1
|
Wang Z, Li W, Jiang Y, Tran TB, Cordova LE, Chung J, Kim M, Wondrak G, Erdrich J, Lu J. Sphingomyelin-derived nanovesicles for the delivery of the IDO1 inhibitor epacadostat enhance metastatic and post-surgical melanoma immunotherapy. Nat Commun 2023; 14:7235. [PMID: 37945606 PMCID: PMC10636136 DOI: 10.1038/s41467-023-43079-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Accepted: 10/31/2023] [Indexed: 11/12/2023] Open
Abstract
Epacadostat (EPA), the most advanced IDO1 inhibitor, in combination with PD-1 checkpoint inhibitor, has failed in a recent Phase III clinical trial for treating metastatic melanoma. Here we report an EPA nanovesicle therapeutic platform (Epacasome) based on chemically attaching EPA to sphingomyelin via an oxime-ester bond highly responsive to hydrolase cleavage. Via clathrin-mediated endocytosis, Epacasome displays higher cellular uptake and enhances IDO1 inhibition and T cell proliferation compared to free EPA. Epacasome shows improved pharmacokinetics and tumour accumulation with efficient intratumoural drug release and deep tumour penetration. Additionally, it outperforms free EPA for anticancer efficacy, potentiating PD-1 blockade with boosted cytotoxic T lymphocytes (CTLs) and reduced regulatory T cells and myeloid-derived suppressor cells responses in a B16-F10 melanoma model in female mice. By co-encapsulating immunogenic dacarbazine, Epacasome further enhances anti-tumor effects and immune responses through the upregulation of NKG2D-mediated CTLs and natural killer cells responses particularly when combined with the PD-1 inhibitor in the late-stage metastatic B16-F10-Luc2 model in female mice. Furthermore, this combination prevents tumour recurrence and prolongs mouse survival in a clinically relevant, post-surgical melanoma model in female mice. Epacasome demonstrates potential to synergize with PD-1 blockade for improved response to melanoma immunotherapy.
Collapse
Affiliation(s)
- Zhiren Wang
- Skaggs Pharmaceutical Sciences Center, Department of Pharmacology & Toxicology, R. Ken Coit College of Pharmacy, The University of Arizona, Tucson, AZ, 85721, USA
| | - Wenpan Li
- Skaggs Pharmaceutical Sciences Center, Department of Pharmacology & Toxicology, R. Ken Coit College of Pharmacy, The University of Arizona, Tucson, AZ, 85721, USA
| | - Yanhao Jiang
- Skaggs Pharmaceutical Sciences Center, Department of Pharmacology & Toxicology, R. Ken Coit College of Pharmacy, The University of Arizona, Tucson, AZ, 85721, USA
| | - Tuyen Ba Tran
- Skaggs Pharmaceutical Sciences Center, Department of Pharmacology & Toxicology, R. Ken Coit College of Pharmacy, The University of Arizona, Tucson, AZ, 85721, USA
| | - Leyla Estrella Cordova
- Skaggs Pharmaceutical Sciences Center, Department of Pharmacology & Toxicology, R. Ken Coit College of Pharmacy, The University of Arizona, Tucson, AZ, 85721, USA
| | - Jinha Chung
- Skaggs Pharmaceutical Sciences Center, Department of Pharmacology & Toxicology, R. Ken Coit College of Pharmacy, The University of Arizona, Tucson, AZ, 85721, USA
| | - Minhyeok Kim
- Skaggs Pharmaceutical Sciences Center, Department of Pharmacology & Toxicology, R. Ken Coit College of Pharmacy, The University of Arizona, Tucson, AZ, 85721, USA
| | - Georg Wondrak
- Skaggs Pharmaceutical Sciences Center, Department of Pharmacology & Toxicology, R. Ken Coit College of Pharmacy, The University of Arizona, Tucson, AZ, 85721, USA
- NCI-designated University of Arizona Comprehensive Cancer Center, Tucson, AZ, 85721, USA
| | - Jennifer Erdrich
- Department of Surgery, Division of Surgical Oncology, The University of Arizona College of Medicine, Tucson, AZ, 85721, USA
| | - Jianqin Lu
- Skaggs Pharmaceutical Sciences Center, Department of Pharmacology & Toxicology, R. Ken Coit College of Pharmacy, The University of Arizona, Tucson, AZ, 85721, USA.
- NCI-designated University of Arizona Comprehensive Cancer Center, Tucson, AZ, 85721, USA.
- BIO5 Institute, The University of Arizona, Tucson, AZ, 85721, USA.
- Southwest Environmental Health Sciences Center, The University of Arizona, Tucson, AZ, 85721, USA.
| |
Collapse
|
2
|
Moschella F, Buccione C, Ruspantini I, Castiello L, Rozo Gonzalez A, Iacobone F, Ferraresi V, Palermo B, Nisticò P, Belardelli F, Proietti E, Macchia I, Urbani F. Blood immune cells as potential biomarkers predicting relapse-free survival of stage III/IV resected melanoma patients treated with peptide-based vaccination and interferon-alpha. Front Oncol 2023; 13:1145667. [PMID: 37274275 PMCID: PMC10233106 DOI: 10.3389/fonc.2023.1145667] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Accepted: 04/24/2023] [Indexed: 06/06/2023] Open
Abstract
Introduction Despite the recent approval of several therapies in the adjuvant setting of melanoma, tumor relapse still occurs in a significant number of completely resected stage III-IV patients. In this context, the use of cancer vaccines is still relevant and may increase the response to immune checkpoint inhibitors. We previously demonstrated safety, immunogenicity and preliminary evidence of clinical efficacy in stage III/IV resected melanoma patients subjected to a combination therapy based on peptide vaccination together with intermittent low-dose interferon-α2b, with or without dacarbazine preconditioning (https://www.clinicaltrialsregister.eu/ctr-search/search, identifier: 2008-008211-26). In this setting, we then focused on pre-treatment patient immune status to highlight possible factors associated with clinical outcome. Methods Multiparametric flow cytometry was used to identify baseline immune profiles in patients' peripheral blood mononuclear cells and correlation with the patient clinical outcome. Receiver operating characteristic curve, Kaplan-Meier survival and principal component analyses were used to evaluate the predictive power of the identified markers. Results We identified 12 different circulating T and NK cell subsets with significant (p ≤ 0.05) differential baseline levels in patients who later relapsed with respect to patients who remained free of disease. All 12 parameters showed a good prognostic accuracy (AUC>0.7, p ≤ 0.05) and 11 of them significantly predicted the relapse-free survival. Remarkably, 3 classifiers also predicted the overall survival. Focusing on immune cell subsets that can be analyzed through simple surface staining, three subsets were identified, namely regulatory T cells, CD56dimCD16- NK cells and central memory γδ T cells. Each subset showed an AUC>0.8 and principal component analysis significantly grouped relapsing and non-relapsing patients (p=0.034). These three subsets were used to calculate a combination score that was able to perfectly distinguish relapsing and non-relapsing patients (AUC=1; p=0). Noticeably, patients with a combined score ≥2 demonstrated a strong advantage in both relapse-free (p=0.002) and overall (p=0.011) survival as compared to patients with a score <2. Discussion Predictive markers may be used to guide patient selection for personalized therapies and/or improve follow-up strategies. This study provides preliminary evidence on the identification of peripheral blood immune biomarkers potentially capable of predicting the clinical response to combined vaccine-based adjuvant therapies in melanoma.
Collapse
Affiliation(s)
- Federica Moschella
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Carla Buccione
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | | | | | - Andrea Rozo Gonzalez
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Floriana Iacobone
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Virginia Ferraresi
- Department of Medical Oncology 1, Scientific Institute for Research, Hospitalization and Healthcare (IRCCS) Regina Elena National Cancer Institute, Rome, Italy
| | - Belinda Palermo
- Tumor Immunology and Immunotherapy Unit, Department of Research, Advanced Diagnostics and Technological Innovation, Scientific Institute for Research, Hospitalization and Healthcare (IRCCS) Regina Elena National Cancer Institute, Rome, Italy
| | - Paola Nisticò
- Tumor Immunology and Immunotherapy Unit, Department of Research, Advanced Diagnostics and Technological Innovation, Scientific Institute for Research, Hospitalization and Healthcare (IRCCS) Regina Elena National Cancer Institute, Rome, Italy
| | - Filippo Belardelli
- Institute of Translational Pharmacology, National Research Council (CNR), Rome, Italy
| | - Enrico Proietti
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Iole Macchia
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Francesca Urbani
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
- Medical Biotechnology and Translational Medicine PhD School, II University of Rome “Tor Vergata”, Rome, Italy
| |
Collapse
|
3
|
Barnestein R, Galland L, Kalfeist L, Ghiringhelli F, Ladoire S, Limagne E. Immunosuppressive tumor microenvironment modulation by chemotherapies and targeted therapies to enhance immunotherapy effectiveness. Oncoimmunology 2022; 11:2120676. [PMID: 36117524 PMCID: PMC9481153 DOI: 10.1080/2162402x.2022.2120676] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022] Open
Abstract
With the rapid clinical development of immune checkpoint inhibitors (ICIs), the standard of care in cancer management has evolved rapidly. However, immunotherapy is not currently beneficial for all patients. In addition to intrinsic tumor factors, other etiologies of resistance to ICIs arise from the complex interplay between cancer and its microenvironment. Recognition of the essential role of the tumor microenvironment (TME) in cancer progression has led to a shift from a tumor-cell-centered view of cancer development, to the concept of a complex tumor ecosystem that supports tumor growth and metastatic dissemination. The expansion of immunosuppressive cells represents a cardinal strategy deployed by tumor cells to escape detection and elimination by the immune system. Regulatory T lymphocytes (Treg), myeloid-derived suppressor cells (MDSCs), and type-2 tumor-associated macrophages (TAM2) are major components of these inhibitory cellular networks, with the ability to suppress innate and adaptive anticancer immunity. They therefore represent major impediments to anticancer therapies, particularly immune-based interventions. Recent work has provided evidence that, beyond their direct cytotoxic effects on cancer cells, several conventional chemotherapeutic (CT) drugs and agents used in targeted therapies (TT) can promote the elimination or inactivation of suppressive immune cells, resulting in enhanced antitumor immunity. In this review, we will analyze findings pertaining to this concept, discuss the possible molecular bases underlying the selective targeting of these immunosuppressive cells by antineoplastic agents (CT and/or TT), and consider current challenges and future prospects related to the integration of these molecules into more efficient anticancer strategies, in the era of immunotherapy.
Collapse
Affiliation(s)
- Robby Barnestein
- University of Burgundy, Dijon, France
- Platform of Transfer in Cancer Biology, Georges François Leclerc Cancer Center, Dijon, France
| | - Loïck Galland
- University of Burgundy, Dijon, France
- Platform of Transfer in Cancer Biology, Georges François Leclerc Cancer Center, Dijon, France
- Department of Medical Oncology, Georges François Leclerc Center, Dijon, France
| | - Laura Kalfeist
- University of Burgundy, Dijon, France
- Platform of Transfer in Cancer Biology, Georges François Leclerc Cancer Center, Dijon, France
- Centre de Recherche INSERM LNC-UMR1231, Dijon, France
- Genomic and Immunotherapy Medical Institute, Dijon University Hospital, Dijon, France
| | - François Ghiringhelli
- University of Burgundy, Dijon, France
- Platform of Transfer in Cancer Biology, Georges François Leclerc Cancer Center, Dijon, France
- Department of Medical Oncology, Georges François Leclerc Center, Dijon, France
- Centre de Recherche INSERM LNC-UMR1231, Dijon, France
- Genomic and Immunotherapy Medical Institute, Dijon University Hospital, Dijon, France
| | - Sylvain Ladoire
- University of Burgundy, Dijon, France
- Platform of Transfer in Cancer Biology, Georges François Leclerc Cancer Center, Dijon, France
- Department of Medical Oncology, Georges François Leclerc Center, Dijon, France
- Centre de Recherche INSERM LNC-UMR1231, Dijon, France
- Genomic and Immunotherapy Medical Institute, Dijon University Hospital, Dijon, France
| | - Emeric Limagne
- University of Burgundy, Dijon, France
- Platform of Transfer in Cancer Biology, Georges François Leclerc Cancer Center, Dijon, France
- Department of Medical Oncology, Georges François Leclerc Center, Dijon, France
- Centre de Recherche INSERM LNC-UMR1231, Dijon, France
- Genomic and Immunotherapy Medical Institute, Dijon University Hospital, Dijon, France
| |
Collapse
|
4
|
Chemotherapie nach Progress unter Immuncheckpoint-Inhibition beim metastasierten malignen Melanom: Fallbericht und Literaturübersicht. AKTUELLE DERMATOLOGIE 2022. [DOI: 10.1055/a-1874-6051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Zusammenfassung
Hintergrund Immuncheckpoint-Inhibitoren (ICI), BRAF- und MEK-Inhibitoren haben die Prognose von Patienten mit metastasiertem malignem Melanom entscheidend verbessert. Nichtsdestotrotz spricht ein erheblicher Prozentsatz nicht oder nur vorübergehend auf diese modernen Therapien an. Insbesondere bei Patienten mit BRAF-Wildtyp-Melanom sind die Therapieoptionen nach Versagen von ICI limitiert.
Kasuistik Eine 24-jährige Patientin stellte sich im Dezember 2015 mit einem akrolentiginösen malignen Melanom am linken Fußballen im Tumorstadium IIB (pT4a, pN0, cM0, Infiltrationstiefe 7,95 mm, BRAF-V600-Wildtyp) vor, das leitliniengerecht operativ versorgt und für 18 Monate adjuvant mit Interferon-alpha behandelt wurde. Nach Diagnose von zwei Lymphknotenmetastasen links inguinal und radikaler Lymphknotendissektion erhielt die Patientin für 12 Monate eine adjuvante Therapie mit Nivolumab. Nach einem 10-monatigen tumorfreien Intervall ohne Therapie erbrachte das Staging eine paraaortale Lymphknotenmetastase. Gemäß Tumorkonferenz-Beschluss wurde bei nunmehr vorliegendem Stadium IV eine Immunkombinationstherapie mit Nivolumab und Ipilimumab begonnen, die nach 2 Gaben aufgrund einer Autoimmunkolitis und -hepatitis Grad 3 beendet und auf eine Nivolumab-Monotherapie umgestellt wurde. Bei erneutem Tumorprogress auch nach Metastasenresektion und fehlender Möglichkeit einer Studienteilnahme wurde eine palliative Chemotherapie mit Dacarbazin 850 mg/m2 alle 3 Wochen eingeleitet. Hiermit ließ sich nach 4 Zyklen eine partielle Remission und nach 10 Zyklen eine Komplettremission erzielen, die nunmehr seit 10 Monaten ohne Therapie anhält.
Diskussion Es gibt Hinweise darauf, dass die Effektivität einer Chemotherapie nach vorangegangener Immuncheckpoint-Inhibition höher ist als ohne diese Vortherapie, möglicherweise durch einen immunmodulatorischen Effekt der Chemotherapie, der synergistisch mit einer Immunaktivierung durch ICI wirkt. Bisher liegen nur wenige Studien zu dieser Therapiesequenz beim Melanom vor, bei denen es sich ausschließlich um retrospektive Analysen handelt. Diese weisen auf einen klinischen Nutzen der Therapiesequenz hin, wenngleich die Evidenz hierfür sehr gering ist. Die meisten Patienten, die nach einer Vortherapie mit ICI auf Dacarbazin oder andere Chemotherapien ansprechen, erreichen eine partielle Remission. Anhaltende Komplettremissionen wie bei unserer Patientin sind eine Rarität.
Collapse
|
5
|
Jones AB, Rocco A, Lamb LS, Friedman GK, Hjelmeland AB. Regulation of NKG2D Stress Ligands and Its Relevance in Cancer Progression. Cancers (Basel) 2022; 14:2339. [PMID: 35565467 PMCID: PMC9105350 DOI: 10.3390/cancers14092339] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 04/22/2022] [Accepted: 04/26/2022] [Indexed: 01/27/2023] Open
Abstract
Under cellular distress, multiple facets of normal homeostatic signaling are altered or disrupted. In the context of the immune landscape, external and internal stressors normally promote the expression of natural killer group 2 member D (NKG2D) ligands that allow for the targeted recognition and killing of cells by NKG2D receptor-bearing effector populations. The presence or absence of NKG2D ligands can heavily influence disease progression and impact the accessibility of immunotherapy options. In cancer, tumor cells are known to have distinct regulatory mechanisms for NKG2D ligands that are directly associated with tumor progression and maintenance. Therefore, understanding the regulation of NKG2D ligands in cancer will allow for targeted therapeutic endeavors aimed at exploiting the stress response pathway. In this review, we summarize the current understanding of regulatory mechanisms controlling the induction and repression of NKG2D ligands in cancer. Additionally, we highlight current therapeutic endeavors targeting NKG2D ligand expression and offer our perspective on considerations to further enhance the field of NKG2D ligand biology.
Collapse
Affiliation(s)
- Amber B. Jones
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL 35233, USA;
| | - Abbey Rocco
- Department of Pediatrics, Division of Pediatric Hematology and Oncology, University of Alabama at Birmingham, Birmingham, AL 35233, USA; (A.R.); (G.K.F.)
| | | | - Gregory K. Friedman
- Department of Pediatrics, Division of Pediatric Hematology and Oncology, University of Alabama at Birmingham, Birmingham, AL 35233, USA; (A.R.); (G.K.F.)
| | - Anita B. Hjelmeland
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL 35233, USA;
| |
Collapse
|
6
|
Kordbacheh T, Honeychurch J, Blackhall F, Faivre-Finn C, Illidge T. Radiotherapy and anti-PD-1/PD-L1 combinations in lung cancer: building better translational research platforms. Ann Oncol 2018; 29:301-310. [PMID: 29309540 DOI: 10.1093/annonc/mdx790] [Citation(s) in RCA: 98] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/11/2024] Open
Abstract
Despite the unheralded success of immune checkpoint blockade in delivering durable responses for some patients with non-small-cell lung cancer (NSCLC), the majority of patients do not respond. PD-L1 tumour expression and pre-existing tumour T-cell infiltration have been correlated with improved clinical outcomes to anti-PD-1/anti-PD-L1. However, patients with tumours that are negative for PD-L1 expression can also respond to treatment. Strategies to combine other treatment modalities like radiotherapy (RT) with immune checkpoint inhibitors are being investigated as means of improving the response rates to PD-1/PD-L1 antibody blockade. RT induces immunogenic changes in cancer cells, can adaptively upregulate tumour cell PD-L1 expression and can improve the efficacy of anti-PD-1/anti-PD-L1 therapy. How we design future clinical trials in NSCLC also depends on practical considerations of delivering these treatment combinations, such as RT dose, fractionation and field volume, as well as scheduling with immune checkpoint blockade. Here, we review reasons for resistance to anti-PD-1/anti-PD-L1 and how RT may be utilised in combination with these drugs to enhance their effect by building better translational research platforms.
Collapse
Affiliation(s)
- T Kordbacheh
- Targeted Therapy Group, Division of Cancer Sciences, University of Manchester, UK; Cancer Research UK Lung Cancer Centre of Excellence, Manchester Cancer Research Centre, Manchester, M20 4BX, UK; The Christie NHS Foundation Trust, Manchester University Hospitals NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, M20 4BX, UK.
| | - J Honeychurch
- Targeted Therapy Group, Division of Cancer Sciences, University of Manchester, UK; Cancer Research UK Lung Cancer Centre of Excellence, Manchester Cancer Research Centre, Manchester, M20 4BX, UK
| | - F Blackhall
- Targeted Therapy Group, Division of Cancer Sciences, University of Manchester, UK; Cancer Research UK Lung Cancer Centre of Excellence, Manchester Cancer Research Centre, Manchester, M20 4BX, UK; The Christie NHS Foundation Trust, Manchester University Hospitals NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, M20 4BX, UK
| | - C Faivre-Finn
- Targeted Therapy Group, Division of Cancer Sciences, University of Manchester, UK; Cancer Research UK Lung Cancer Centre of Excellence, Manchester Cancer Research Centre, Manchester, M20 4BX, UK; The Christie NHS Foundation Trust, Manchester University Hospitals NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, M20 4BX, UK
| | - T Illidge
- Targeted Therapy Group, Division of Cancer Sciences, University of Manchester, UK; Cancer Research UK Lung Cancer Centre of Excellence, Manchester Cancer Research Centre, Manchester, M20 4BX, UK; The Christie NHS Foundation Trust, Manchester University Hospitals NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, M20 4BX, UK
| |
Collapse
|
7
|
Ciprofloxacin-mediated induction of S-phase cell cycle arrest and apoptosis in COLO829 melanoma cells. Pharmacol Rep 2017; 70:6-13. [PMID: 29306115 DOI: 10.1016/j.pharep.2017.07.007] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2017] [Revised: 07/03/2017] [Accepted: 07/12/2017] [Indexed: 02/03/2023]
Abstract
BACKGROUND Low effectiveness of anti-melanoma therapies makes it necessary to search for new drugs that could improve or replace the standard chemotherapy. Fluoroquinolones are a group of synthetic antibiotics, used in the treatment of wide range of bacterial infections. Moreover, this class of antibiotics has shown promising anti-tumor activity in several cancer cell lines. The aim of this study was to examine the effect of ciprofloxacin on cell viability, apoptosis and cell cycle distribution in COLO829 melanoma cells. METHODS Cell viability was evaluated by the WST-1 assay. Cell cycle distribution and apoptosis in cells exposed to ciprofloxacin was analyzed by the use of fluorescence image cytometer NucleoCounter NC-3000. RESULTS Ciprofloxacin decreased the cell viability in a dose- and time-dependent manner. For COLO829 cells treated with ciprofloxacin for 24 h, 48 h and 72 h the values of IC50 were found to be 0.74 mM, 0.17 mM and 0.10 mM, respectively. The oligonucleosomal DNA fragmentation was observed when the cells were exposed to ciprofloxacin in concentration of 1.0 mM for 48 h and 72 h. At lower ciprofloxacin concentrations (0.01 mM and 0.1 mM) cells were arrested in S-phase suggesting a mechanism related to topoisomerase II inhibition. Moreover, it was demonstrated that ciprofloxacin induced apoptosis as a result of mitochondrial membrane breakdown. CONCLUSIONS The obtained results for COLO829 melanoma cells were compared with data for normal dark pigmented melanocytes and the use of ciprofloxacin as a potential anticancer drug for the treatment of melanoma in vivo was considered.
Collapse
|
8
|
Sottile R, Pangigadde PN, Tan T, Anichini A, Sabbatino F, Trecroci F, Favoino E, Orgiano L, Roberts J, Ferrone S, Kärre K, Colucci F, Carbone E. HLA class I downregulation is associated with enhanced NK-cell killing of melanoma cells with acquired drug resistance to BRAF inhibitors. Eur J Immunol 2015; 46:409-19. [PMID: 26564811 PMCID: PMC4832274 DOI: 10.1002/eji.201445289] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2014] [Revised: 09/22/2015] [Accepted: 11/05/2015] [Indexed: 01/06/2023]
Abstract
The frequent development of drug resistance to targeted therapies in cancer patients has stimulated interest in strategies counteracting resistance. Combining immunotherapies with targeted therapies is one such strategy. In this context, we asked whether human NK cells can target melanoma cells that have acquired resistance to selective inhibitors targeting activating mutants of the B‐Raf kinase (BRAF inhibitors, BRAFi). We generated drug‐resistant cell variants in vitro from human BRAF‐mutant melanoma cell lines MEL‐HO, COLO‐38, SK‐MEL‐37, 1520 and from primary melanoma cells freshly isolated from two patients. All drug‐resistant cell variants remained susceptible to lysis by IL‐2‐activated NK cells; and two BRAFi‐resistant lines (BRAFi‐R) became significantly more susceptible to NK‐cell lysis than their parental lines. This was associated with significant HLA class I antigen downregulation and PD‐L1 upregulation on the drug‐resistant lines. Although blocking HLA class I enhanced the extent of lysis of both BRAFi‐R and parental cells to NK‐cell‐mediated lysis, antibody‐mediated inhibition of PD1–PD‐L1 interactions had no detectable effect. HLA class I antigen expression on BRAFi‐R melanoma variants thus appears to play a major role in their susceptibility to NK‐cell cytotoxicity. These findings suggest that NK‐cell‐based immunotherapy may be a viable approach to treat melanoma patients with acquired resistance to BRAF inhibitors.
Collapse
Affiliation(s)
- Rosa Sottile
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institute, Stockholm, Sweden.,University Magna Graecia of Catanzaro, Catanzaro, Italy
| | - Pradeepa N Pangigadde
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institute, Stockholm, Sweden.,University Magna Graecia of Catanzaro, Catanzaro, Italy
| | - Thomas Tan
- Department of Obstetrics and Gynaecology, University of Cambridge School of Clinical Medicine, NIHR Cambridge Biomedical Research Centre, Cambridge, UK
| | - Andrea Anichini
- Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | | | | | - Elvira Favoino
- Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Laura Orgiano
- Department of Obstetrics and Gynaecology, University of Cambridge School of Clinical Medicine, NIHR Cambridge Biomedical Research Centre, Cambridge, UK
| | - James Roberts
- Department of Obstetrics and Gynaecology, University of Cambridge School of Clinical Medicine, NIHR Cambridge Biomedical Research Centre, Cambridge, UK
| | - Soldano Ferrone
- Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Klas Kärre
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institute, Stockholm, Sweden
| | - Francesco Colucci
- Department of Obstetrics and Gynaecology, University of Cambridge School of Clinical Medicine, NIHR Cambridge Biomedical Research Centre, Cambridge, UK
| | - Ennio Carbone
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institute, Stockholm, Sweden.,University Magna Graecia of Catanzaro, Catanzaro, Italy
| |
Collapse
|
9
|
Metastatic melanoma treatment: Combining old and new therapies. Crit Rev Oncol Hematol 2015; 98:242-53. [PMID: 26616525 DOI: 10.1016/j.critrevonc.2015.11.011] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2014] [Revised: 10/16/2015] [Accepted: 11/12/2015] [Indexed: 01/04/2023] Open
Abstract
Metastatic melanoma is an aggressive form of cancer characterised by poor prognosis and a complex etiology. Until 2010, the treatment options for metastatic melanoma were very limited. Largely ineffective dacarbazine, temozolamide or fotemustine were the only agents in use for 35 years. In recent years, the development of molecularly targeted inhibitors in parallel with the development of checkpoint inhibition immunotherapies has rapidly improved the outcomes for metastatic melanoma patients. Despite these new therapies showing initial promise; resistance and poor duration of response have limited their effectiveness as monotherapies. Here we provide an overview of the history of melanoma treatment, as well as the current treatments in development. We also discuss the future of melanoma treatment as we go beyond monotherapies to a combinatorial approach. Combining older therapies with the new molecular and immunotherapies will be the most promising way forward for treatment of metastatic melanoma.
Collapse
|
10
|
The Effect of Clinically Therapeutic Plasma Concentrations of Lidocaine on Natural Killer Cell Cytotoxicity. Reg Anesth Pain Med 2015; 40:43-8. [DOI: 10.1097/aap.0000000000000191] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
11
|
Zegers CML, Rekers NH, Quaden DHF, Lieuwes NG, Yaromina A, Germeraad WTV, Wieten L, Biessen EAL, Boon L, Neri D, Troost EGC, Dubois LJ, Lambin P. Radiotherapy combined with the immunocytokine L19-IL2 provides long-lasting antitumor effects. Clin Cancer Res 2014; 21:1151-60. [PMID: 25552483 DOI: 10.1158/1078-0432.ccr-14-2676] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Radiotherapy modifies the tumor microenvironment and causes the release of tumor antigens, which can enhance the effect of immunotherapy. L19 targets the extra domain B (ED-B) of fibronectin, a marker for tumor neoangiogenesis, and can be used as immunocytokine when coupled to IL2. We hypothesize that radiotherapy in combination with L19-IL2 provides an enhanced antitumor effect, which is dependent on ED-B expression. EXPERIMENTAL DESIGN Mice were injected with syngeneic C51 colon carcinoma, Lewis lung carcinoma (LLC), or 4T1 mammary carcinoma cells. Tumor growth delay, underlying immunologic parameters, and treatment toxicity were evaluated after single-dose local tumor irradiation and systemic administration of L19-IL2 or equimolar controls. RESULTS ED-B expression was high, intermediate, and low for C51, LLC, and 4T1, respectively. The combination therapy showed (i) a long-lasting synergistic effect for the C51 model with 75% of tumors being cured, (ii) an additive effect for the LLC model, and (iii) no effect for the 4T1 model. The combination treatment resulted in a significantly increased cytotoxic (CD8(+)) T-cell population for both C51 and LLC. Depletion of CD8(+) T cells abolished the benefit of the combination therapy. CONCLUSIONS These data provide the first evidence for an increased therapeutic potential by combining radiotherapy with L19-IL2 in ED-B-positive tumors. This new opportunity in cancer treatment will be investigated in a phase I clinical study for patients with an oligometastatic solid tumor (NCT02086721). An animation summarizing our results is available at https://www.youtube.com/watch?v=xHbwQuCTkRc.
Collapse
Affiliation(s)
- Catharina M L Zegers
- Department of Radiation Oncology (MAASTRO), Maastricht University Medical Centre, Maastricht, the Netherlands.
| | - Nicolle H Rekers
- Department of Radiation Oncology (MAASTRO), Maastricht University Medical Centre, Maastricht, the Netherlands
| | - Dana H F Quaden
- Department of Radiation Oncology (MAASTRO), Maastricht University Medical Centre, Maastricht, the Netherlands. Department of Internal Medicine, Maastricht University Medical Centre, Maastricht, the Netherlands
| | - Natasja G Lieuwes
- Department of Radiation Oncology (MAASTRO), Maastricht University Medical Centre, Maastricht, the Netherlands
| | - Ala Yaromina
- Department of Radiation Oncology (MAASTRO), Maastricht University Medical Centre, Maastricht, the Netherlands
| | - Wilfred T V Germeraad
- Department of Internal Medicine, Maastricht University Medical Centre, Maastricht, the Netherlands
| | - Lotte Wieten
- Department of Transplantation Immunology, Maastricht University Medical Centre, Maastricht, the Netherlands
| | - Erik A L Biessen
- Experimental Vascular Pathology Group, Cardiovascular Research Institute Maastricht, GROW - School for Oncology and Developmental Biology, Maastricht University Medical Centre, Maastricht, the Netherlands
| | | | - Dario Neri
- Department of Chemistry and Applied Biosciences, Swiss Federal Institute of Technology (ETH Zürich), Zürich, Switzerland
| | - Esther G C Troost
- Department of Radiation Oncology (MAASTRO), Maastricht University Medical Centre, Maastricht, the Netherlands
| | - Ludwig J Dubois
- Department of Radiation Oncology (MAASTRO), Maastricht University Medical Centre, Maastricht, the Netherlands
| | - Philippe Lambin
- Department of Radiation Oncology (MAASTRO), Maastricht University Medical Centre, Maastricht, the Netherlands
| |
Collapse
|
12
|
Mignot G, Hervieu A, Vabres P, Dalac S, Jeudy G, Bel B, Apetoh L, Ghiringhelli F. Prospective study of the evolution of blood lymphoid immune parameters during dacarbazine chemotherapy in metastatic and locally advanced melanoma patients. PLoS One 2014; 9:e105907. [PMID: 25170840 PMCID: PMC4149472 DOI: 10.1371/journal.pone.0105907] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2014] [Accepted: 07/29/2014] [Indexed: 12/22/2022] Open
Abstract
Background The importance of immune responses in the control of melanoma growth is well known. However, the implication of these antitumor immune responses in the efficacy of dacarbazine, a cytotoxic drug classically used in the treatment of melanoma, remains poorly understood in humans. Methods In this prospective observational study, we performed an immunomonitoring of eleven metastatic or locally advanced patients treated with dacarbazine as a first line of treatment. We assessed by flow cytometry lymphoid populations and their activation state; we also isolated NK cells to perform in vitro cytotoxicity tests. Results We found that chemotherapy induces lymphopenia and that a significantly higher numbers of naïve CD4+ T cells and lower proportion of Treg before chemotherapy are associated with disease control after dacarbazine treatment. Interestingly, NK cell cytotoxicity against dacarbazine-pretreated melanoma cells is only observed in NK cells from patients who achieved disease control. Conclusion Together, our data pinpoint that some immune factors could help to predict the response of melanoma patients to dacarbazine. Future larger scale studies are warranted to test their validity as prediction markers.
Collapse
Affiliation(s)
- Grégoire Mignot
- INSERM, UMR866, Dijon, France
- Faculté de Médecine, Université de Bourgogne, Dijon, France
| | - Alice Hervieu
- INSERM, UMR866, Dijon, France
- Faculté de Médecine, Université de Bourgogne, Dijon, France
- Service de Dermatologie, Centre Hospitalier Universitaire le Bocage, Dijon, France
| | - Pierre Vabres
- Faculté de Médecine, Université de Bourgogne, Dijon, France
- Service de Dermatologie, Centre Hospitalier Universitaire le Bocage, Dijon, France
| | - Sophie Dalac
- Service de Dermatologie, Centre Hospitalier Universitaire le Bocage, Dijon, France
| | - Geraldine Jeudy
- Service de Dermatologie, Centre Hospitalier Universitaire le Bocage, Dijon, France
| | - Blandine Bel
- Service de Dermatologie, Centre Hospitalier Universitaire le Bocage, Dijon, France
| | - Lionel Apetoh
- INSERM, UMR866, Dijon, France
- Faculté de Médecine, Université de Bourgogne, Dijon, France
- Oncologie médicale, Centre Georges François Leclerc, Dijon, France
| | - François Ghiringhelli
- INSERM, UMR866, Dijon, France
- Faculté de Médecine, Université de Bourgogne, Dijon, France
- Oncologie médicale, Centre Georges François Leclerc, Dijon, France
- * E-mail:
| |
Collapse
|
13
|
Sukkurwala AQ, Adjemian S, Senovilla L, Michaud M, Spaggiari S, Vacchelli E, Baracco EE, Galluzzi L, Zitvogel L, Kepp O, Kroemer G. Screening of novel immunogenic cell death inducers within the NCI Mechanistic Diversity Set. Oncoimmunology 2014; 3:e28473. [PMID: 25050214 PMCID: PMC4063139 DOI: 10.4161/onci.28473] [Citation(s) in RCA: 103] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2014] [Accepted: 03/08/2014] [Indexed: 12/22/2022] Open
Abstract
Immunogenic cell death (ICD) inducers can be defined as agents that exert cytotoxic effects while stimulating an immune response against dead cell-associated antigens. When initiated by anthracyclines, ICD is accompanied by stereotyped molecular changes, including the pre-apoptotic exposure of calreticulin (CRT) on the cell surface, the lysosomal secretion of ATP during the blebbing phase of apoptosis, and the release of high mobility group box 1 (HMGB1) from dead cells. By means of genetically engineered human osteosarcoma U2OS cells, we screened the 879 anticancer compounds of the National Cancer Institute (NCI) Mechanistic Diversity Set for their ability to promote all these hallmarks of ICD in vitro. In line with previous findings from our group, several cardiac glycosides exhibit a robust propensity to elicit the major manifestations of ICD in cultured neoplastic cells. This screen pointed to septacidin, an antibiotic produced by Streptomyces fibriatus, as a novel putative inducer of ICD. In low-throughput validation experiments, septacidin promoted CRT exposure, ATP secretion and HGMB1 release from both U2OS cells and murine fibrosarcoma MCA205 cells. Moreover, septacidin-killed MCA205 cells protected immunocompetent mice against a re-challenge with living cancer cells of the same type. Finally, the antineoplastic effects of septacidin on established murine tumors were entirely dependent on T lymphocytes. Altogether, these results underscore the suitability of the high-throughput screening system described here for the identification of novel ICD inducers.
Collapse
Affiliation(s)
- Abdul Qader Sukkurwala
- Equipe 11 labellisée par la Ligue Nationale contre le cancer, Centre de Recherche des Cordeliers; Paris, France ; INSERM, U1138; Paris, France ; Gustave Roussy Comprehensive Cancer Center; Villejuif, France
| | - Sandy Adjemian
- Equipe 11 labellisée par la Ligue Nationale contre le cancer, Centre de Recherche des Cordeliers; Paris, France ; INSERM, U1138; Paris, France ; Gustave Roussy Comprehensive Cancer Center; Villejuif, France
| | - Laura Senovilla
- Equipe 11 labellisée par la Ligue Nationale contre le cancer, Centre de Recherche des Cordeliers; Paris, France ; INSERM, U1138; Paris, France ; Gustave Roussy Comprehensive Cancer Center; Villejuif, France ; INSERM, U1015; Villejuif, France
| | - Mickaël Michaud
- Equipe 11 labellisée par la Ligue Nationale contre le cancer, Centre de Recherche des Cordeliers; Paris, France ; INSERM, U1138; Paris, France ; Gustave Roussy Comprehensive Cancer Center; Villejuif, France
| | - Sabrina Spaggiari
- Equipe 11 labellisée par la Ligue Nationale contre le cancer, Centre de Recherche des Cordeliers; Paris, France ; INSERM, U1138; Paris, France ; Metabolomics and Cell Biology Platforms, Gustave Roussy Comprehensive Cancer Center; Villejuif, France
| | - Erika Vacchelli
- Equipe 11 labellisée par la Ligue Nationale contre le cancer, Centre de Recherche des Cordeliers; Paris, France ; INSERM, U1138; Paris, France ; Gustave Roussy Comprehensive Cancer Center; Villejuif, France
| | - Elisa Elena Baracco
- Equipe 11 labellisée par la Ligue Nationale contre le cancer, Centre de Recherche des Cordeliers; Paris, France ; INSERM, U1138; Paris, France ; Gustave Roussy Comprehensive Cancer Center; Villejuif, France
| | - Lorenzo Galluzzi
- Equipe 11 labellisée par la Ligue Nationale contre le cancer, Centre de Recherche des Cordeliers; Paris, France ; INSERM, U1138; Paris, France ; Gustave Roussy Comprehensive Cancer Center; Villejuif, France ; Université Paris Descartes/Paris 5; Sorbonne Paris Cité; Paris, France
| | - Laurence Zitvogel
- Gustave Roussy Comprehensive Cancer Center; Villejuif, France ; INSERM, U1015; Villejuif, France ; Université de Paris Saclay, Faculté de Médecine; Le Kremlin Bicêtre, France ; Center of Clinical Investigations in Biotherapies of Cancer (CICBT) 507; Villejuif, France
| | - Oliver Kepp
- Equipe 11 labellisée par la Ligue Nationale contre le cancer, Centre de Recherche des Cordeliers; Paris, France ; INSERM, U1138; Paris, France ; Metabolomics and Cell Biology Platforms, Gustave Roussy Comprehensive Cancer Center; Villejuif, France
| | - Guido Kroemer
- Equipe 11 labellisée par la Ligue Nationale contre le cancer, Centre de Recherche des Cordeliers; Paris, France ; INSERM, U1138; Paris, France ; Metabolomics and Cell Biology Platforms, Gustave Roussy Comprehensive Cancer Center; Villejuif, France ; Université Paris Descartes/Paris 5; Sorbonne Paris Cité; Paris, France ; Pôle de Biologie, Hôpital Européen Georges Pompidou, AP-HP; Paris, France
| |
Collapse
|