uptake or increased efflux, drug target alteration, activation of compensatory pathways for cell survival, regulation of DNA repair and cell death, tumor plasticity, and the regulation from tumor microenvironments (TMEs). To overcome cancer resistance, a variety of strategies have been proposed, which are designed to enhance the effectiveness of cancer treatment or reduce drug resistance. These include identifying biomarkers that can predict drug response and resistance, identifying new targets, developing new targeted drugs, combination therapies targeting multiple signaling pathways, and modulating the TME. The present article focuses on the different mechanisms of drug resistance in cancer and the corresponding tackling approaches with recent updates. Perspectives on polytherapy targeting multiple resistance mechanisms, novel nanoparticle delivery systems, and advanced drug design tools for overcoming resistance are also reviewed.
Collapse
Affiliation(s)
- Zi‐Ning Lei
- PrecisionMedicine CenterScientific Research CenterThe Seventh Affiliated HospitalSun Yat‐Sen UniversityShenzhenP. R. China
- Department of Pharmaceutical SciencesCollege of Pharmacy and Health SciencesSt. John's UniversityQueensNew YorkUSA
| | - Qin Tian
- PrecisionMedicine CenterScientific Research CenterThe Seventh Affiliated HospitalSun Yat‐Sen UniversityShenzhenP. R. China
| | - Qiu‐Xu Teng
- Department of Pharmaceutical SciencesCollege of Pharmacy and Health SciencesSt. John's UniversityQueensNew YorkUSA
| | - John N. D. Wurpel
- Department of Pharmaceutical SciencesCollege of Pharmacy and Health SciencesSt. John's UniversityQueensNew YorkUSA
| | - Leli Zeng
- PrecisionMedicine CenterScientific Research CenterThe Seventh Affiliated HospitalSun Yat‐Sen UniversityShenzhenP. R. China
| | - Yihang Pan
- PrecisionMedicine CenterScientific Research CenterThe Seventh Affiliated HospitalSun Yat‐Sen UniversityShenzhenP. R. China
| | - Zhe‐Sheng Chen
- Department of Pharmaceutical SciencesCollege of Pharmacy and Health SciencesSt. John's UniversityQueensNew YorkUSA
| |
Collapse
|
13
|
Cui Q, Liang XL, Wang JQ, Zhang JY, Chen ZS. Therapeutic implication of carbon monoxide in drug resistant cancers. Biochem Pharmacol 2022; 201:115061. [PMID: 35489394 DOI: 10.1016/j.bcp.2022.115061] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Revised: 04/19/2022] [Accepted: 04/20/2022] [Indexed: 12/14/2022]
Abstract
Drug resistance is the major obstacle that undermines effective cancer treatment. Recently, the application of gas signaling molecules, e.g., carbon monoxide (CO), in overcoming drug resistance has gained significant attention. Growing evidence showed that CO could inhibit mitochondria respiratory effect and glycolysis, two major ATP production pathways in cancer cells, and suppress angiogenesis and inhibit the activity of cystathionine β-synthase that is important in regulating cancer cells homeostasis, leading to synergistic effects when combined with cisplatin, doxorubicin, or phototherapy, etc. in certain resistant cancer cells. In the current review, we attempted to have a summary of these research conducted in the past decade using CO in treating drug resistant cancers, and have a detailed interpretation of the underlying mechanisms. The critical challenges will be discussed and potential solutions will also be provided. The information collected in this work will hopefully evoke more effects in using CO for the treatment of drug resistant cancers.
Collapse
Affiliation(s)
- Qingbin Cui
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, The NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou 511436, China
| | - Xiao-Lan Liang
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, The NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou 511436, China
| | - Jing-Quan Wang
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY 11439, USA
| | - Jian-Ye Zhang
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, The NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou 511436, China.
| | - Zhe-Sheng Chen
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY 11439, USA; Institute for Biotechnology, St. John's University, Queens, NY 11439, USA.
| |
Collapse
|