1
|
Oh Y, An DE, Park J, Koh B, Cho KJ, Jeon H. Synthesis and evaluation of KX-01 analogs with an exploration of linker attachment points for antibody-drug conjugates. Bioorg Med Chem Lett 2025; 120:130114. [PMID: 39880175 DOI: 10.1016/j.bmcl.2025.130114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2024] [Revised: 12/26/2024] [Accepted: 01/26/2025] [Indexed: 01/31/2025]
Abstract
KX-01 (tirbanibulin, Klisyri®) is a recently FDA-approved drug for treating actinic keratosis, with a distinct dual mechanism of action combining microtubule disruption and non-ATP-competitive Src inhibition. This unique mechanism and novel chemotype highlight KX-01's potential as a payload for antibody-drug conjugates. In this study, we synthesized and evaluated KX-01 derivatives to enhance anticancer potency and explore functional groups suitable for antibody conjugation. Notably, replacing the morpholine group with an N-benzoylpiperazine scaffold resulted in an analog with significantly improved in vitro antiproliferative activity, attributed to enhanced microtubule disruption and Src inhibition. Furthermore, introducing a phenol or aniline functionality as a common linker attachment point preserved substantial cytotoxicity. These results suggest the potential of KX-01 derivatives for future use as ADC payloads.
Collapse
Affiliation(s)
- Yeju Oh
- Therapeutics & Biotechnology Division, Korea Research Institute of Chemical Technology, Daejeon 34114 Republic of Korea; Medicinal Chemistry & Pharmacology, University of Science & Technology, Daejeon 34113 Republic of Korea
| | - Da Eun An
- Therapeutics & Biotechnology Division, Korea Research Institute of Chemical Technology, Daejeon 34114 Republic of Korea
| | - Jaebeom Park
- Therapeutics & Biotechnology Division, Korea Research Institute of Chemical Technology, Daejeon 34114 Republic of Korea
| | - Byumseok Koh
- Therapeutics & Biotechnology Division, Korea Research Institute of Chemical Technology, Daejeon 34114 Republic of Korea; Medicinal Chemistry & Pharmacology, University of Science & Technology, Daejeon 34113 Republic of Korea
| | - Kyung-Jin Cho
- Therapeutics & Biotechnology Division, Korea Research Institute of Chemical Technology, Daejeon 34114 Republic of Korea.
| | - Hongjun Jeon
- Therapeutics & Biotechnology Division, Korea Research Institute of Chemical Technology, Daejeon 34114 Republic of Korea; Medicinal Chemistry & Pharmacology, University of Science & Technology, Daejeon 34113 Republic of Korea.
| |
Collapse
|
2
|
Valenti M, Bianco M, Narcisi A, Costanzo A, Borroni R, Ardigò M. Topical Pharmacological Treatment of Actinic Keratoses: Focus on Tirbanibulin 1% Ointment. Dermatol Pract Concept 2024; 14:dpc.1403S1a145S. [PMID: 39133636 PMCID: PMC11566729 DOI: 10.5826/dpc.1403s1a145s] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/22/2024] [Indexed: 11/17/2024] Open
Abstract
Actinic keratosis (AK) is a frequent precancerous skin lesion that mostly affects chronically sun-exposed areas. Chronic sun damage leads to various mutations in onco-suppressor and oncogenic genes which cause an uncontrolled proliferation of atypical keratinocytes. Untreated AKs may evolve in cutaneous squamous cell carcinoma (cSCC), with the consequent need for dermato-surgical excision or even for systemic immunotherapy in case of invasive/metastatic cSCCs. Epidemiology data on AK prevalence are various, however, the literature unanimously reports an increasing prevalence due to the aging of the population. Clinically AKs appear as a scaly, erythematous macule or papule or hyperkeratotic plaque. Management of AKs and the field of cancerization is important to avoid the natural evolution into squamous cell carcinomas (SCCs). Both physical and topical treatments are approved for managing AKs. Patient compliance with topical regimens is usually low due to the length of the posology and frequent skin adverse events. A recently approved tirbanibulin-based ointment, showed potential for inhibiting cell proliferation and blocking SRC-kinases, implicated in the progression of AKs in SCCs. The advantage of this new treatment is the practical posology, with a daily application for 5 consecutive days on AKs of the face-scalp area. Local skin reactions are usually mild and do not require treatment discontinuation. The short course of this new therapy and its excellent tolerance massively increased patient compliance. This article reviews what is currently known about this new therapy from its mechanism of action to clinical trial outcomes regarding safety, effectiveness, and patient adherence to the treatment.
Collapse
Affiliation(s)
- Mario Valenti
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Italy
- Dermatology Unit, IRCCS Humanitas Research Hospital, Rozzano, Italy
| | - Matteo Bianco
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Italy
- Dermatology Unit, IRCCS Humanitas Research Hospital, Rozzano, Italy
| | - Alessandra Narcisi
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Italy
- Dermatology Unit, IRCCS Humanitas Research Hospital, Rozzano, Italy
| | - Antonio Costanzo
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Italy
- Dermatology Unit, IRCCS Humanitas Research Hospital, Rozzano, Italy
| | - Riccardo Borroni
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Italy
- Dermatology Unit, IRCCS Humanitas Research Hospital, Rozzano, Italy
| | - Marco Ardigò
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Italy
- Dermatology Unit, IRCCS Humanitas Research Hospital, Rozzano, Italy
| |
Collapse
|
3
|
Tang L, Chen Z, Yang J, Li Q, Wang S, Mo T, Zeng W, Ding H, Pan S. Single-cell and Bulk RNA-Seq reveal angiogenic heterogeneity and microenvironmental features to evaluate prognosis and therapeutic response in lung adenocarcinoma. Front Immunol 2024; 15:1352893. [PMID: 38390340 PMCID: PMC10882092 DOI: 10.3389/fimmu.2024.1352893] [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] [Received: 12/09/2023] [Accepted: 01/23/2024] [Indexed: 02/24/2024] Open
Abstract
Background Angiogenesis stands as a pivotal hallmark in lung adenocarcinoma (LUAD), intricately shaping the tumor microenvironment (TME) and influencing LUAD progression. It emerges as a promising therapeutic target for LUAD, affecting patients' prognosis. However, its role in TME, LUAD prognosis, and its clinical applicability remain shrouded in mystery. Methods We employed integrated single-cell and bulk transcriptome sequencing to unravel the heterogeneity of angiogenesis within LUAD cells. Through "consensus clustering", we delineated distinct angiogenic clusters and deciphered their TME features. "Monocle2" was used to unravel divergent trajectories within malignant cell subpopulations of LUAD. Additionally, regulon submodules and specific cellular communication patterns of cells in different angiogenic states were analyzed by "pyscenic" and "Cellchat" algorithms. The "univariate Cox" and "LASSO" algorithms were applied to build angiogenic prognostic models. Immunohistochemistry (IHC) on clinical samples validated the role of model factors in LUAD angiogenesis. We utilized CTRP 2.0 and PRISM databases for pinpointing sensitive drugs against lung adenocarcinoma. Results Two clusters for the activation of angiogenesis were identified, with Cluster 1 showing a poor prognosis and a pro-cancerous TME. Three differentiated states of malignant epithelial LUAD cells were identified, which had different degrees of angiogenic activation, were regulated by three different regulon submodules, and had completely different crosstalk from other cells in TME. The experiments validate that SLC2A1 promotes angiogenesis in LUAD. ARS (Angiogenesis related score) had a high prognostic value; low ARSs showed immunotherapy benefits, whereas high ARSs were sensitive to 15 chemotherapeutic agents. Conclusion The assessment of angiogenic clusters helps to determine the prognostic and TME characteristics of LUAD. Angiogenic prognostic models can be used to assess the prognosis, immunotherapeutic response, and chemotherapeutic drug sensitivity of LUAD.
Collapse
Affiliation(s)
- Lijuan Tang
- Dalian Medical University, Dalian, China
- Department of Pathology, Affiliated Hospital of Nantong University, Nantong, China
| | - Zhike Chen
- Department of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Jian Yang
- Department of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Qifan Li
- Department of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Sichu Wang
- Dalian Medical University, Dalian, China
- Department of Pathology, Affiliated Hospital of Nantong University, Nantong, China
| | - Taoming Mo
- Department of Pathology, Affiliated Hospital of Nantong University, Nantong, China
- Medical School of Nantong University, Nantong, China
| | - Weibiao Zeng
- Department of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Hao Ding
- Department of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Shu Pan
- Department of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
- Suzhou Gene Pharma Co., Ltd, Suzhou, China
| |
Collapse
|
4
|
Chen L, Chau WY, Yuen HT, Liu XH, Qi RZ, Lung ML, Lung HL. THY1 (CD90) Maintains the Adherens Junctions in Nasopharyngeal Carcinoma via Inhibition of SRC Activation. Cancers (Basel) 2023; 15:cancers15072189. [PMID: 37046850 PMCID: PMC10093038 DOI: 10.3390/cancers15072189] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2023] [Revised: 03/31/2023] [Accepted: 04/04/2023] [Indexed: 04/14/2023] Open
Abstract
We had previously shown that THY1 (CD90) is a tumor suppressor in nasopharyngeal carcinoma (NPC) and that its down-regulation and loss of expression are associated with tumor metastasis, yet the mechanism leading to such effects remains unknown. In this study we show that tumor invasion could be suppressed by THY1 via adherens junction formation in a few NPC cell lines, and knockdown of THY1 would disrupt this cell-cell adhesion phenotype. Mechanistically, the activity of the SRC family kinase (SFK) member, SRC, and canonical Wnt signaling were dramatically reduced when THY1 was constitutively expressed. Previous studies by others have found that high levels of SRC activity in NPCs are associated with EMT and a poor prognosis. We hypothesized that THY1 can suppress tumor invasion in NPC via inhibition of SRC. By gene silencing of SRC, we found that the in vitro NPC cell invasion was significantly reduced and adherens junctions were restored. Through proteomic analysis, we identified that platelet-derived growth factor receptor β (PDGF-Rβ) and protein tyrosine phosphatase nonreceptor type 22 (PTPN22) are novel and potential binding partners of THY1, which were subsequently verified by co-immunoprecipitation (co-IP) analysis. The ligand of PDGF-Rβ (PDGF-BB) could highly induce SRC activation and NPC cell invasion, which could be almost completely suppressed by THY1 expression. On the other hand, the PTPN22 siRNA could enhance both the SRC activities and the cell invasion and could also disrupt the adherens junctions in the THY1-expressing NPC cells; the original THY1-induced phenotypes were reverted when the PTPN22 expression was reduced. Together, our results identified that PTPN22 is essential for THY1 to suppress cell invasion and SRC activity, maintain tight adherens junctions, and prevent NPC metastasis. These results suggested that PDGF-Rβ and SRC can be used as drug targets for suppressing NPC metastasis. Indeed, our in vivo assay using the SRC inhibitor KX2-391, clearly showed that inhibition of SRC signaling can prevent the metastasis of NPC, indicating that targeting SRC can be a promising approach to control the NPC progression.
Collapse
Affiliation(s)
- Luo Chen
- Department of Chemistry, Faculty of Science, Hong Kong Baptist University, Hong Kong 999077, China
| | - Wai Yin Chau
- Department of Chemistry, Faculty of Science, Hong Kong Baptist University, Hong Kong 999077, China
| | - Hei Tung Yuen
- Department of Chemistry, Faculty of Science, Hong Kong Baptist University, Hong Kong 999077, China
| | - Xiao Han Liu
- Department of Chemistry, Faculty of Science, Hong Kong Baptist University, Hong Kong 999077, China
| | - Robert Zhong Qi
- Bioscience and Biomedical Engineering Thrust, The Hong Kong University of Science and Technology (Guangzhou), Guangzhou 511400, China
- Division of Life Science, The Hong Kong University of Science and Technology, Hong Kong 999077, China
| | - Maria Li Lung
- Department of Clinical Oncology, University of Hong Kong, Hong Kong 999077, China
| | - Hong Lok Lung
- Department of Chemistry, Faculty of Science, Hong Kong Baptist University, Hong Kong 999077, China
| |
Collapse
|
5
|
Arcuri D, Ramchatesingh B, Lagacé F, Iannattone L, Netchiporouk E, Lefrançois P, Litvinov IV. Pharmacological Agents Used in the Prevention and Treatment of Actinic Keratosis: A Review. Int J Mol Sci 2023; 24:ijms24054989. [PMID: 36902419 PMCID: PMC10003023 DOI: 10.3390/ijms24054989] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 02/17/2023] [Accepted: 02/21/2023] [Indexed: 03/08/2023] Open
Abstract
Actinic keratosis (AK) is among the most commonly diagnosed skin diseases with potentially life-threatening repercussions if left untreated. Usage of pharmacologic agents represents one of many therapeutic strategies that can be used to help manage these lesions. Ongoing research into these compounds continues to change our clinical understanding as to which agents most benefit particular patient populations. Indeed, factors such as past personal medical history, lesion location and tolerability of therapy only represent a few considerations that clinicians must account for when prescribing appropriate treatment. This review focuses on specific drugs used in either the prevention or treatment of AKs. Nicotinamide, acitretin and topical 5-fluorouracil (5-FU) continue to be used with fidelity in the chemoprevention of actinic keratosis, although some uncertainty persists in regard to which agents should be used in immunocompetent vs. immunodeficient/immunosuppressed patients. Topical 5-FU, including combination formulations with either calcipotriol or salicylic acid, as well as imiquimod, diclofenac and photodynamic light therapy are all accepted treatment strategies employed to target and eliminate AKs. Five percent of 5-FU is regarded as the most effective therapy in the condition, although the literature has conflictingly shown that lower concentrations of the drug might also be as effective. Topical diclofenac (3%) appears to be less efficacious than 5% 5-FU, 3.75-5% imiquimod and photodynamic light therapy despite its favorable side effect profile. Finally, traditional photodynamic light therapy, while painful, appears to be of higher efficacy in comparison to its more tolerable counterpart, daylight phototherapy.
Collapse
Affiliation(s)
- Domenico Arcuri
- Department of Medicine, McGill University, Montreal, QC H4A 3J1, Canada
| | | | - François Lagacé
- Department of Medicine, McGill University, Montreal, QC H4A 3J1, Canada
| | - Lisa Iannattone
- Department of Medicine, McGill University, Montreal, QC H4A 3J1, Canada
| | | | | | - Ivan V. Litvinov
- Department of Medicine, McGill University, Montreal, QC H4A 3J1, Canada
- Division of Dermatology, McGill University Health Center, Montreal, QC H4A 3J1, Canada
- Correspondence:
| |
Collapse
|
6
|
Gomari MM, Abkhiz S, Pour TG, Lotfi E, Rostami N, Monfared FN, Ghobari B, Mosavi M, Alipour B, Dokholyan NV. Peptidomimetics in cancer targeting. Mol Med 2022; 28:146. [PMID: 36476230 PMCID: PMC9730693 DOI: 10.1186/s10020-022-00577-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Accepted: 11/16/2022] [Indexed: 12/12/2022] Open
Abstract
The low efficiency of treatment strategies is one of the main obstacles to developing cancer inhibitors. Up to now, various classes of therapeutics have been developed to inhibit cancer progression. Peptides due to their small size and easy production compared to proteins are highly regarded in designing cancer vaccines and oncogenic pathway inhibitors. Although peptides seem to be a suitable therapeutic option, their short lifespan, instability, and low binding affinity for their target have not been widely applicable against malignant tumors. Given the peptides' disadvantages, a new class of agents called peptidomimetic has been introduced. With advances in physical chemistry and biochemistry, as well as increased knowledge about biomolecule structures, it is now possible to chemically modify peptides to develop efficient peptidomimetics. In recent years, numerous studies have been performed to the evaluation of the effectiveness of peptidomimetics in inhibiting metastasis, angiogenesis, and cancerous cell growth. Here, we offer a comprehensive review of designed peptidomimetics to diagnose and treat cancer.
Collapse
Affiliation(s)
- Mohammad Mahmoudi Gomari
- grid.411746.10000 0004 4911 7066Department of Medical Biotechnology, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Shadi Abkhiz
- grid.411746.10000 0004 4911 7066Department of Medical Biotechnology, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Taha Ghantab Pour
- grid.411746.10000 0004 4911 7066Department of Anatomy, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Ehsan Lotfi
- grid.411746.10000 0004 4911 7066Department of Medical Biotechnology, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Neda Rostami
- grid.411425.70000 0004 0417 7516Department of Chemical Engineering, Faculty of Engineering, Arak University, Arak, Iran
| | - Fatemeh Nafe Monfared
- grid.411705.60000 0001 0166 0922Department of Virology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Babak Ghobari
- grid.412831.d0000 0001 1172 3536Department of Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran
| | - Mona Mosavi
- grid.411746.10000 0004 4911 7066Department of Medical Biotechnology, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Behruz Alipour
- grid.411705.60000 0001 0166 0922Medical Biotechnology Department, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Nikolay V. Dokholyan
- grid.240473.60000 0004 0543 9901Department of Pharmacology, Penn State College of Medicine, Hershey, PA USA ,grid.240473.60000 0004 0543 9901Department of Biochemistry & Molecular Biology, Penn State College of Medicine, Hershey, PA USA
| |
Collapse
|
7
|
Schlesinger T, Stockfleth E, Grada A, Berman B. Tirbanibulin for Actinic Keratosis: Insights into the Mechanism of Action. Clin Cosmet Investig Dermatol 2022; 15:2495-2506. [PMID: 36415541 PMCID: PMC9675993 DOI: 10.2147/ccid.s374122] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Accepted: 10/19/2022] [Indexed: 08/13/2023]
Abstract
Actinic keratosis (AK) is a common pre-neoplastic skin lesion constituted by uncontrolled proliferation of atypical keratinocytes that may evolve to squamous cell carcinoma. With global prevalence increasing, AK is expected to be the most common carcinoma of the skin. Tirbanibulin is a reversible tubulin polymerization inhibitor with potent anti-proliferative and anti-tumoral effects. In-vivo and in-vitro studies have shown that tirbanibulin significantly inhibits cell proliferation, tumor growth and downregulates Src signaling with no overt toxicity. Early phase and Phase III trials have shown high lesion clearance, compliance, and few side effects of once daily tirbanibulin treatment. This review discusses tirbanibulin anti-cancer activity, focusing on tubulin polymerization and Src signaling inhibitory effects, highlighting relevant literature and novel preclinical results from the ATNXUS-KX01-001 study. Furthermore, we address the relevant findings obtained in recent clinical trials to evaluate the safety, efficacy, pharmacokinetics, clearance efficacy, and side effects of the 1% tirbanibulin ointment applied once daily. In summary, we highlight preclinical and clinical evidence on the use of tirbanibulin as an effective and safe treatment option for AK.
Collapse
Affiliation(s)
| | - Eggert Stockfleth
- Department of Dermatology, Venereology and Allergology, Ruhr-University Bochum, Bochum, Germany
| | - Ayman Grada
- Department of Dermatology, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Brian Berman
- Dr. Phillip Frost Department of Dermatology and Cutaneous Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
| |
Collapse
|
8
|
Topical Tirbanibulin, a Dual Src Kinase and Tubulin Polymerization Inhibitor, for the Treatment of Plaque-Type Psoriasis: Phase I Results. Pharmaceutics 2022; 14:pharmaceutics14102159. [PMID: 36297594 PMCID: PMC9608911 DOI: 10.3390/pharmaceutics14102159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 10/04/2022] [Accepted: 10/05/2022] [Indexed: 11/17/2022] Open
Abstract
Plaque-type psoriasis is a common skin disorder. Tirbanibulin (KX01) is a new Src kinase inhibitor with potent antiproliferative activity against keratinocytes and has been approved for treatment of actinic keratosis. This Phase I study investigates the safety and activity of KX01 ointment in patients with plaque-type psoriasis. We recruited 28 patients from two medical centers in Taiwan. This study was performed in four stages. Double-blind treatments were randomized in stages I (KX01 0.01% + placebo, two rounds of two-week treatment) and II (KX01 0.1% + placebo, four weeks) and open-labelled in stages III (KX01 1%, five days) and IV (KX01 1%, five days weekly for four weeks). The safety, tolerability, KX01 concentration, target area score, physician global assessment, and disease relapse were determined. Most treatment-emergent adverse events were mild-to-moderate application site reactions. Three (50.0%) subjects from the stage IV group showed ≥50% reduction in the target area score (TAS50), while two subjects (33.3%) showed a clinically meaningful improvement in the physician global assessment score. KX01 0.01%, 0.1%, and 1% were safe and well-tolerated. KX01 1% at four weeks showed a promising activity for the treatment of plaque-type psoriasis.
Collapse
|
9
|
Peng J, Zeng Y, Hu X, Huang S, Gao X, Tian D, Tian S, Qiu L, Liu J, Dong R, Zhan W, Qin C, Guang B, Yang T. KC-180-2 Exerts Anti-SCLC Effects via Dual Inhibition of Tubulin Polymerization and Src Signaling. ACS OMEGA 2022; 7:32164-32175. [PMID: 36120000 PMCID: PMC9476193 DOI: 10.1021/acsomega.2c03408] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Accepted: 08/17/2022] [Indexed: 06/15/2023]
Abstract
In this study, a series of N-benzyl-2-(5-phenylpyridin-2-yl) acetamide-based derivatives were successfully designed and synthesized as anti-cancer agents. KC-180-2 was screened as a potentially leading compound with dual mechanisms of action: Src signaling and tubulin polymerization inhibition. It efficiently suppressed the proliferation of five cancer cell lines (MDA-MB-231, H446, SKOV-3, HepG2, and HT29), with IC50 values ranging from 5 to 188 nM, especially small-cell lung cancer (SCLC) cells (IC50, 5 nM). Correspondingly, it exerted a significant therapeutic effect on the H446 small-cell lung cancer xenograft model, significantly reducing the volume of tumors without obvious toxicity. Mechanistically, this compound significantly inhibited the polymerization of purified tubulin in vitro, inducing G2/M cell cycle arrest and binding to the kinase catalytic domain of the Src protein, which reduced the phosphorylation of Src. Thus, KC-180-2 is a potential lead compound for the further development of a new anti-tumor drug against SCLC.
Collapse
Affiliation(s)
- Jian Peng
- School
of Pharmacy, Chengdu Medical College, No. 783, Xindu Avenue, Xindu District, Chengdu, Sichuan Province 610500, China
| | - Yisheng Zeng
- School
of Pharmacy, Chengdu Medical College, No. 783, Xindu Avenue, Xindu District, Chengdu, Sichuan Province 610500, China
| | - Xiaojun Hu
- School
of Pharmacy, Chengdu Medical College, No. 783, Xindu Avenue, Xindu District, Chengdu, Sichuan Province 610500, China
| | - Sheng Huang
- Chengdu
Biobel Biotechnology Co., Ltd., No. 88, Keyuan South Road, New and High-Tech Zone, Chengdu, Sichuan Province 610094, China
| | - Xiaofang Gao
- School
of Pharmacy, Chengdu Medical College, No. 783, Xindu Avenue, Xindu District, Chengdu, Sichuan Province 610500, China
| | - Dong Tian
- School
of Pharmacy, Chengdu Medical College, No. 783, Xindu Avenue, Xindu District, Chengdu, Sichuan Province 610500, China
| | - Shuting Tian
- School
of Pharmacy, Chengdu Medical College, No. 783, Xindu Avenue, Xindu District, Chengdu, Sichuan Province 610500, China
| | - Lan Qiu
- School
of Pharmacy, Chengdu Medical College, No. 783, Xindu Avenue, Xindu District, Chengdu, Sichuan Province 610500, China
| | - Jin Liu
- School
of Pharmacy, Chengdu Medical College, No. 783, Xindu Avenue, Xindu District, Chengdu, Sichuan Province 610500, China
| | - Renhan Dong
- Chengdu
Biobel Biotechnology Co., Ltd., No. 88, Keyuan South Road, New and High-Tech Zone, Chengdu, Sichuan Province 610094, China
| | - Wei Zhan
- Chengdu
Biobel Biotechnology Co., Ltd., No. 88, Keyuan South Road, New and High-Tech Zone, Chengdu, Sichuan Province 610094, China
| | - Chuanjun Qin
- Chengdu
Biobel Biotechnology Co., Ltd., No. 88, Keyuan South Road, New and High-Tech Zone, Chengdu, Sichuan Province 610094, China
| | - Bing Guang
- Chengdu
Biobel Biotechnology Co., Ltd., No. 88, Keyuan South Road, New and High-Tech Zone, Chengdu, Sichuan Province 610094, China
| | - Tai Yang
- School
of Pharmacy, Chengdu Medical College, No. 783, Xindu Avenue, Xindu District, Chengdu, Sichuan Province 610500, China
| |
Collapse
|
10
|
[Translated article] Tirbanibulin: review of its novel mechanism of action and how it fits into the treatment of actinic keratosis. ACTAS DERMO-SIFILIOGRAFICAS 2022. [DOI: 10.1016/j.ad.2021.07.016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
|
11
|
Gilaberte Y, Fernández-Figueras M. Tirbanibulina: revisión de su mecanismo de acción novedoso y de cómo encaja en el tratamiento de la queratosis actínica. ACTAS DERMO-SIFILIOGRAFICAS 2022; 113:58-66. [DOI: 10.1016/j.ad.2021.07.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Accepted: 07/18/2021] [Indexed: 11/24/2022] Open
|
12
|
Tirbanibulin: review of its novel mechanism of action and how it fits into the treatment of actinic keratosis. ACTAS DERMO-SIFILIOGRAFICAS 2021. [DOI: 10.1016/j.adengl.2021.11.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
|
13
|
Yang L, Zhang W, Qiu Q, Su Z, Tang M, Bai P, Si W, Zhu Z, Liu Y, Yang J, Kuang S, Liu J, Yan W, Shi M, Ye H, Yang Z, Chen L. Discovery of a Series of Hydroxamic Acid-Based Microtubule Destabilizing Agents with Potent Antitumor Activity. J Med Chem 2021; 64:15379-15401. [PMID: 34648295 DOI: 10.1021/acs.jmedchem.1c01451] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Hydroxamic acid group is one of the characteristic pharmacophores of histone deacetylase (HDAC) inhibitors. But here, we discovered a series of hydroxamic acid-based microtubule destabilizing agents (MDAs), which were derived from shortening the length of the linker in HDAC6 inhibitor SKLB-23bb. Interestingly, the low nanomolar antiproliferative activity of these MDAs depended on the presence of hydroxamic acid groups, but their inhibitory effects on HDAC were lost. Among them, 12b showed favorable metabolism stability, high bioavailability, and potent antitumor activity in multidrug-resistant cell lines and A2780/T xenograft model. More importantly, in the patient-derived xenograft models of triple-negative breast cancer and osimertinib-resistant non-small-cell lung cancer, both 20 mg/kg oral and 10 mg/kg intravenous administration of 12b could induce more than 70% tumor inhibition without obvious toxicity. Overall, we discovered that 12b, as a novel MDA based on hydroxamic acid, could serve as a potential MDA for further investigation.
Collapse
Affiliation(s)
- Linyu Yang
- State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu 610041, Sichuan, China
| | - Wanhua Zhang
- State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu 610041, Sichuan, China.,Department of Hematology, West China Hospital of Sichuan University, Chengdu 610041, Sichuan, China
| | - Qiang Qiu
- State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu 610041, Sichuan, China
| | - Zhengying Su
- State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu 610041, Sichuan, China
| | - Minghai Tang
- State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu 610041, Sichuan, China
| | - Peng Bai
- State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu 610041, Sichuan, China
| | - Wenting Si
- State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu 610041, Sichuan, China
| | - Zejiang Zhu
- State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu 610041, Sichuan, China
| | - Yan Liu
- State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu 610041, Sichuan, China
| | - Jianhong Yang
- State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu 610041, Sichuan, China
| | - Shuang Kuang
- State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu 610041, Sichuan, China
| | - Jiang Liu
- State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu 610041, Sichuan, China
| | - Wei Yan
- State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu 610041, Sichuan, China
| | - Mingsong Shi
- State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu 610041, Sichuan, China
| | - Haoyu Ye
- State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu 610041, Sichuan, China
| | - Zhuang Yang
- State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu 610041, Sichuan, China
| | - Lijuan Chen
- State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu 610041, Sichuan, China
| |
Collapse
|
14
|
Shuai W, Wang G, Zhang Y, Bu F, Zhang S, Miller DD, Li W, Ouyang L, Wang Y. Recent Progress on Tubulin Inhibitors with Dual Targeting Capabilities for Cancer Therapy. J Med Chem 2021; 64:7963-7990. [PMID: 34101463 DOI: 10.1021/acs.jmedchem.1c00100] [Citation(s) in RCA: 74] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Microtubules play a crucial role in multiple cellular functions including mitosis, cell signaling, and organelle trafficking, which makes the microtubule an important target for cancer therapy. Despite the great successes of microtubule-targeting agents in the clinic, the development of drug resistance and dose-limiting toxicity restrict their clinical efficacy. In recent years, multitarget therapy has been considered an effective strategy to achieve higher therapeutic efficacy, in particular dual-target drugs. In terms of the synergetic effect of tubulin and other antitumor agents such as receptor tyrosine kinases inhibitors, histone deacetylases inhibitors, DNA-damaging agents, and topoisomerase inhibitors in combination therapy, designing dual-target tubulin inhibitors is regarded as a promising approach to overcome these limitations and improve therapeutic efficacy. In this Perspective, we discussed rational target combinations, design strategies, structure-activity relationships, and future directions of dual-target tubulin inhibitors.
Collapse
Affiliation(s)
- Wen Shuai
- State Key Laboratory of Biotherapy and Cancer Center, Department of Respiratory and Critical Care Medicine, Innovation Center of Nursing Research, National Clinical Research Center for Geriatrics, West China Hospital, and Collaborative Innovation Center of Biotherapy, Sichuan University, Chengdu 610041, Sichuan, China
| | - Guan Wang
- State Key Laboratory of Biotherapy and Cancer Center, Department of Respiratory and Critical Care Medicine, Innovation Center of Nursing Research, National Clinical Research Center for Geriatrics, West China Hospital, and Collaborative Innovation Center of Biotherapy, Sichuan University, Chengdu 610041, Sichuan, China
| | - Yiwen Zhang
- State Key Laboratory of Biotherapy and Cancer Center, Department of Respiratory and Critical Care Medicine, Innovation Center of Nursing Research, National Clinical Research Center for Geriatrics, West China Hospital, and Collaborative Innovation Center of Biotherapy, Sichuan University, Chengdu 610041, Sichuan, China
| | - Faqian Bu
- State Key Laboratory of Biotherapy and Cancer Center, Department of Respiratory and Critical Care Medicine, Innovation Center of Nursing Research, National Clinical Research Center for Geriatrics, West China Hospital, and Collaborative Innovation Center of Biotherapy, Sichuan University, Chengdu 610041, Sichuan, China
| | - Sicheng Zhang
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee 38163, United States
| | - Duane D Miller
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee 38163, United States
| | - Wei Li
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee 38163, United States
| | - Liang Ouyang
- State Key Laboratory of Biotherapy and Cancer Center, Department of Respiratory and Critical Care Medicine, Innovation Center of Nursing Research, National Clinical Research Center for Geriatrics, West China Hospital, and Collaborative Innovation Center of Biotherapy, Sichuan University, Chengdu 610041, Sichuan, China
| | - Yuxi Wang
- State Key Laboratory of Biotherapy and Cancer Center, Department of Respiratory and Critical Care Medicine, Innovation Center of Nursing Research, National Clinical Research Center for Geriatrics, West China Hospital, and Collaborative Innovation Center of Biotherapy, Sichuan University, Chengdu 610041, Sichuan, China.,Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China.,Precision Medicine Key Laboratory of Sichuan Province & Precision Medicine Center, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China
| |
Collapse
|
15
|
Novel bone-targeted parathyroid hormone-related peptide antagonists inhibit breast cancer bone metastases. Anticancer Drugs 2021; 32:365-375. [PMID: 33595947 DOI: 10.1097/cad.0000000000001051] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Patients with advanced breast cancer often develop bone metastases. Treatment is limited to palliative care. Parathyroid hormone (PTH)/parathyroid hormone-related peptide (PTHrP) antagonists for bone metastases failed clinically due to short half-life and inadequate concentration in bone. We synthesized two novel PTHrP antagonists fused to an inert bacterial collagen binding domain (CBD) that directs drugs to bone. PTH(7-33)-CBD is an N-terminal truncated PTHrP antagonist. [W2]PTH(1-33)-CBD is an PTHrP inverse-agonist. The aim of this study was to assess PTH(7-33)-CBD to reduce breast cancer bone metastases and prevent osteolytic destruction in mice and to assess both drugs for apoptosis of breast cancer cells in vitro and inhibition of PTH receptor (PTHR1). PTH(7-33)-CBD (1000 µg/kg, subcutaneous) or vehicle was administered 24 h prior to MDA-MB-231 breast cancer cell inoculation into the tibia marrow. Weekly tumor burden and bone density were measured. Pharmacokinetic analysis of PTH(7-33)-CBD in rat serum was evaluated. Drug effect on cAMP accumulation in SaOS-2 osteosarcoma cells and apoptosis of MDA-MB-231 cells was assessed. PTH(7-33)-CBD reduced MDA-MB-231 tumor burden and osteolytic destruction in mice 4-5 weeks post-treatment. PTH(7-33)-CBD (1000 μg/kg i.v. and subcutaneous) in rats was rapidly absorbed with peak concentration 5-min and terminal half-life 3-h. Bioavailability by the subcutaneous route was 43% relative to the i.v. route. PTH(7-33)-CBD was detected only on rat periosteal bone surfaces that stained positive for collagen-1. PTH(7-33)-CBD and [W2]PTH(1-33)-CBD (10-8M) blocked basal and PTH agonist-induced cAMP accumulation in SaOS-2 osteosarcoma cells. Both drugs induced PTHR1-dependent apoptosis of MDA-MB-231 cells in vitro. Novel bone-targeted PTHrP antagonists represent a new paradigm for treatment of breast cancer bone metastases.
Collapse
|
16
|
Stanković T, Dinić J, Podolski-Renić A, Musso L, Burić SS, Dallavalle S, Pešić M. Dual Inhibitors as a New Challenge for Cancer Multidrug Resistance Treatment. Curr Med Chem 2019; 26:6074-6106. [PMID: 29874992 DOI: 10.2174/0929867325666180607094856] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2018] [Revised: 05/28/2018] [Accepted: 05/28/2018] [Indexed: 12/23/2022]
Abstract
BACKGROUND Dual-targeting in cancer treatment by a single drug is an unconventional approach in relation to drug combinations. The rationale for the development of dualtargeting agents is to overcome incomplete efficacy and drug resistance frequently present when applying individual targeting agents. Consequently, -a more favorable outcome of cancer treatment is expected with dual-targeting strategies. METHODS We reviewed the literature, concentrating on the association between clinically relevant and/or novel dual inhibitors with the potential to modulate multidrug resistant phenotype of cancer cells, particularly the activity of P-glycoprotein. A balanced analysis of content was performed to emphasize the most important findings and optimize the structure of this review. RESULTS Two-hundred and forty-five papers were included in the review. The introductory part was interpreted by 9 papers. Tyrosine kinase inhibitors' role in the inhibition of Pglycoprotein and chemosensitization was illustrated by 87 papers. The contribution of naturalbased compounds in overcoming multidrug resistance was reviewed using 92 papers, while specific dual inhibitors acting against microtubule assembling and/or topoisomerases were described with 55 papers. Eleven papers gave an insight into a novel and less explored approach with hybrid drugs. Their influence on P-glycoprotein and multidrug resistance was also evaluated. CONCLUSION These findings bring into focus rational anticancer strategies with dual-targeting agents. Most evaluated synthetic and natural drugs showed a great potential in chemosensitization. Further steps in this direction are needed for the optimization of anticancer treatment.
Collapse
Affiliation(s)
- Tijana Stanković
- Department of Neurobiology, Institute for Biological Research "Sinisa Stankovic", University of Belgrade, Belgrade, Serbia
| | - Jelena Dinić
- Department of Neurobiology, Institute for Biological Research "Sinisa Stankovic", University of Belgrade, Belgrade, Serbia
| | - Ana Podolski-Renić
- Department of Neurobiology, Institute for Biological Research "Sinisa Stankovic", University of Belgrade, Belgrade, Serbia
| | - Loana Musso
- DeFENS, Department of Food, Environmental and Nutritional Sciences, Universita degli Studi di Milano, Milano, Italy
| | - Sonja Stojković Burić
- Department of Neurobiology, Institute for Biological Research "Sinisa Stankovic", University of Belgrade, Belgrade, Serbia
| | - Sabrina Dallavalle
- DeFENS, Department of Food, Environmental and Nutritional Sciences, Universita degli Studi di Milano, Milano, Italy
| | - Milica Pešić
- Department of Neurobiology, Institute for Biological Research "Sinisa Stankovic", University of Belgrade, Belgrade, Serbia
| |
Collapse
|
17
|
Arnst KE, Banerjee S, Chen H, Deng S, Hwang DJ, Li W, Miller DD. Current advances of tubulin inhibitors as dual acting small molecules for cancer therapy. Med Res Rev 2019; 39:1398-1426. [PMID: 30746734 DOI: 10.1002/med.21568] [Citation(s) in RCA: 94] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Revised: 01/16/2019] [Accepted: 01/19/2019] [Indexed: 12/25/2022]
Abstract
Microtubule (MT)-targeting agents are highly successful drugs as chemotherapeutic agents, and this is attributed to their ability to target MT dynamics and interfere with critical cellular functions, including, mitosis, cell signaling, intracellular trafficking, and angiogenesis. Because MT dynamics vary in the different stages of the cell cycle, these drugs tend to be the most effective against mitotic cells. While this class of drug has proven to be effective against many cancer types, significant hurdles still exist and include overcoming aspects such as dose limited toxicities and the development of resistance. Newer generations of developed drugs attack these problems and alternative approaches such as the development of dual tubulin and kinase inhibitors are being investigated. This approach offers the potential to show increased efficacy and lower toxicities. This review covers different categories of MT-targeting agents, recent advances in dual inhibitors, and current challenges for this drug target.
Collapse
Affiliation(s)
- Kinsie E Arnst
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee
| | - Souvik Banerjee
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee
| | - Hao Chen
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee
| | - Shanshan Deng
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee
| | - Dong-Jin Hwang
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee
| | - Wei Li
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee
| | - Duane D Miller
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee
| |
Collapse
|
18
|
KX2-361: a novel orally bioavailable small molecule dual Src/tubulin inhibitor that provides long term survival in a murine model of glioblastoma. J Neurooncol 2018; 140:519-527. [DOI: 10.1007/s11060-018-2992-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Accepted: 08/27/2018] [Indexed: 10/28/2022]
|
19
|
Smolinski MP, Bu Y, Clements J, Gelman IH, Hegab T, Cutler DL, Fang JWS, Fetterly G, Kwan R, Barnett A, Lau JYN, Hangauer DG. Discovery of Novel Dual Mechanism of Action Src Signaling and Tubulin Polymerization Inhibitors (KX2-391 and KX2-361). J Med Chem 2018; 61:4704-4719. [PMID: 29617135 DOI: 10.1021/acs.jmedchem.8b00164] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
The discovery of potent, peptide site directed, tyrosine kinase inhibitors has remained an elusive goal. Herein we describe the discovery of two such clinical candidates that inhibit the tyrosine kinase Src. Compound 1 is a phase 3 clinical trial candidate that is likely to provide a first in class topical treatment for actinic keratosis (AK) with good efficacy and dramatically less toxicity compared to existing standard therapy. Compound 2 is a phase 1 clinical trial candidate that is likely to provide a first in class treatment of malignant glioblastoma and induces 30% long-term complete tumor remission in animal models. The discovery strategy for these compounds iteratively utilized molecular modeling, along with the synthesis and testing of increasingly elaborated proof of concept compounds, until the final clinical candidates were arrived at. This was followed with mechanism of action (MOA) studies that revealed tubulin polymerization inhibition as the second MOA.
Collapse
Affiliation(s)
- Michael P Smolinski
- Athenex Inc. , Conventus Building, 1001 Main Street, Suite 600 , Buffalo , New York 14203 , United States
| | - Yahao Bu
- Athenex Inc. , Conventus Building, 1001 Main Street, Suite 600 , Buffalo , New York 14203 , United States
| | - James Clements
- Athenex Inc. , Conventus Building, 1001 Main Street, Suite 600 , Buffalo , New York 14203 , United States
| | - Irwin H Gelman
- Department of Cancer Genetics & Genomics , Roswell Park Comprehensive Cancer Center , Elm and Carlton Streets , Buffalo , New York 14263 , United States
| | - Taher Hegab
- Athenex Inc. , Conventus Building, 1001 Main Street, Suite 600 , Buffalo , New York 14203 , United States
| | - David L Cutler
- Athenex Inc. , Conventus Building, 1001 Main Street, Suite 600 , Buffalo , New York 14203 , United States
| | - Jane W S Fang
- Athenex Inc. , Conventus Building, 1001 Main Street, Suite 600 , Buffalo , New York 14203 , United States
| | - Gerald Fetterly
- Athenex Inc. , Conventus Building, 1001 Main Street, Suite 600 , Buffalo , New York 14203 , United States
| | - Rudolf Kwan
- Athenex Inc. , Conventus Building, 1001 Main Street, Suite 600 , Buffalo , New York 14203 , United States
| | - Allen Barnett
- Athenex Inc. , Conventus Building, 1001 Main Street, Suite 600 , Buffalo , New York 14203 , United States
| | - Johnson Y N Lau
- Athenex Inc. , Conventus Building, 1001 Main Street, Suite 600 , Buffalo , New York 14203 , United States
| | - David G Hangauer
- Athenex Inc. , Conventus Building, 1001 Main Street, Suite 600 , Buffalo , New York 14203 , United States
| |
Collapse
|
20
|
Resistance to dasatinib is associated with the activation of Akt in oral squamous cell carcinoma. TRANSLATIONAL RESEARCH IN ORAL ONCOLOGY 2017. [DOI: 10.1177/2057178x17702920] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
|
21
|
Anbalagan M, Sheng M, Fleischer B, Zhang Y, Gao Y, Hoang V, Matossian M, Burks HE, Burow ME, Collins-Burow BM, Hangauer D, Rowan BG. Dual Src Kinase/Pretubulin Inhibitor KX-01, Sensitizes ERα-negative Breast Cancers to Tamoxifen through ERα Reexpression. Mol Cancer Res 2017; 15:1491-1502. [PMID: 28751463 DOI: 10.1158/1541-7786.mcr-16-0297-t] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2016] [Revised: 02/22/2017] [Accepted: 07/19/2017] [Indexed: 12/20/2022]
Abstract
Unlike breast cancer that is positive for estrogen receptor-α (ERα), there are no targeted therapies for triple-negative breast cancer (TNBC). ERα is silenced in TNBC through epigenetic changes including DNA methylation and histone acetylation. Restoring ERα expression in TNBC may sensitize patients to endocrine therapy. Expression of c-Src and ERα are inversely correlated in breast cancer suggesting that c-Src inhibition may lead to reexpression of ERα in TNBC. KX-01 is a peptide substrate-targeted Src/pretubulin inhibitor in clinical trials for solid tumors. KX-01 (1 mg/kg body weight-twice daily) inhibited growth of tamoxifen-resistant MDA-MB-231 and MDA-MB-157 TNBC xenografts in nude mice that was correlated with Src kinase inhibition. KX-01 also increased ERα mRNA and protein, as well as increased the ERα targets progesterone receptor (PR), pS2 (TFF1), cyclin D1 (CCND1), and c-myc (MYC) in MDA-MB-231 and MDA-MB-468, but not MDA-MB-157 xenografts. MDA-MB-231 and MDA-MB-468 tumors exhibited reduction in mesenchymal markers (vimentin, β-catenin) and increase in epithelial marker (E-cadherin) suggesting mesenchymal-to-epithelial transition (MET). KX-01 sensitized MDA-MB-231 and MDA-MB-468 tumors to tamoxifen growth inhibition and tamoxifen repression of the ERα targets pS2, cyclin D1, and c-myc. Chromatin immunoprecipitation (ChIP) of the ERα promoter in KX-01-treated tumors demonstrated enrichment of active transcription marks (acetyl-H3, acetyl-H3Lys9), dissociation of HDAC1, and recruitment of RNA polymerase II. Methylation-specific PCR and bisulfite sequencing demonstrated no alteration in ERα promoter methylation by KX-01. These data demonstrate that in addition to Src kinase inhibition, peptidomimetic KX-01 restores ERα expression in TNBC through changes in histone acetylation that sensitize tumors to tamoxifen.Implications: Src kinase/pretubulin inhibitor KX-01 restores functional ERα expression in ERα- breast tumors, a novel treatment strategy to treat triple-negative breast cancer. Mol Cancer Res; 15(11); 1491-502. ©2017 AACR.
Collapse
Affiliation(s)
- Muralidharan Anbalagan
- Department of Structural and Cellular Biology, Tulane University School of Medicine, New Orleans, Louisiana
| | - Mei Sheng
- Department of Structural and Cellular Biology, Tulane University School of Medicine, New Orleans, Louisiana
| | - Brian Fleischer
- Department of Structural and Cellular Biology, Tulane University School of Medicine, New Orleans, Louisiana
| | - Yifang Zhang
- Department of Structural and Cellular Biology, Tulane University School of Medicine, New Orleans, Louisiana.,Department of Obstetrics and Gynecology, Affiliated Hospital of Taishan Medical University, Taishan, Shandong, China
| | - Yuanjun Gao
- Department of Structural and Cellular Biology, Tulane University School of Medicine, New Orleans, Louisiana.,Department of Gastroenterology, Taihe Hospital, Hubei University of Medicine, Hubei, China
| | - Van Hoang
- Department of Medicine, Section of Hematology and Medical Oncology, Tulane University School of Medicine, New Orleans, Louisiana
| | - Margarite Matossian
- Department of Medicine, Section of Hematology and Medical Oncology, Tulane University School of Medicine, New Orleans, Louisiana
| | - Hope E Burks
- Department of Medicine, Section of Hematology and Medical Oncology, Tulane University School of Medicine, New Orleans, Louisiana
| | - Matthew E Burow
- Department of Medicine, Section of Hematology and Medical Oncology, Tulane University School of Medicine, New Orleans, Louisiana
| | - Bridgette M Collins-Burow
- Department of Medicine, Section of Hematology and Medical Oncology, Tulane University School of Medicine, New Orleans, Louisiana
| | - David Hangauer
- Athenex Pharmaceuticals LLC, New York State Center of Excellence in Bioinformatics and Life Sciences, Buffalo, New York
| | - Brian G Rowan
- Department of Structural and Cellular Biology, Tulane University School of Medicine, New Orleans, Louisiana.
| |
Collapse
|
22
|
Kim S, Min A, Lee KH, Yang Y, Kim TY, Lim JM, Park SJ, Nam HJ, Kim JE, Song SH, Han SW, Oh DY, Kim JH, Kim TY, Hangauer D, Lau JYN, Im K, Lee DS, Bang YJ, Im SA. Antitumor Effect of KX-01 through Inhibiting Src Family Kinases and Mitosis. Cancer Res Treat 2016; 49:643-655. [PMID: 27737538 PMCID: PMC5512373 DOI: 10.4143/crt.2016.168] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2016] [Accepted: 09/17/2016] [Indexed: 12/26/2022] Open
Abstract
PURPOSE KX-01 is a novel dual inhibitor of Src and tubulin. Unlike previous Src inhibitors that failed to show clinical benefit during treatment of breast cancer, KX-01 can potentially overcome the therapeutic limitations of current Src inhibitors through inhibition of both Src and tubulin. The present study further evaluates the activity and mechanism of KX-01 in vitro and in vivo. MATERIALS AND METHODS The antitumor effect of KX-01 in triple negative breast cancer (TNBC) cell lines was determined by MTT assay. Wound healing and immunofluorescence assays were performed to evaluate the action mechanisms of KX-01. Changes in the cell cycle and molecular changes induced by KX-01 were also evaluated. A MDA-MB-231 mouse xenograft model was used to demonstrate the in vivo effects. RESULTS KX-01 effectively inhibited the growth of breast cancer cell lines. The expression of phospho-Src and proliferative-signaling molecules were down-regulated in KX-01-sensitive TNBC cell lines. In addition, migration inhibition was observed by wound healing assay. KX-01-induced G2/M cell cycle arrest and increased the aneuploid cell population in KX-01-sensitive cell lines. Multi-nucleated cells were significantly increased after KX-01 treatment. Furthermore, KX-01 effectively delayed tumor growth in a MDA-MB-231 mouse xenograft model. CONCLUSION KX-01 effectively inhibited cell growth and migration of TNBC cells. Moreover, this study demonstrated that KX-01 showed antitumor effects through the inhibition of Src signaling and the induction of mitotic catastrophe. The antitumor effects of KX-01 were also demonstrated in vivo using a mouse xenograft model.
Collapse
Affiliation(s)
- Seongyeong Kim
- Cancer Research Institute, Seoul National University, Seoul, Korea
| | - Ahrum Min
- Cancer Research Institute, Seoul National University, Seoul, Korea.,Biomedical Research Institute, Seoul National University Hospital, Seoul, Korea
| | - Kyung-Hun Lee
- Cancer Research Institute, Seoul National University, Seoul, Korea.,Biomedical Research Institute, Seoul National University Hospital, Seoul, Korea.,Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea
| | - Yaewon Yang
- Cancer Research Institute, Seoul National University, Seoul, Korea.,Biomedical Research Institute, Seoul National University Hospital, Seoul, Korea.,Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea
| | - Tae-Yong Kim
- Cancer Research Institute, Seoul National University, Seoul, Korea.,Biomedical Research Institute, Seoul National University Hospital, Seoul, Korea.,Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea
| | - Jee Min Lim
- Cancer Research Institute, Seoul National University, Seoul, Korea
| | - So Jung Park
- Cancer Research Institute, Seoul National University, Seoul, Korea
| | - Hyun-Jin Nam
- Cancer Research Institute, Seoul National University, Seoul, Korea
| | - Jung Eun Kim
- Cancer Research Institute, Seoul National University, Seoul, Korea
| | - Sang-Hyun Song
- Cancer Research Institute, Seoul National University, Seoul, Korea
| | - Sae-Won Han
- Cancer Research Institute, Seoul National University, Seoul, Korea.,Biomedical Research Institute, Seoul National University Hospital, Seoul, Korea.,Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea
| | - Do-Youn Oh
- Cancer Research Institute, Seoul National University, Seoul, Korea.,Biomedical Research Institute, Seoul National University Hospital, Seoul, Korea.,Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea
| | - Jee Hyun Kim
- Cancer Research Institute, Seoul National University, Seoul, Korea.,Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea.,Department of Internal Medicine, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Korea
| | - Tae-You Kim
- Cancer Research Institute, Seoul National University, Seoul, Korea.,Biomedical Research Institute, Seoul National University Hospital, Seoul, Korea.,Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea
| | - David Hangauer
- Kinex Pharmaceutical Corporation, New York State Center of Excellence in Bioinformartics and Life Sciences, NY, USA
| | - Johnson Yiu-Nam Lau
- Kinex Pharmaceutical Corporation, New York State Center of Excellence in Bioinformartics and Life Sciences, NY, USA
| | - Kyongok Im
- Cancer Research Institute, Seoul National University, Seoul, Korea
| | - Dong Soon Lee
- Cancer Research Institute, Seoul National University, Seoul, Korea.,Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea
| | - Yung-Jue Bang
- Cancer Research Institute, Seoul National University, Seoul, Korea.,Biomedical Research Institute, Seoul National University Hospital, Seoul, Korea.,Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea
| | - Seock-Ah Im
- Cancer Research Institute, Seoul National University, Seoul, Korea.,Biomedical Research Institute, Seoul National University Hospital, Seoul, Korea.,Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea
| |
Collapse
|
23
|
Rowan BG, Lacayo EA, Sheng M, Anbalagan M, Gimble JM, Jones RK, Joseph WJ, Friedlander PL, Chiu ES. Human Adipose Tissue-Derived Stromal/Stem Cells Promote Migration and Early Metastasis of Head and Neck Cancer Xenografts. Aesthet Surg J 2016; 36:93-104. [PMID: 26063833 DOI: 10.1093/asj/sjv090] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/22/2015] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Fat grafting has become popular for repair of postsurgical/postradiation defects after head/neck cancers resection. Fat graft supplementation with adipose tissue-derived stromal/stem cells (ASCs) is proposed to improve graft viability/efficacy, although the impact of ASCs on head/neck cancer cells is unknown. OBJECTIVES To determine whether ASCs affect growth, migration, and metastasis of human head/neck cancer. METHODS Human Cal-27 and SCC-4 head/neck cancer cells were co-cultured human ASCs, or treated with ASC conditioned medium (CM), and cancer cell growth/migration was assessed by MTT, cell count, and scratch/wound healing assays in vitro. Co-injection of 3 × 10(6) Cal-27/green fluorescent protein (GFP) cells and ASCs into the flank of NUDE mice assessed ASC effect on tumor growth/morphology. Quantitation of human chromosome 17 DNA in mouse organs assessed ASC effects on micrometastasis. Primary tumors were evaluated for markers of epithelial-to-mesenchymal transition, matrix metalloproteinases, and angiogenesis by immunohistochemistry. RESULTS Co-culture of Cal-27 or SCC-4 cells with ASCs from 2 different donors or ASC CM had no effect on cell growth in vitro. However, ASC CM stimulated Cal-27 and SCC-4 migration. Co-injection of ASCs from 2 different donors with Cal-27 cells did not affect tumor volume at 6 weeks, but increased Cal-27 micrometastasis to the brain. Evaluation of tumors sections from 1 ASC donor co-injection revealed that ASCs were viable and well integrated with Cal-27/GFP cells. These tumors exhibited increased MMP2, MMP9, IL-8, and microvessel density. CONCLUSIONS Human ASCs did not alter growth of human head/neck cancer cells or tumor xenografts, but stimulated migration and early micrometastasis to mouse brain.
Collapse
Affiliation(s)
- Brian G Rowan
- Dr Rowan is Piltz Professor of Cancer Research, Tulane Cancer Center, and Associate Professor and Interim Chair, Department of Structural and Cellular Biology, Tulane University School of Medicine, New Orleans, Louisiana. Drs Lacayo and Sheng are Postdoctoral Researchers, Dr Anbalagan is an Instructor, and Mr Jones is a Technician, Department of Structural and Cellular Biology, Tulane University School of Medicine, New Orleans, Louisiana. Dr Gimble is Adjunct Professor, Departments of Medicine, Surgery, and Structural and Cellular Biology, and the Center for Stem Cell Research and Regenerative Medicine, Tulane University School of Medicine, New Orleans, Louisiana. Mr Joseph is a Medical Student and Dr Chiu is an Associate Professor, Department of Plastic Surgery, New York University Langone Medical Center, New York, New York. Dr Friedlander is the Chairman, Department of Otolaryngology, Tulane University School of Medicine, New Orleans, Louisiana
| | - Eduardo A Lacayo
- Dr Rowan is Piltz Professor of Cancer Research, Tulane Cancer Center, and Associate Professor and Interim Chair, Department of Structural and Cellular Biology, Tulane University School of Medicine, New Orleans, Louisiana. Drs Lacayo and Sheng are Postdoctoral Researchers, Dr Anbalagan is an Instructor, and Mr Jones is a Technician, Department of Structural and Cellular Biology, Tulane University School of Medicine, New Orleans, Louisiana. Dr Gimble is Adjunct Professor, Departments of Medicine, Surgery, and Structural and Cellular Biology, and the Center for Stem Cell Research and Regenerative Medicine, Tulane University School of Medicine, New Orleans, Louisiana. Mr Joseph is a Medical Student and Dr Chiu is an Associate Professor, Department of Plastic Surgery, New York University Langone Medical Center, New York, New York. Dr Friedlander is the Chairman, Department of Otolaryngology, Tulane University School of Medicine, New Orleans, Louisiana
| | - Mei Sheng
- Dr Rowan is Piltz Professor of Cancer Research, Tulane Cancer Center, and Associate Professor and Interim Chair, Department of Structural and Cellular Biology, Tulane University School of Medicine, New Orleans, Louisiana. Drs Lacayo and Sheng are Postdoctoral Researchers, Dr Anbalagan is an Instructor, and Mr Jones is a Technician, Department of Structural and Cellular Biology, Tulane University School of Medicine, New Orleans, Louisiana. Dr Gimble is Adjunct Professor, Departments of Medicine, Surgery, and Structural and Cellular Biology, and the Center for Stem Cell Research and Regenerative Medicine, Tulane University School of Medicine, New Orleans, Louisiana. Mr Joseph is a Medical Student and Dr Chiu is an Associate Professor, Department of Plastic Surgery, New York University Langone Medical Center, New York, New York. Dr Friedlander is the Chairman, Department of Otolaryngology, Tulane University School of Medicine, New Orleans, Louisiana
| | - Muralidharan Anbalagan
- Dr Rowan is Piltz Professor of Cancer Research, Tulane Cancer Center, and Associate Professor and Interim Chair, Department of Structural and Cellular Biology, Tulane University School of Medicine, New Orleans, Louisiana. Drs Lacayo and Sheng are Postdoctoral Researchers, Dr Anbalagan is an Instructor, and Mr Jones is a Technician, Department of Structural and Cellular Biology, Tulane University School of Medicine, New Orleans, Louisiana. Dr Gimble is Adjunct Professor, Departments of Medicine, Surgery, and Structural and Cellular Biology, and the Center for Stem Cell Research and Regenerative Medicine, Tulane University School of Medicine, New Orleans, Louisiana. Mr Joseph is a Medical Student and Dr Chiu is an Associate Professor, Department of Plastic Surgery, New York University Langone Medical Center, New York, New York. Dr Friedlander is the Chairman, Department of Otolaryngology, Tulane University School of Medicine, New Orleans, Louisiana
| | - Jeffrey M Gimble
- Dr Rowan is Piltz Professor of Cancer Research, Tulane Cancer Center, and Associate Professor and Interim Chair, Department of Structural and Cellular Biology, Tulane University School of Medicine, New Orleans, Louisiana. Drs Lacayo and Sheng are Postdoctoral Researchers, Dr Anbalagan is an Instructor, and Mr Jones is a Technician, Department of Structural and Cellular Biology, Tulane University School of Medicine, New Orleans, Louisiana. Dr Gimble is Adjunct Professor, Departments of Medicine, Surgery, and Structural and Cellular Biology, and the Center for Stem Cell Research and Regenerative Medicine, Tulane University School of Medicine, New Orleans, Louisiana. Mr Joseph is a Medical Student and Dr Chiu is an Associate Professor, Department of Plastic Surgery, New York University Langone Medical Center, New York, New York. Dr Friedlander is the Chairman, Department of Otolaryngology, Tulane University School of Medicine, New Orleans, Louisiana
| | - Ryan K Jones
- Dr Rowan is Piltz Professor of Cancer Research, Tulane Cancer Center, and Associate Professor and Interim Chair, Department of Structural and Cellular Biology, Tulane University School of Medicine, New Orleans, Louisiana. Drs Lacayo and Sheng are Postdoctoral Researchers, Dr Anbalagan is an Instructor, and Mr Jones is a Technician, Department of Structural and Cellular Biology, Tulane University School of Medicine, New Orleans, Louisiana. Dr Gimble is Adjunct Professor, Departments of Medicine, Surgery, and Structural and Cellular Biology, and the Center for Stem Cell Research and Regenerative Medicine, Tulane University School of Medicine, New Orleans, Louisiana. Mr Joseph is a Medical Student and Dr Chiu is an Associate Professor, Department of Plastic Surgery, New York University Langone Medical Center, New York, New York. Dr Friedlander is the Chairman, Department of Otolaryngology, Tulane University School of Medicine, New Orleans, Louisiana
| | - Walter J Joseph
- Dr Rowan is Piltz Professor of Cancer Research, Tulane Cancer Center, and Associate Professor and Interim Chair, Department of Structural and Cellular Biology, Tulane University School of Medicine, New Orleans, Louisiana. Drs Lacayo and Sheng are Postdoctoral Researchers, Dr Anbalagan is an Instructor, and Mr Jones is a Technician, Department of Structural and Cellular Biology, Tulane University School of Medicine, New Orleans, Louisiana. Dr Gimble is Adjunct Professor, Departments of Medicine, Surgery, and Structural and Cellular Biology, and the Center for Stem Cell Research and Regenerative Medicine, Tulane University School of Medicine, New Orleans, Louisiana. Mr Joseph is a Medical Student and Dr Chiu is an Associate Professor, Department of Plastic Surgery, New York University Langone Medical Center, New York, New York. Dr Friedlander is the Chairman, Department of Otolaryngology, Tulane University School of Medicine, New Orleans, Louisiana
| | - Paul L Friedlander
- Dr Rowan is Piltz Professor of Cancer Research, Tulane Cancer Center, and Associate Professor and Interim Chair, Department of Structural and Cellular Biology, Tulane University School of Medicine, New Orleans, Louisiana. Drs Lacayo and Sheng are Postdoctoral Researchers, Dr Anbalagan is an Instructor, and Mr Jones is a Technician, Department of Structural and Cellular Biology, Tulane University School of Medicine, New Orleans, Louisiana. Dr Gimble is Adjunct Professor, Departments of Medicine, Surgery, and Structural and Cellular Biology, and the Center for Stem Cell Research and Regenerative Medicine, Tulane University School of Medicine, New Orleans, Louisiana. Mr Joseph is a Medical Student and Dr Chiu is an Associate Professor, Department of Plastic Surgery, New York University Langone Medical Center, New York, New York. Dr Friedlander is the Chairman, Department of Otolaryngology, Tulane University School of Medicine, New Orleans, Louisiana
| | - Ernest S Chiu
- Dr Rowan is Piltz Professor of Cancer Research, Tulane Cancer Center, and Associate Professor and Interim Chair, Department of Structural and Cellular Biology, Tulane University School of Medicine, New Orleans, Louisiana. Drs Lacayo and Sheng are Postdoctoral Researchers, Dr Anbalagan is an Instructor, and Mr Jones is a Technician, Department of Structural and Cellular Biology, Tulane University School of Medicine, New Orleans, Louisiana. Dr Gimble is Adjunct Professor, Departments of Medicine, Surgery, and Structural and Cellular Biology, and the Center for Stem Cell Research and Regenerative Medicine, Tulane University School of Medicine, New Orleans, Louisiana. Mr Joseph is a Medical Student and Dr Chiu is an Associate Professor, Department of Plastic Surgery, New York University Langone Medical Center, New York, New York. Dr Friedlander is the Chairman, Department of Otolaryngology, Tulane University School of Medicine, New Orleans, Louisiana
| |
Collapse
|
24
|
Zheng MW, Zhang CH, Chen K, Huang M, Li YP, Lin WT, Zhang RJ, Zhong L, Xiang R, Li LL, Liu XY, Wei YQ, Yang SY. Preclinical Evaluation of a Novel Orally Available SRC/Raf/VEGFR2 Inhibitor, SKLB646, in the Treatment of Triple-Negative Breast Cancer. Mol Cancer Ther 2015; 15:366-78. [PMID: 26721945 DOI: 10.1158/1535-7163.mct-15-0501] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2015] [Accepted: 11/20/2015] [Indexed: 11/16/2022]
Affiliation(s)
- Ming-Wu Zheng
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China. Collaborative Innovation Center for Biotherapy, Chengdu, Sichuan, China
| | - Chun-Hui Zhang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China. Collaborative Innovation Center for Biotherapy, Chengdu, Sichuan, China
| | - Kai Chen
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China. Collaborative Innovation Center for Biotherapy, Chengdu, Sichuan, China
| | - Mei Huang
- Key Laboratory of Drug Targeting and Drug Delivery System of Ministry of Education, West China School of Pharmacy, Sichuan University, Chengdu, Sichuan, China
| | - Ya-Ping Li
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China. Collaborative Innovation Center for Biotherapy, Chengdu, Sichuan, China
| | - Wan-Ting Lin
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China. Collaborative Innovation Center for Biotherapy, Chengdu, Sichuan, China
| | - Rong-Jie Zhang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China. Collaborative Innovation Center for Biotherapy, Chengdu, Sichuan, China
| | - Lei Zhong
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China. Collaborative Innovation Center for Biotherapy, Chengdu, Sichuan, China
| | - Rong Xiang
- Department of Clinical Medicine, School of Medicine, Nankai University, Tianjin, China
| | - Lin-Li Li
- Key Laboratory of Drug Targeting and Drug Delivery System of Ministry of Education, West China School of Pharmacy, Sichuan University, Chengdu, Sichuan, China
| | - Xin-Yu Liu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China. Collaborative Innovation Center for Biotherapy, Chengdu, Sichuan, China
| | - Yu-Quan Wei
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China. Collaborative Innovation Center for Biotherapy, Chengdu, Sichuan, China
| | - Sheng-Yong Yang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China. Collaborative Innovation Center for Biotherapy, Chengdu, Sichuan, China.
| |
Collapse
|
25
|
Zhang G, Liu X, Li J, Ledet E, Alvarez X, Qi Y, Fu X, Sartor O, Dong Y, Zhang H. Androgen receptor splice variants circumvent AR blockade by microtubule-targeting agents. Oncotarget 2015; 6:23358-71. [PMID: 26160840 PMCID: PMC4695123 DOI: 10.18632/oncotarget.4396] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2015] [Accepted: 06/09/2015] [Indexed: 01/25/2023] Open
Abstract
Docetaxel-based chemotherapy is established as a first-line treatment and standard of care for patients with metastatic castration-resistant prostate cancer. However, half of the patients do not respond to treatment and those do respond eventually become refractory. A better understanding of the resistance mechanisms to taxane chemotherapy is both urgent and clinical significant, as taxanes (docetaxel and cabazitaxel) are being used in various clinical settings. Sustained signaling through the androgen receptor (AR) has been established as a hallmark of CRPC. Recently, splicing variants of AR (AR-Vs) that lack the ligand-binding domain (LBD) have been identified. These variants are constitutively active and drive prostate cancer growth in a castration-resistant manner. In taxane-resistant cell lines, we found the expression of a major variant, AR-V7, was upregulated. Furthermore, ectopic expression of two clinically relevant AR-Vs (AR-V7 and ARV567es), but not the full-length AR (AR-FL), reduced the sensitivities to taxanes in LNCaP cells. Treatment with taxanes inhibited the transcriptional activity of AR-FL, but not those of AR-Vs. This could be explained, at least in part, due to the inability of taxanes to block the nuclear translocation of AR-Vs. Through a series of deletion constructs, the microtubule-binding activity was mapped to the LBD of AR. Finally, taxane-induced cytoplasm sequestration of AR-FL was alleviated when AR-Vs were present. These findings provide evidence that constitutively active AR-Vs maintain the AR signaling axis by evading the inhibitory effects of microtubule-targeting agents, suggesting that these AR-Vs play a role in resistance to taxane chemotherapy.
Collapse
Affiliation(s)
- Guanyi Zhang
- College of Life Sciences, Jilin University, Changchun, P.R. China
- Department of Pathology, Tulane University School of Medicine, New Orleans, Louisiana, USA
- Tulane Cancer Center, Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Xichun Liu
- Department of Pathology, Tulane University School of Medicine, New Orleans, Louisiana, USA
- Tulane Cancer Center, Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Jianzhuo Li
- College of Life Sciences, Jilin University, Changchun, P.R. China
- Department of Pathology, Tulane University School of Medicine, New Orleans, Louisiana, USA
- Tulane Cancer Center, Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Elisa Ledet
- Department of Medicine, Tulane University School of Medicine, New Orleans, Louisiana, USA
- Department of Urology, Tulane University School of Medicine, New Orleans, Louisiana, USA
- Tulane Cancer Center, Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Xavier Alvarez
- Division of Comparative Pathology, Tulane National Primate Research Center, Covington, Louisiana, USA
| | - Yanfeng Qi
- Department of Structural and Cellular Biology, Tulane University School of Medicine, New Orleans, Louisiana, USA
- Tulane Cancer Center, Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Xueqi Fu
- College of Life Sciences, Jilin University, Changchun, P.R. China
| | - Oliver Sartor
- Department of Medicine, Tulane University School of Medicine, New Orleans, Louisiana, USA
- Department of Urology, Tulane University School of Medicine, New Orleans, Louisiana, USA
- Tulane Cancer Center, Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Yan Dong
- College of Life Sciences, Jilin University, Changchun, P.R. China
- Department of Structural and Cellular Biology, Tulane University School of Medicine, New Orleans, Louisiana, USA
- Tulane Cancer Center, Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Haitao Zhang
- Department of Pathology, Tulane University School of Medicine, New Orleans, Louisiana, USA
- Tulane Cancer Center, Tulane University School of Medicine, New Orleans, Louisiana, USA
| |
Collapse
|
26
|
Abstract
Knowledge of the molecular events that contribute to prostate cancer progression has created opportunities to develop novel therapy strategies. It is now well established that c-Src, a non-receptor tyrosine kinase, regulates a complex signaling network that drives the development of castrate-resistance and bone metastases, events that signal the lethal phenotype of advanced disease. Preclinical studies have established a role for c-Src and Src Family Kinases (SFKs) in proliferation, angiogenesis, invasion and bone metabolism, thus implicating Src signaling in both epithelial and stromal mechanisms of disease progression. A number of small molecule inhibitors of SFK now exist, many of which have demonstrated efficacy in preclinical models and several that have been tested in patients with metastatic castrate-resistant prostate cancer. These agents have demonstrated provocative clinic activity, particularly in modulating the bone microenvironment in a therapeutically favorable manner. Here, we review the discovery and basic biology of c-Src and further discuss the role of SFK inhibitors in the treatment of advanced prostate cancer.
Collapse
|
27
|
Salzano G, Navarro G, Trivedi MS, De Rosa G, Torchilin VP. Multifunctional Polymeric Micelles Co-loaded with Anti-Survivin siRNA and Paclitaxel Overcome Drug Resistance in an Animal Model of Ovarian Cancer. Mol Cancer Ther 2015; 14:1075-84. [PMID: 25657335 DOI: 10.1158/1535-7163.mct-14-0556] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2014] [Accepted: 01/29/2015] [Indexed: 12/23/2022]
Abstract
Ovarian cancer is a dreadful disease estimated to be the second most common gynecologic malignancy worldwide. Its current therapy, based on cytoreductive surgery followed by the combination of platinum and taxanes, is frequently complicated by the onset of multidrug resistance (MDR). The discovery that survivin, a small antiapoptotic protein, is involved in chemoresistance provided a new prospect to overcome MDR in cancer, because siRNA could be used to inhibit the expression of survivin in cancer cells. With this in mind, we have developed self-assembly polymeric micelles (PM) able to efficiently co-load an anti-survivin siRNA and a chemotherapeutic agent, such as paclitaxel (PXL; survivin siRNA/PXL PM). Previously, we have successfully demonstrated that the downregulation of survivin by using siRNA-containing PM strongly sensitizes different cancer cells to paclitaxel. Here, we have evaluated the applicability of the developed multifunctional PM in vivo. Changes in survivin expression, therapeutic efficacy, and biologic effects of the nanopreparation were investigated in an animal model of paclitaxel-resistant ovarian cancer. The results obtained in mice xenografed with SKOV3-tr revealed a significant downregulation of survivin expression in tumor tissues together with a potent anticancer activity of survivin siRNA/PXL PM, while the tumors remained unaffected with the same quantity of free paclitaxel. These promising results introduce a novel type of nontoxic and easy-to-obtain nanodevice for the combined therapy of siRNA and anticancer agents in the treatment of chemoresistant tumors.
Collapse
Affiliation(s)
- Giuseppina Salzano
- Center for Pharmaceutical Biotechnology and Nanomedicine, Northeastern University, Boston, Massachusetts. Department of Pharmacy, School of Pharmacy Federico II, Naples, Italy
| | - Gemma Navarro
- Center for Pharmaceutical Biotechnology and Nanomedicine, Northeastern University, Boston, Massachusetts
| | - Malav S Trivedi
- Department of Pharmaceutical Sciences, Northeastern University, Boston, Massachusetts
| | - Giuseppe De Rosa
- Department of Pharmacy, School of Pharmacy Federico II, Naples, Italy
| | - Vladimir P Torchilin
- Center for Pharmaceutical Biotechnology and Nanomedicine, Northeastern University, Boston, Massachusetts. Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia.
| |
Collapse
|
28
|
Breen ME, Soellner MB. Small molecule substrate phosphorylation site inhibitors of protein kinases: approaches and challenges. ACS Chem Biol 2015; 10:175-89. [PMID: 25494294 PMCID: PMC4301090 DOI: 10.1021/cb5008376] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
![]()
Protein kinases are
important mediators of cellular communication
and attractive drug targets for many diseases. Although success has
been achieved with developing ATP-competitive kinase inhibitors, the
disadvantages of ATP-competitive inhibitors have led to increased
interest in targeting sites outside of the ATP binding pocket. Kinase
inhibitors with substrate-competitive, ATP-noncompetitive binding
modes are promising due to the possibility of increased selectivity
and better agreement between biochemical and in vitro potency. However, the difficulty of identifying these types of inhibitors
has resulted in significantly fewer small molecule substrate phosphorylation
site inhibitors being reported compared to ATP-competitive inhibitors.
This review surveys reported substrate phosphorylation site inhibitors
and methods that can be applied to the discovery of such inhibitors,
including a discussion of the challenges inherent to these screening
methods.
Collapse
Affiliation(s)
- Meghan E. Breen
- Department of Medicinal Chemistry and ‡Department of
Chemistry, University of Michigan, 930 N. University Avenue, Ann Arbor, Michigan 48109, United States
| | - Matthew B. Soellner
- Department of Medicinal Chemistry and ‡Department of
Chemistry, University of Michigan, 930 N. University Avenue, Ann Arbor, Michigan 48109, United States
| |
Collapse
|
29
|
Zimmer AS, Steeg PS. Meaningful prevention of breast cancer metastasis: candidate therapeutics, preclinical validation, and clinical trial concerns. J Mol Med (Berl) 2015; 93:13-29. [PMID: 25412774 PMCID: PMC6545582 DOI: 10.1007/s00109-014-1226-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2014] [Revised: 10/08/2014] [Accepted: 10/30/2014] [Indexed: 12/31/2022]
Abstract
The development of drugs to treat breast and other cancers proceeds through phase I dose finding, phase II efficacy, and phase III comparative studies in the metastatic setting, only then asking if metastasis can be prevented in adjuvant trials. Compounds without overt cytotoxic activity, such as those developed to inhibit metastatic colonization, will likely fail to shrink established lesions in the metastatic setting and never be tested in a metastasis prevention scenario where they were preclinically validated. We and others have proposed phase II primary and secondary metastasis prevention studies to address this need. Herein, we have asked whether preclinical metastasis prevention data agrees with the positive adjuvant setting trials. The data are limited but complimentary. We also review fundamental pathways involved in metastasis, including Src, integrins, focal adhesion kinase (FAK), and fibrosis, for their clinical progress to date and potential for metastasis prevention. Issues of inadequate preclinical validation and clinical toxicity profiles are discussed.
Collapse
Affiliation(s)
- Alexandra S Zimmer
- Women's Malignancies Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, 20892, USA,
| | | |
Collapse
|
30
|
Delle Monache S, Sanità P, Calgani A, Schenone S, Botta L, Angelucci A. Src inhibition potentiates antitumoral effect of paclitaxel by blocking tumor-induced angiogenesis. Exp Cell Res 2014; 328:20-31. [PMID: 25128812 DOI: 10.1016/j.yexcr.2014.08.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2014] [Revised: 07/31/2014] [Accepted: 08/02/2014] [Indexed: 12/25/2022]
Abstract
The protein kinase Src is frequently over-activated in advanced cancers where it modulates the signaling transduction cascade of several growth factors. The feasibility of combination treatment of Src inhibitors with chemotherapy is currently under investigation. We evaluated the anti-tumoral effect of paclitaxel (PTX) in combination with S13, a tyrosine kinase inhibitor with a prevalent specificity for Src, in a hormone-insensible prostate cancer (PCa) cell model. In vivo, combination treatment with PTX and S13 reduced dramatically PCa tumor growth with a relevant difference in the density of new blood vessels with respect to control and single treatments. This reduction was determined by a concomitant impairment of endothelial cell migration and of VEGF release by cancer cells. In fact, S13, when used alone, was sufficient to reduce tubule formation in vivo, and to inhibit VEGFR2 activation and FAK expression in endothelial cells. In addition, the combination treatment determined a significant reduction in ROS production and HIF-1 stabilization in PCa cells respect to single treatments with S13 or PTX. In conclusion, Src-inhibition could be an effective therapeutic strategy aimed at supporting the anti-angiogenic action of PTX in aggressive PCa.
Collapse
Affiliation(s)
- Simona Delle Monache
- Dipartimento di Scienze Cliniche Applicate e Biotecnologiche, University of L׳Aquila, via Vetoio Coppito, 67100 L׳Aquila, Italy.
| | - Patrizia Sanità
- Dipartimento di Scienze Cliniche Applicate e Biotecnologiche, University of L׳Aquila, via Vetoio Coppito, 67100 L׳Aquila, Italy
| | - Alessia Calgani
- Dipartimento di Scienze Cliniche Applicate e Biotecnologiche, University of L׳Aquila, via Vetoio Coppito, 67100 L׳Aquila, Italy
| | - Silvia Schenone
- Dipartimento di Farmacia, University of Genova, viale Benedetto XV, 3, 16132 Genova, Italy
| | - Lorenzo Botta
- Dipartimento di Biotecnologie, Chimica e Farmacia, University of via A. Moro, snc, 53100, Siena, Italy
| | - Adriano Angelucci
- Dipartimento di Scienze Cliniche Applicate e Biotecnologiche, University of L׳Aquila, via Vetoio Coppito, 67100 L׳Aquila, Italy
| |
Collapse
|
31
|
Identification of DW532 as a novel anti-tumor agent targeting both kinases and tubulin. Acta Pharmacol Sin 2014; 35:916-28. [PMID: 24858311 DOI: 10.1038/aps.2014.33] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2014] [Accepted: 04/07/2014] [Indexed: 01/19/2023] Open
Abstract
AIM 7,8-Dihydroxy-4-(3-hydroxy-4-methoxyphenyl)-2H-chromen-2-one (DW532) is one of simplified analogues of hematoxylin that has shown broad-spectrum inhibition on tyrosine kinases and in vitro anti-cancer activities. The aim of this study was to identify DW532 as a agent targeting both kinases and tubulin, and to investigate its anti-cancer and anti-angiogenesis activities. METHODS In vitro tyrosine kinases activity was examined with ELISA, and tyrosine kinases activity in cells was evaluated with Western blot analysis. Tubulin turbidity assay, surface plasmon resonance and immunofluorescence technique were used to characterize the tubulin inhibitory activity. Cell proliferation was examined with SRB assay, and cell apoptosis and cell cycle distribution were analyzed with Annexin-V/PI staining and flow cytometry. Tube formation, aortic ring and chick chorioallantoic membrane assays were used to evaluate the anti-angiogenesis efficacy. RESULTS DW532 inhibited EGFR and VEGFR2 in vitro kinase activity (the IC50 values were 4.9 and 5.5 μmol/L, respectively), and suppressed their downstream signaling. DW532 dose-dependently inhibited tubulin polymerization via direct binding to tubulin, thus disrupting the mitotic spindle assembly and leading to abnormal cell division. In a panel of human cancer cells, DW532 (1 and 10 μmol/L) induced G2/M phase arrest and cell apoptosis, which subsequently resulted in cytotoxicity. Knockdown of BubR1 or Mps1, the two core proteins of the spindle assembly checkpoint dramatically decreased DW532-induced cell cycle arrest in MDA-MB-468 cells. Moreover, treatment with DW532 potently and dose-dependently suppressed angiogenesis in vitro and in vivo. CONCLUSION DW532 is a dual inhibitor against tubulin and tyrosine kinases, and deserves further development as a novel anti-cancer agent.
Collapse
|
32
|
Rowan BG, Gimble JM, Sheng M, Anbalagan M, Jones RK, Frazier TP, Asher M, Lacayo EA, Friedlander PL, Kutner R, Chiu ES. Human adipose tissue-derived stromal/stem cells promote migration and early metastasis of triple negative breast cancer xenografts. PLoS One 2014; 9:e89595. [PMID: 24586900 PMCID: PMC3938488 DOI: 10.1371/journal.pone.0089595] [Citation(s) in RCA: 141] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2012] [Accepted: 01/22/2014] [Indexed: 01/01/2023] Open
Abstract
Background Fat grafting is used to restore breast defects after surgical resection of breast tumors. Supplementing fat grafts with adipose tissue-derived stromal/stem cells (ASCs) is proposed to improve the regenerative/restorative ability of the graft and retention. However, long term safety for ASC grafting in proximity of residual breast cancer cells is unknown. The objective of this study was to determine the impact of human ASCs derived from abdominal lipoaspirates of three donors, on a human breast cancer model that exhibits early metastasis. Methodology/Principal Findings Human MDA-MB-231 breast cancer cells represents “triple negative” breast cancer that exhibits early micrometastasis to multiple mouse organs [1]. Human ASCs were derived from abdominal adipose tissue from three healthy female donors. Indirect co-culture of MDA-MB-231 cells with ASCs, as well as direct co-culture demonstrated that ASCs had no effect on MDA-MB-231 growth. Indirect co-culture, and ASC conditioned medium (CM) stimulated migration of MDA-MB-231 cells. ASC/RFP cells from two donors co-injected with MDA-MB-231/GFP cells exhibited a donor effect for stimulation of primary tumor xenografts. Both ASC donors stimulated metastasis. ASC/RFP cells were viable, and integrated with MDA-MB-231/GFP cells in the tumor. Tumors from the co-injection group of one ASC donor exhibited elevated vimentin, matrix metalloproteinase-9 (MMP-9), IL-8, VEGF and microvessel density. The co-injection group exhibited visible metastases to the lung/liver and enlarged spleen not evident in mice injected with MDA-MB-231/GFP alone. Quantitation of the total area of GFP fluorescence and human chromosome 17 DNA in mouse organs, H&E stained paraffin sections and fluorescent microscopy confirmed multi-focal metastases to lung/liver/spleen in the co-injection group without evidence of ASC/RFP cells. Conclusions Human ASCs derived from abdominal lipoaspirates of two donors stimulated metastasis of MDA-MB-231 breast tumor xenografts to multiple mouse organs. MDA-MB-231 tumors co-injected with ASCs from one donor exhibited partial EMT, expression of MMP-9, and increased angiogenesis.
Collapse
Affiliation(s)
- Brian G. Rowan
- Department of Structural and Cellular Biology, Tulane University School of Medicine, New Orleans, Louisiana, United States of America
- * E-mail: (BGR); (ESC)
| | - Jeffrey M. Gimble
- Stem Cell Biology Laboratory, Pennington Biomedical Research Center, Louisiana State University, Baton Rouge, Louisiana, United States of America
| | - Mei Sheng
- Department of Otolaryngology, Tulane University School of Medicine, New Orleans, Louisiana, United States of America
| | - Muralidharan Anbalagan
- Department of Structural and Cellular Biology, Tulane University School of Medicine, New Orleans, Louisiana, United States of America
| | - Ryan K. Jones
- Department of Structural and Cellular Biology, Tulane University School of Medicine, New Orleans, Louisiana, United States of America
| | - Trivia P. Frazier
- Department of Structural and Cellular Biology, Tulane University School of Medicine, New Orleans, Louisiana, United States of America
| | - Majdouline Asher
- Department of Structural and Cellular Biology, Tulane University School of Medicine, New Orleans, Louisiana, United States of America
| | - Eduardo A. Lacayo
- Department of Otolaryngology, Tulane University School of Medicine, New Orleans, Louisiana, United States of America
| | - Paul L. Friedlander
- Department of Otolaryngology, Tulane University School of Medicine, New Orleans, Louisiana, United States of America
| | - Robert Kutner
- Gene Therapy Program, Louisiana State University Health Sciences Center, New Orleans, Louisiana, United States of America
| | - Ernest S. Chiu
- Department of Plastic Surgery, New York University Langone Medical Center, New York, New York, United States of America
- * E-mail: (BGR); (ESC)
| |
Collapse
|
33
|
Liu T, Hu W, Dalton HJ, Choi HJ, Huang J, Kang Y, Pradeep S, Miyake T, Song JH, Wen Y, Lu C, Pecot CV, Bottsford-Miller J, Zand B, Jennings NB, Ivan C, Gallick GE, Baggerly KA, Hangauer DG, Coleman RL, Frumovitz M, Sood AK. Targeting SRC and tubulin in mucinous ovarian carcinoma. Clin Cancer Res 2013; 19:6532-43. [PMID: 24100628 PMCID: PMC3852199 DOI: 10.1158/1078-0432.ccr-13-1305] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
PURPOSE To investigate the antitumor effects of targeting Src and tubulin in mucinous ovarian carcinoma. EXPERIMENTAL DESIGN The in vitro and in vivo effects and molecular mechanisms of KX-01, which inhibits Src pathway and tubulin polymerization, were examined in mucinous ovarian cancer models. RESULTS In vitro studies using RMUG-S and RMUG-L cell lines showed that KX-01 inhibited cell proliferation, induced apoptosis, arrested the cell cycle at the G2-M phase, and enhanced the cytotoxicity of oxaliplatin in the KX-01-sensitive cell line, RMUG-S. In vivo studies showed that KX-01 significantly decreased tumor burden in RMUG-S and RMUG-L mouse models relative to untreated controls, and the effects were greater when KX-01 was combined with oxaliplatin. KX-01 alone and in combination with oxaliplatin significantly inhibited tumor growth by reducing cell proliferation and inducing apoptosis in vivo. PTEN knock-in experiments in RMUG-L cells showed improved response to KX-01. Reverse phase protein array analysis showed that in addition to blocking downstream molecules of Src family kinases, KX-01 also activated acute stress-inducing molecules. CONCLUSION Our results showed that targeting both the Src pathway and tubulin with KX-01 significantly inhibited tumor growth in preclinical mucinous ovarian cancer models, suggesting that this may be a promising therapeutic approach for patients with mucinous ovarian carcinoma.
Collapse
Affiliation(s)
- Tao Liu
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- Department of General Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Wei Hu
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Heather J. Dalton
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Hyun Jin Choi
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Jie Huang
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Yu Kang
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- Department of Gynecology, Obstetrics and Gynecology, Hospital of Fudan University, Shanghai 20001, P.R. China
| | - Sunila Pradeep
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Takahito Miyake
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Jian H. Song
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Yunfei Wen
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Chunhua Lu
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Chad V. Pecot
- Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Justin Bottsford-Miller
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Behrouz Zand
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Nicholas B Jennings
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Cristina Ivan
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- Center for RNA Interference and Non-Coding RNA, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Gary E. Gallick
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Keith A Baggerly
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - David G. Hangauer
- Kinex Pharmaceuticals LLC, New York State Center of Excellence in Bioinformatics and Life Sciences, Buffalo, NY 14203, USA
| | - Robert L. Coleman
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Michael Frumovitz
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Anil K. Sood
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- Center for RNA Interference and Non-Coding RNA, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| |
Collapse
|
34
|
Marsden CG, Wright MJ, Carrier L, Moroz K, Rowan BG. Disseminated breast cancer cells acquire a highly malignant and aggressive metastatic phenotype during metastatic latency in the bone. PLoS One 2012; 7:e47587. [PMID: 23173031 PMCID: PMC3500091 DOI: 10.1371/journal.pone.0047587] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2012] [Accepted: 09/18/2012] [Indexed: 12/19/2022] Open
Abstract
Background Disseminated tumor cells (DTCs) in the bone marrow may exist in a dormant state for extended periods of time, maintaining the ability to proliferate upon activation, engraft at new sites, and form detectable metastases. However, understanding of the behavior and biology of dormant breast cancer cells in the bone marrow niche remains limited, as well as their potential involvement in tumor recurrence and metastasis. Therefore, the purpose of this study was to investigate the tumorigenicity and metastatic potential of dormant disseminated breast cancer cells (prior to activation) in the bone marrow. Methodology/Principal Findings Total bone marrow, isolated from mice previously injected with tumorspheres into the mammary fat pad, was injected into the mammary fat pad of NUDE mice. As a negative control, bone marrow isolated from non-injected mice was injected into the mammary fat pad of NUDE mice. The resultant tumors were analyzed by immunohistochemistry for expression of epithelial and mesenchymal markers. Mouse lungs, livers, and kidneys were analyzed by H+E staining to detect metastases. The injection of bone marrow isolated from mice previously injected with tumorspheres into the mammary fat pad, resulted in large tumor formation in the mammary fat pad 2 months post-injection. However, the injection of bone marrow isolated from non-injected mice did not result in tumor formation in the mammary fat pad. The DTC-derived tumors exhibited accelerated development of metastatic lesions within the lung, liver and kidney. The resultant tumors and the majority of metastatic lesions within the lung and liver exhibited a mesenchymal-like phenotype. Conclusions/Significance Dormant DTCs within the bone marrow are highly malignant upon injection into the mammary fat pad, with the accelerated development of metastatic lesions within the lung, liver and kidney. These results suggest the acquisition of a more aggressive phenotype of DTCs during metastatic latency within the bone marrow microenvironment.
Collapse
Affiliation(s)
- Carolyn G. Marsden
- Department of Structural and Cellular Biology, The Louisiana Cancer Research Consortium, Tulane University Health Sciences Center, New Orleans, Louisiana, United States of America
| | - Mary Jo Wright
- Department of Surgery, The Louisiana Cancer Research Consortium, Tulane University School of Medicine, New Orleans, Louisiana, United States of America
| | - Latonya Carrier
- Department of Structural and Cellular Biology, The Louisiana Cancer Research Consortium, Tulane University Health Sciences Center, New Orleans, Louisiana, United States of America
| | - Krzysztof Moroz
- Section of Surgical Pathology and Cytopathology, Louisiana Cancer Research Consortium, Tulane University School of Medicine, New Orleans, Louisiana, United States of America
| | - Brian G. Rowan
- Department of Structural and Cellular Biology, The Louisiana Cancer Research Consortium, Tulane University Health Sciences Center, New Orleans, Louisiana, United States of America
- * E-mail:
| |
Collapse
|