1
|
Mark M, Rusakiewicz S, Früh M, Hayoz S, Grosso F, Pless M, Zucali P, Ceresoli G, Maconi A, Schneider M, Froesch P, Tarussio D, Benedetti F, Dagher J, Kandalaft L, von Moos R, Tissot-Renaud S, Schmid S, Metaxas Y. Long-term benefit of lurbinectedin as palliative chemotherapy in progressive malignant pleural mesothelioma (MPM): final efficacy and translational data of the SAKK 17/16 study. ESMO Open 2022; 7:100446. [PMID: 35427834 PMCID: PMC9271468 DOI: 10.1016/j.esmoop.2022.100446] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 01/24/2022] [Accepted: 02/09/2022] [Indexed: 01/01/2023] Open
Abstract
Background The SAKK 17/16 study showed promising efficacy data with lurbinectedin as second- or third-line palliative therapy in malignant pleural mesothelioma. Here, we evaluated long-term outcome and analyzed the impact of lurbinectedin monotherapy on the tumor microenvironment at the cellular and molecular level to predict outcomes. Material and methods Forty-two patients were treated with lurbinectedin in this single-arm study. Twenty-nine samples were available at baseline, and seven additional matched samples at day one of cycle two of treatment. Survival curves and rates between groups were compared using the log-rank test and Kaplan–Meier method. Statistical significance was set at P value <0.05. Results Updated median overall survival (OS) was slightly increased to 11.5 months [95% confidence interval (CI) 8.8-13.8 months]. Thirty-six patients (85%) had died. The OS rate at 12 and 18 months was 47% (95% CI 32.1% to 61.6%) and 31% (95% CI 17.8% to 45.0%), respectively. Median progression-free survival was 4.1 months (95% CI 2.6-5.5 months). No new safety signals were observed. Patients with lower frequencies of regulatory T cells, as well as lower tumor-associated macrophages (TAMs) at baseline, had a better OS. Comparing matched biopsies, a decrease of M2 macrophages was observed in five out of seven patients after exposure to lurbinectedin, and two out of four patients showed increased CD8+ T-cell infiltrates in tumor. Discussion Lurbinectedin continues to be active in patients with progressing malignant pleural mesothelioma. According to our very small sample size, we hypothesize that baseline TAMs and regulatory T cells are associated with survival. Lurbinectedin seems to inhibit conversion of TAMs to M2 phenotype in humans. Lurbinectedin continues to be active in patients with progressing MPM with a median OS of 11.5 months (95% CI 8.8-13.8 months). TAMs and regulatory T cells are associated with survival. Lurbinectedin seems to inhibit conversion of TAMs to M2 phenotype in humans.
Collapse
|
2
|
Popat S, Curioni-Fontecedro A, Dafni U, Shah R, O'Brien M, Pope A, Fisher P, Spicer J, Roy A, Gilligan D, Gautschi O, Nadal E, Janthur WD, López Castro R, García Campelo R, Rusakiewicz S, Letovanec I, Polydoropoulou V, Roschitzki-Voser H, Ruepp B, Gasca-Ruchti A, Peters S, Stahel RA. A multicentre randomised phase III trial comparing pembrolizumab versus single-agent chemotherapy for advanced pre-treated malignant pleural mesothelioma: the European Thoracic Oncology Platform (ETOP 9-15) PROMISE-meso trial. Ann Oncol 2020; 31:1734-1745. [PMID: 32976938 DOI: 10.1016/j.annonc.2020.09.009] [Citation(s) in RCA: 167] [Impact Index Per Article: 33.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Revised: 08/19/2020] [Accepted: 09/13/2020] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Malignant pleural mesothelioma (MPM) is an aggressive malignancy characterised by limited treatment options and a poor prognosis. At relapse after platinum-based chemotherapy, single-agent chemotherapy is commonly used and single-arm trials of immune-checkpoint inhibitors have demonstrated encouraging activity. PATIENTS AND METHODS PROMISE-meso is an open-label 1:1 randomised phase III trial investigating the efficacy of pembrolizumab (200 mg/Q3W) versus institutional choice single-agent chemotherapy (gemcitabine or vinorelbine) in relapsed MPM patients with progression after/on previous platinum-based chemotherapy. Patients were performance status 0-1 and unselected for programmed cell death ligand 1 (PD-L1) status. At progression, patients randomly assigned to receive chemotherapy were allowed to crossover to pembrolizumab. The primary end point was progression-free survival (PFS), assessed by blinded independent central review (BICR). Secondary end points were overall survival (OS), investigator-assessed PFS, objective response rate (ORR), and safety. Efficacy by PD-L1 status was investigated in exploratory analyses. RESULTS Between September 2017 and August 2018, 144 patients were randomly allocated (pembrolizumab: 73; chemotherapy: 71). At data cut-off [20 February 2019, median follow-up of 11.8 months (interquartile range: 9.9-14.5)], 118 BICR-PFS events were observed. No difference in BICR-PFS was detected [hazard ratio = 1.06, 95% confidence interval (CI): 0.73-1.53; P = 0.76], and median BICR-PFS (95% CI) for pembrolizumab was 2.5 (2.1-4.2), compared with 3.4 (2.2-4.3) months for chemotherapy. A difference in ORR for pembrolizumab was identified (22%, 95% CI: 13% to 33%), over chemotherapy (6%, 95% CI: 2% to 14%; P = 0.004). Forty-five patients (63%) assigned to chemotherapy received pembrolizumab at progression. With follow-up to 21 August 2019 [17.5 months: (14.8-19.7)], no difference in OS was detected between groups (HR = 1.12, 95% CI: 0.74-1.69; P = 0.59), even after adjusting for crossover. Pembrolizumab safety was consistent with previous observations. Exploratory efficacy analyses by PD-L1 status demonstrated no improvements in ORR/PFS/OS. CONCLUSION This is the first randomised trial evaluating the efficacy of pembrolizumab in MPM patients progressing after/on previous platinum-based chemotherapy. In biologically unselected patients, although associated with an improved ORR, pembrolizumab improves neither PFS nor OS over single-agent chemotherapy.
Collapse
Affiliation(s)
- S Popat
- Royal Marsden Hospital Fulham Road, London, UK
| | - A Curioni-Fontecedro
- University Hospital Zürich, Department of Medical Oncology and Hematology, Zürich, Switzerland
| | - U Dafni
- National and Kapodistrian University of Athens & Frontier Science Foundation-Hellas, Athens, Greece
| | - R Shah
- Kent Oncology Centre, Maidstone, UK
| | - M O'Brien
- Royal Marsden Hospital Sutton, London, UK
| | - A Pope
- Clatterbridge Cancer Centre, Liverpool, UK
| | - P Fisher
- Weston Park Hospital, Sheffield, UK
| | - J Spicer
- King's College London, Guy's Hospital, London, UK
| | - A Roy
- University Hospital Plymouth, Plymouth, UK
| | | | - O Gautschi
- University of Bern and Cantonal Hospital Luzern, Luzern Swiss Group for Clinical Cancer Research (SAKK), Bern, Switzerland; Swiss Group for Clinical Cancer Research, Switzerland (SAKK), Bern
| | - E Nadal
- Catalan Institute of Oncology (ICO), L'Hospitalet, Barcelona, Spain
| | - W D Janthur
- Swiss Group for Clinical Cancer Research, Switzerland (SAKK), Bern; Cantonal Hospital Aarau, Aarau, Switzerland
| | - R López Castro
- Hospital Clínico Universitario de Valladolid, Valladolid, Spain
| | | | - S Rusakiewicz
- Centre Hospitalier Universitaire Vaudois CHUV, Centre of Experimental Therapies and Department of Oncology, Lausanne, Switzerland
| | - I Letovanec
- Centre Hospitalier Universitaire Vaudois CHUV, Institute of Pathology, Lausanne, Switzerland
| | | | | | - B Ruepp
- European Thoracic Oncology Platform (ETOP), Bern, Switzerland
| | - A Gasca-Ruchti
- European Thoracic Oncology Platform (ETOP), Bern, Switzerland
| | - S Peters
- Centre Hospitalier Universitaire Vaudois, Department of Oncology, Lausanne, Switzerland
| | - R A Stahel
- University Hospital Zürich, Department of Medical Oncology and Hematology, Zürich, Switzerland.
| |
Collapse
|
3
|
Metaxas Y, Früh M, Eboulet EI, Grosso F, Pless M, Zucali PA, Ceresoli GL, Mark M, Schneider M, Maconi A, Perrino M, Biaggi-Rudolf C, Froesch P, Schmid S, Waibel C, Appenzeller C, Rauch D, von Moos R. Lurbinectedin as second- or third-line palliative therapy in malignant pleural mesothelioma: an international, multi-centre, single-arm, phase II trial (SAKK 17/16). Ann Oncol 2020; 31:495-500. [PMID: 32085891 DOI: 10.1016/j.annonc.2019.12.009] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Revised: 12/20/2019] [Accepted: 12/24/2019] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Systemic second- and third-line therapies for malignant pleural mesothelioma (MPM) result in a median progression-free survival (mPFS) of <2 months and median overall survival (mOS) of 6-9 months. Lurbinectedin binds to the DNA of the regulatory region while inhibiting tumour-associated macrophage transcription. In early trials, encouraging outcomes occurred in patients (pts) with MPM treated with lurbinectedin. We aimed to generate lurbinectedin efficacy and safety data among pts with progressive MPM. PATIENTS AND METHODS Pts with progressing MPM treated with first-line platinum-pemetrexed chemotherapy with or without immunotherapy received lurbinectedin monotherapy. Treatment was given intravenously at 3.2 mg/m2 dose every 3 weeks until progression or unacceptable toxicity. Using Simon's two-stage design, the primary endpoint, progression-free survival (PFS) at 12 weeks (PFS12wks), was met if achieved by ≥21 pts (p0 ≤35% versus p1 ≥55%). RESULTS Forty-two pts from nine centres across Switzerland and Italy were recruited. Histology was epithelioid in 33 cases, sarcomatoid in 5, and biphasic in 4. Overall 10/42 (23.8%) underwent prior immunotherapy and 14/42 (33.3%) had progressed ≤6 months after first-line chemotherapy. At data cut-off PFS12wks was met by 22/42 pts (52.4%; 90% confidence interval (CI): 38.7% to 63.5%; P = 0.015) with an mPFS of 4.1 months and mOS of 11.1 months. The best response was complete and partial remission observed in one patient each and stable disease in 20 pts. The duration of disease control was 6.6 months (95% CI: 5.2-7.4). No significant difference in PFS12wks, mPFS, and mOS was recorded in epithelioid versus non-epithelioid cases and pts with prior immunotherapy versus those without. Similar mPFS but shorter mOS were observed among pts who progressed within ≤6 months after first-line chemotherapy. Lurbinectedin-related grade 3-4 toxicity was seen in 21 pts, mostly being neutropenia (23.8%) and fatigue (16.7%). CONCLUSIONS The primary efficacy endpoint was reached with acceptable toxicity. Lurbinectedin showed promising activity regardless of histology, prior immunotherapy, or outcome on prior treatment. CLINICALTRIALS. GOV IDENTIFIER NCT03213301.
Collapse
Affiliation(s)
- Y Metaxas
- Department of Oncology/Haematology, Kantonsspital Graubünden, Chur, Switzerland.
| | - M Früh
- Department of Medical Oncology and Haematology, Kantonsspital St. Gallen, St Gallen, Switzerland; University of Bern, Bern, Switzerland
| | | | - F Grosso
- Mesothelioma Unit - Oncology, SS. Antonio and C. Arrigo Hospital, Alessandria, Italy
| | - M Pless
- Department of Medical Oncology and Haematology, Kantonsspital Winterthur, Winterthur, Switzerland
| | - P A Zucali
- Humanitas Cancer Centre, Humanitas Research Hospital, Rozzano, Italy
| | - G L Ceresoli
- Oncology Unit, Humanitas Clinic Gavazzeni, Bergamo, Italy
| | - M Mark
- Department of Oncology/Haematology, Kantonsspital Graubünden, Chur, Switzerland
| | | | - A Maconi
- Scientific Research and Development Department, SS Antonio e Biagio e Cesare Arrigo, General Hospital, Alessandria, Italy
| | - M Perrino
- Humanitas Cancer Centre, Humanitas Research Hospital, Rozzano, Italy
| | | | - P Froesch
- Department of Oncology, Ospedale Regionale di Bellinzona e Valli, Bellinzona, Switzerland
| | - S Schmid
- Department of Medical Oncology and Haematology, Kantonsspital St. Gallen, St Gallen, Switzerland
| | - C Waibel
- Division of Haematology and Oncology, Kantonsspital Baden, Baden, Switzerland
| | - C Appenzeller
- Department of Medical Oncology and Haematology, Kantonsspital St. Gallen, St Gallen, Switzerland
| | - D Rauch
- Oncology Centre, Hospital STS AG, Thun, Switzerland
| | - R von Moos
- Department of Oncology/Haematology, Kantonsspital Graubünden, Chur, Switzerland
| |
Collapse
|
4
|
Ceresoli GL, Rossi A. Approved and emerging treatments of malignant pleural mesothelioma in elderly patients. Expert Rev Respir Med 2019; 13:1179-1188. [PMID: 31596154 DOI: 10.1080/17476348.2019.1678386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Introduction: Malignant pleural mesothelioma (MPM) is a rare neoplasm with asbestos exposure as the dominant etiologic agent. Owing to the long latent period following exposure, MPM is often diagnosed late in life. Despite this, elderly patients are under-represented in clinical trials. To date, data regarding the tolerability and efficacy of anticancer treatments for elderly patients affected by MPM are still lacking.Areas covered: The current state-of-the-art of approved treatments employed in the treatment of MPM elderly patients is reviewed and discussed, with a look to emerging therapies. A structured search of bibliographic databases for peer-reviewed research literature and of main meeting abstracts using a focused review question was undertaken.Expert opinion: Even though the median age of MPM patients enrolled in the most recent experimental trials is increasing, no specific analysis has been reported so far in the elderly. Moreover, no data are available for the 'oldest of the elderly' (>75 years). Treatment of elderly patients with MPM is one of the major challenges to the clinician. There is a clear need of large, well-conducted retrospective studies and above all of prospective investigations in this patient population, both in the first-and in the second-line setting.
Collapse
Affiliation(s)
- Giovanni Luca Ceresoli
- Division of Medical Oncology, Thoracic and Urologic Oncology Unit, Cliniche Humanitas Gavazzeni, Bergamo, Italy
| | - Antonio Rossi
- Division of Medical Oncology, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo (FG), Italy
| |
Collapse
|
5
|
Sato-Kaneko F, Wang X, Yao S, Hosoya T, Lao FS, Messer K, Pu M, Shukla NM, Cottam HB, Chan M, Carson DA, Corr M, Hayashi T. Discovery of a Novel Microtubule Targeting Agent as an Adjuvant for Cancer Immunotherapy. BIOMED RESEARCH INTERNATIONAL 2018; 2018:8091283. [PMID: 30406141 PMCID: PMC6199861 DOI: 10.1155/2018/8091283] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Accepted: 09/17/2018] [Indexed: 12/26/2022]
Abstract
For an activating immunotherapy such as adjuvants, a compound that can prolong immune stimulation may enhance efficacy. We leveraged data from two prior high throughput screens with NF-κB and interferon reporter cell lines to identify 4H-chromene-3-carbonitriles as a class of compounds that prolonged activation in both screens. We repurchased 23 of the most promising candidates. Out of these compounds we found #1 to be the most effective agent in stimulating the release of cytokines and chemokines from immune cells, including murine primary bone marrow derived dendritic cells. Mechanistically, #1 inhibited tubulin polymerization, and its effect on immune cell activation was abolished in cells mutated in the beta-tubulin gene (TUBB) encoding the site where colchicine binds. Treatment with #1 resulted in mitochondrial depolarization followed by mitogen-activated protein kinase activation. Because tubulin polymerization modulating agents have been used for chemotherapy to treat malignancy and #1 activated cytokine responses, we hypothesized that #1 could be effective for cancer immunotherapy. Intratumoral injection of #1 delayed tumor growth in a murine syngeneic model of head and neck cancer. When combined with PD-1 blockade, tumor growth slowed in the injected tumor nodule and there was an abscopal effect in an uninjected nodule on the contralateral flank, suggesting central antitumor immune activation. Thus, we identified a new class of tubulin depolymerizing agent that acts as both an innate and an adaptive immune activating agent and that limits solid tumor growth when used concurrently with a checkpoint inhibitor.
Collapse
Affiliation(s)
- Fumi Sato-Kaneko
- Moores Cancer Center, University of California San Diego, La Jolla 92093, USA
| | - Xiaodong Wang
- Moores Cancer Center, University of California San Diego, La Jolla 92093, USA
- School of Pharmaceutical Sciences, Shenzhen University Health Science Center, Nanhai Ave 3688, Shenzhen, Guangdong 518060, China
| | - Shiyin Yao
- Moores Cancer Center, University of California San Diego, La Jolla 92093, USA
| | - Tadashi Hosoya
- Moores Cancer Center, University of California San Diego, La Jolla 92093, USA
| | - Fitzgerald S. Lao
- Moores Cancer Center, University of California San Diego, La Jolla 92093, USA
| | - Karen Messer
- Division of Biostatistics, University of California San Diego, La Jolla 92093, USA
| | - Minya Pu
- Division of Biostatistics, University of California San Diego, La Jolla 92093, USA
| | - Nikunj M. Shukla
- Moores Cancer Center, University of California San Diego, La Jolla 92093, USA
| | - Howard B. Cottam
- Moores Cancer Center, University of California San Diego, La Jolla 92093, USA
| | - Michael Chan
- Moores Cancer Center, University of California San Diego, La Jolla 92093, USA
| | - Dennis A. Carson
- Moores Cancer Center, University of California San Diego, La Jolla 92093, USA
| | - Maripat Corr
- Department of Medicine, University of California San Diego, La Jolla 92093, USA
| | - Tomoko Hayashi
- Moores Cancer Center, University of California San Diego, La Jolla 92093, USA
| |
Collapse
|
6
|
Microtubule-Actin Crosslinking Factor 1 and Plakins as Therapeutic Drug Targets. Int J Mol Sci 2018; 19:ijms19020368. [PMID: 29373494 PMCID: PMC5855590 DOI: 10.3390/ijms19020368] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Revised: 01/22/2018] [Accepted: 01/23/2018] [Indexed: 12/16/2022] Open
Abstract
Plakins are a family of seven cytoskeletal cross-linker proteins (microtubule-actin crosslinking factor 1 (MACF), bullous pemphigoid antigen (BPAG1) desmoplakin, envoplakin, periplakin, plectin, epiplakin) that network the three major filaments that comprise the cytoskeleton. Plakins have been found to be involved in disorders and diseases of the skin, heart, nervous system, and cancer that are attributed to autoimmune responses and genetic alterations of these macromolecules. Despite their role and involvement across a spectrum of several diseases, there are no current drugs or pharmacological agents that specifically target the members of this protein family. On the contrary, microtubules have traditionally been targeted by microtubule inhibiting agents, used for the treatment of diseases such as cancer, in spite of the deleterious toxicities associated with their clinical utility. The Research Collaboratory for Structural Bioinformatics (RCSB) was used here to identify therapeutic drugs targeting the plakin proteins, particularly the spectraplakins MACF1 and BPAG1, which contain microtubule-binding domains. RCSB analysis revealed that plakin proteins had 329 ligands, of which more than 50% were MACF1 and BPAG1 ligands and 10 were documented, clinically or experimentally, to have several therapeutic applications as anticancer, anti-inflammatory, and antibiotic agents.
Collapse
|
7
|
Petrini I, Lucchesi M, Puppo G, Chella A. Medical treatment of malignant pleural mesothelioma relapses. J Thorac Dis 2018; 10:S333-S341. [PMID: 29507803 PMCID: PMC5830550 DOI: 10.21037/jtd.2017.10.159] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2017] [Accepted: 10/23/2017] [Indexed: 12/29/2022]
Abstract
There are not established treatments for patients with advanced malignant pleural mesothelioma that progressed after first-line chemotherapy with cisplatin and pemetrexed. Retrospective analyses suggest a possible role for rechallenge with pemetrexed for selected patients. Phase II trials demonstrate a modest efficacy of vinorelbine monotherapy with a response rate ranging between 0% and 18% and a tolerable toxicity profile. Combination schedules, despite an increased toxicity, fail to demonstrate an improved efficacy. To date, genome wide analyses did not show molecular targets suitable for therapy and biological drugs did not exert a significant efficacy in clinical trials. Immunotherapy has given a hint of efficacy in early clinical trials but definitive evaluations are still ongoing.
Collapse
Affiliation(s)
- Iacopo Petrini
- Department of Cardiothoracic and Vascular, University Hospital of Pisa, Pisa, Italy
- General Pathology, University of Pisa, Pisa, Italy
| | - Maurizio Lucchesi
- Department of Cardiothoracic and Vascular, University Hospital of Pisa, Pisa, Italy
| | - Gianfranco Puppo
- Department of Cardiothoracic and Vascular, University Hospital of Pisa, Pisa, Italy
| | - Antonio Chella
- Department of Cardiothoracic and Vascular, University Hospital of Pisa, Pisa, Italy
| |
Collapse
|
8
|
Schunselaar LM, Quispel-Janssen JMMF, Kim Y, Alifrangis C, Zwart W, Baas P, Neefjes J. Chemical Profiling of Primary Mesothelioma Cultures Defines Subtypes with Different Expression Profiles and Clinical Responses. Clin Cancer Res 2017; 24:1761-1770. [PMID: 29066506 DOI: 10.1158/1078-0432.ccr-17-1345] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Revised: 08/25/2017] [Accepted: 10/19/2017] [Indexed: 11/16/2022]
Abstract
Purpose: Finding new treatment options for patients with malignant pleural mesothelioma is challenging due to the rarity and heterogeneity of this cancer type. The absence of druggable targets further complicates the development of new therapies. Current treatment options are therefore limited, and prognosis remains poor.Experimental Design: We performed drug screening on primary mesothelioma cultures to guide treatment decisions of corresponding patients that were progressive after first- or second-line treatment.Results: We observed a high concordance between in vitro results and clinical outcomes. We defined three subgroups responding differently to the anticancer drugs tested. In addition, gene expression profiling yielded distinct signatures that segregated the differently responding subgroups. These genes signatures involved various pathways, most prominently the fibroblast growth factor pathway.Conclusions: Our primary mesothelioma culture system has proved to be suitable to test novel drugs. Chemical profiling of primary mesothelioma cultures allows personalizing treatment for a group of patients with a rare tumor type where clinical trials are notoriously difficult. This personalized treatment strategy is expected to improve the poor prospects of patients with mesothelioma. Clin Cancer Res; 24(7); 1761-70. ©2017 AACRSee related commentary by John and Chia, p. 1513.
Collapse
Affiliation(s)
- Laurel M Schunselaar
- Division of Oncogenomics, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | | | - Yongsoo Kim
- Division of Oncogenomics, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | | | - Wilbert Zwart
- Division of Oncogenomics, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Paul Baas
- Department of Thoracic Oncology, Netherlands Cancer Institute, Amsterdam, the Netherlands.
| | - Jacques Neefjes
- Department of Chemical Immunology, Leiden University Medical Center, Leiden, the Netherlands
| |
Collapse
|
9
|
Discovery and Optimization of Novel 5-Indolyl-7-arylimidazo[1,2-a]pyridine-8-carbonitrile Derivatives as Potent Antitubulin Agents Targeting Colchicine-binding Site. Sci Rep 2017; 7:43398. [PMID: 28240326 PMCID: PMC5327470 DOI: 10.1038/srep43398] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Accepted: 01/24/2017] [Indexed: 12/19/2022] Open
Abstract
Aiming at development of potent antitubulin agents targeting colchicine-binding site, a series of novel 5-indolyl-7-arylimidazo[1,2-a]pyridine-8-carbonitrilederivatives (5a–5v and 7a–7h) were designed based on bioisosterism and hybridization strategies. All these compounds were concisely synthesized via a three-step process and examined against five human cancer cell lines (HT-29, A549, MKN-45, MDA-MB-231 and SMMC-7721) along with a normal human cell (L02) in vitro. A structure-activity relationships (SARs) study was carried out and optimization towards this series of compounds in cellular assay resulted in the discovery of 5k, which displayed similar or better antitumor potency against the tested cancer cells with IC50 value ranging from 0.02 to 1.22 μM superior to CA-4 and Crolibulin. Significantly, a cell cycle study disclosed the ability of 5k to arrest cell cycle at the G2/M phase, and immunofluorescence assay as well as a colchicine competition assay revealed that tubulin polymerization was disturbed by 5k by binding to the colchicine site. Moreover, the molecular modeling mode showed the posture of 5k and Crolibulin was similar in the colchcine-binding pocket of tubulin as identified with the SARs and pharmacological results. Together, all these results rationalized 5k might serve as a promising lead for a novel class of antitubulin agents for cancer treatments.
Collapse
|
10
|
Zhong J, Zhu X, Luo K, Li L, Tang M, Liu Y, Zhou Z, Huang Y. Direct Cytoplasmic Delivery and Nuclear Targeting Delivery of HPMA-MT Conjugates in a Microtubules Dependent Fashion. Mol Pharm 2016; 13:3069-79. [PMID: 27417390 DOI: 10.1021/acs.molpharmaceut.6b00181] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
As the hearts of tumor cells, the nucleus is the ultimate target of many chemotherapeutic agents and genes. However, nuclear drug delivery is always hampered by multiple intracellular obstacles, such as low efficiency of lysosome escape and insufficient nuclear trafficking. Herein, an N-(2-hydroxypropyl) methacrylamide (HPMA) polymer-based drug delivery system was designed, which could achieve direct cytoplasmic delivery by a nonendocytic pathway and transport into the nucleus in a microtubules dependent fashion. A special targeting peptide (MT), derived from an endogenic parathyroid hormone-related protein, was conjugated to the polymer backbone, which could accumulate into the nucleus a by microtubule-mediated pathway. The in vitro studies found that low temperature and NaN3 could not influence the cell internalization of the conjugates. Besides, no obvious overlay of the conjugates with lysosome demonstrated that the polymer conjugates could enter the tumor cell cytoplasm by a nonendocytic pathway, thus avoiding the drug degradation in the lysosome. Furthermore, after suppression of the microtubule dynamics with microtubule stabilizing docetaxel (DTX) and destabilizing nocodazole (Noc), the nuclear accumulation of polymeric conjugates was significantly inhibited. Living cells fluorescence recovery after photobleaching study found that the nuclear import rate of conjugates was 2-fold faster compared with the DTX and Noc treated groups. These results demonstrated that the conjugates transported into the nucleus in a microtubules dependent way. Therefore, in addition to direct cytoplasmic delivery, our peptide conjugated polymeric platform could simultaneously mediate nuclear drug accumulation, which may open a new path for further intracellular genes/peptides delivery.
Collapse
Affiliation(s)
- Jiaju Zhong
- Key Laboratory of Drug Targeting and Drug Delivery System (Ministery of Education), West China School of Pharmacy, Sichuan University , NO. 17, Block 3, South Renmin Road, Chengdu 610041, P.R. China
| | - Xi Zhu
- Key Laboratory of Drug Targeting and Drug Delivery System (Ministery of Education), West China School of Pharmacy, Sichuan University , NO. 17, Block 3, South Renmin Road, Chengdu 610041, P.R. China
| | - Kui Luo
- Huaxi MR Research Center (HMRRC), Department of Radiology, West China Hospital, Sichuan University , Chengdu 610041, China
| | - Lian Li
- Key Laboratory of Drug Targeting and Drug Delivery System (Ministery of Education), West China School of Pharmacy, Sichuan University , NO. 17, Block 3, South Renmin Road, Chengdu 610041, P.R. China
| | - Manlin Tang
- Key Laboratory of Drug Targeting and Drug Delivery System (Ministery of Education), West China School of Pharmacy, Sichuan University , NO. 17, Block 3, South Renmin Road, Chengdu 610041, P.R. China
| | - Yanxi Liu
- Key Laboratory of Drug Targeting and Drug Delivery System (Ministery of Education), West China School of Pharmacy, Sichuan University , NO. 17, Block 3, South Renmin Road, Chengdu 610041, P.R. China
| | - Zhou Zhou
- Key Laboratory of Drug Targeting and Drug Delivery System (Ministery of Education), West China School of Pharmacy, Sichuan University , NO. 17, Block 3, South Renmin Road, Chengdu 610041, P.R. China
| | - Yuan Huang
- Key Laboratory of Drug Targeting and Drug Delivery System (Ministery of Education), West China School of Pharmacy, Sichuan University , NO. 17, Block 3, South Renmin Road, Chengdu 610041, P.R. China
| |
Collapse
|
11
|
Spanò V, Pennati M, Parrino B, Carbone A, Montalbano A, Cilibrasi V, Zuco V, Lopergolo A, Cominetti D, Diana P, Cirrincione G, Barraja P, Zaffaroni N. Preclinical Activity of New [1,2]Oxazolo[5,4-e]isoindole Derivatives in Diffuse Malignant Peritoneal Mesothelioma. J Med Chem 2016; 59:7223-38. [PMID: 27428868 DOI: 10.1021/acs.jmedchem.6b00777] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
A series of 22 derivatives of the [1,2]oxazolo[5,4-e]isoindole system were synthesized through an efficient and versatile procedure that involves the annelation of the [1,2]oxazole moiety to the isoindole ring, producing derivatives with a wide substitution pattern. The structure-activity relationship indicates that the N-4-methoxybenzyl group appears crucial for potent activity. In addition, the presence of a 6-phenyl moiety is important and the best activity is reached with a 3,4,5-trimethoxy substituent. The most active compound, bearing both the structural features, was able to inhibit tumor cell proliferation at nanomolar concentrations when tested against the full NCI human tumor cell line panel. Interestingly, this compound was effective in reducing in vitro and in vivo cell growth, impairing cell cycle progression and inducing apoptosis, as a consequence of the inhibition of tubulin polymerization, in experimental models of diffuse malignant peritoneal mesothelioma (DMPM), a rapidly lethal disease, poorly responsive to conventional therapeutic strategies.
Collapse
Affiliation(s)
- Virginia Spanò
- Dipartimento di Scienze e Tecnologie Biologiche Chimiche e Farmaceutiche (STEBICEF), Università degli Studi di Palermo , Via Archirafi 32, 90123 Palermo, Italy
| | - Marzia Pennati
- Molecular Pharmacology Unit, Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale Tumori , Via Amadeo 42, 20133 Milano, Italy
| | - Barbara Parrino
- Dipartimento di Scienze e Tecnologie Biologiche Chimiche e Farmaceutiche (STEBICEF), Università degli Studi di Palermo , Via Archirafi 32, 90123 Palermo, Italy
| | - Anna Carbone
- Dipartimento di Scienze e Tecnologie Biologiche Chimiche e Farmaceutiche (STEBICEF), Università degli Studi di Palermo , Via Archirafi 32, 90123 Palermo, Italy
| | - Alessandra Montalbano
- Dipartimento di Scienze e Tecnologie Biologiche Chimiche e Farmaceutiche (STEBICEF), Università degli Studi di Palermo , Via Archirafi 32, 90123 Palermo, Italy
| | - Vincenzo Cilibrasi
- Dipartimento di Scienze e Tecnologie Biologiche Chimiche e Farmaceutiche (STEBICEF), Università degli Studi di Palermo , Via Archirafi 32, 90123 Palermo, Italy
| | - Valentina Zuco
- Molecular Pharmacology Unit, Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale Tumori , Via Amadeo 42, 20133 Milano, Italy
| | - Alessia Lopergolo
- Molecular Pharmacology Unit, Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale Tumori , Via Amadeo 42, 20133 Milano, Italy
| | - Denis Cominetti
- Molecular Pharmacology Unit, Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale Tumori , Via Amadeo 42, 20133 Milano, Italy
| | - Patrizia Diana
- Dipartimento di Scienze e Tecnologie Biologiche Chimiche e Farmaceutiche (STEBICEF), Università degli Studi di Palermo , Via Archirafi 32, 90123 Palermo, Italy
| | - Girolamo Cirrincione
- Dipartimento di Scienze e Tecnologie Biologiche Chimiche e Farmaceutiche (STEBICEF), Università degli Studi di Palermo , Via Archirafi 32, 90123 Palermo, Italy
| | - Paola Barraja
- Dipartimento di Scienze e Tecnologie Biologiche Chimiche e Farmaceutiche (STEBICEF), Università degli Studi di Palermo , Via Archirafi 32, 90123 Palermo, Italy
| | - Nadia Zaffaroni
- Molecular Pharmacology Unit, Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale Tumori , Via Amadeo 42, 20133 Milano, Italy
| |
Collapse
|