1
|
Mohammad-Jafari K, Naghib SM, Mozafari MR. Cisplatin-based Liposomal Nanocarriers for Drug Delivery in Lung Cancer Therapy: Recent Progress and Future Outlooks. Curr Pharm Des 2024; 30:2850-2881. [PMID: 39051580 DOI: 10.2174/0113816128304923240704113319] [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: 02/13/2024] [Accepted: 05/23/2024] [Indexed: 07/27/2024]
Abstract
In order to improve the treatment of lung cancer, this paper looks at the development of cisplatinbased liposomal nanocarriers. It focuses on addressing the drawbacks of conventional cisplatin therapy, including systemic toxicity, inadequate tumor targeting, and drug resistance. Liposomes, or spherical lipid vesicles, offer a potentially effective way to encapsulate cisplatin, enhancing its transport and minimizing harmful effects on healthy tissues. The article discusses many liposomal cisplatin formulations, including pH-sensitive liposomes, sterically stabilized liposomes, and liposomes coupled with specific ligands like EGFR antibodies. These novel formulations show promise in reducing cisplatin resistance, optimizing pharmacokinetics, and boosting therapeutic results in the two in vitro and in vivo models. They also take advantage of the Enhanced Permeability and Retention (EPR) effect in the direction of improved tumor accumulation. The study highlights the need for more investigation to move these liposomal formulations from experimental to clinical settings, highlighting their potential to offer less harmful and more effective cancer therapy alternatives.
Collapse
Affiliation(s)
- Kave Mohammad-Jafari
- Nanotechnology Department, School of Advanced Technologies, Iran University of Science and Technology, Tehran, Iran
| | - Seyed Morteza Naghib
- Nanotechnology Department, School of Advanced Technologies, Iran University of Science and Technology, Tehran, Iran
| | - M R Mozafari
- Australasian Nanoscience and Nanotechnology Initiative (ANNI), Monash University LPO, Clayton, VIC 3168, Australia
| |
Collapse
|
2
|
Alshahrani MY, Alkhathami AG, Almoyad MAA, Ahmad MZ, Mohanto S, Ahmad W, Wahab S. Phytochemicals as potential inhibitors of interleukin-8 for anticancer therapy: in silico evaluation and molecular dynamics analysis. J Biomol Struct Dyn 2023:1-12. [PMID: 38116764 DOI: 10.1080/07391102.2023.2294387] [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: 10/09/2023] [Accepted: 12/03/2023] [Indexed: 12/21/2023]
Abstract
Within the realm of soluble factors that have emerged as potential targets for therapeutic intervention, the chemokine interleukin-8 (IL-8) has garnered attention as a potential contributor to treatment responses in various cancer types. The utilization of naturally occurring anticancer compounds for treating cancer patients has shown substantial advancements in survival rates across early and advanced stages of the disease. In silico research findings provide support for the application of phytochemicals as potential inhibitors of IL-8, and phytochemicals exhibiting a high binding free energy and crucial interactions display promising anticancer properties, positioning them as candidates for future drug development. Noteworthy phytochemicals such as IMPHY006634 (Isohydnocarpin), IMPHY007957 (Chitranone) and IMPHY013015 (1-Hydroxyrutaecarpine) were predicted to possess inhibitory activity against IL-8, with calculated energies ranging from -9.9 to -9.1 kcal/mol, respectively. Several hydrogen bonds, including common amino acid residues Lys9 and CYS48, were identified. Molecular dynamics calculations conducted on these potent inhibitors demonstrated their stability throughout a 200 ns simulation, as indicated by metrics such as RMSD, RMSF, Rg, SASA, H-bonds, PCA and FEL analysis. Moreover, PASS analysis and adherence of these natural compounds to drug-likeness rules like Lipinski's further strengthen their candidacy. Considering these calculations and various parameters, these three prominent natural compounds emerge as promising candidates for anti-IL-8 therapy in the management of cancer.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Mohammad Y Alshahrani
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia
| | - Ali Gaithan Alkhathami
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia
| | - Mohammad Ali Abdullah Almoyad
- Department of Basic Medical Sciences, College of Applied Medical Sciences, King Khalid University, Abha, Khamis Mushyt, Saudi Arabia
| | - Mohammad Zaki Ahmad
- Department of Pharmaceutics, College of Pharmacy, Najran University, Najran, Saudi Arabia
| | - Sourav Mohanto
- Department of Pharmaceutics, Yenepoya Pharmacy College & Research Centre, Yenepoya (Deemed to be University), Mangalore, Karnataka, India
| | - Wasim Ahmad
- Department of Pharmacy, Mohammed Al-Mana College for Medical Sciences, Dammam, Saudi Arabia
| | - Shadma Wahab
- Department of Pharmacognosy, College of Pharmacy, King Khalid University, Abha, Saudi Arabia
| |
Collapse
|
3
|
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
|
4
|
Eerkens AL, Vledder A, Rooij N, Foijer F, Nijman HW, Bruyn M. Rapid and efficient generation of antigen‐specific isogenic T cells from cryopreserved blood samples. Immunol Cell Biol 2022; 100:285-295. [PMID: 35194830 PMCID: PMC9314923 DOI: 10.1111/imcb.12538] [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: 11/11/2021] [Revised: 02/04/2022] [Accepted: 02/18/2022] [Indexed: 11/28/2022]
Abstract
Clustered regularly interspaced short palindromic repeats/CRISPR‐associated protein 9 (CRISPR/Cas9)‐mediated gene editing has been leveraged for the modification of human and mouse T cells. However, limited experience is available on the application of CRISPR/Cas9 electroporation in cryopreserved T cells collected during clinical trials. To address this, we aimed to optimize a CRISPR/Cas9‐mediated gene editing protocol compatible with peripheral blood mononuclear cells (PBMCs) samples routinely produced during clinical trials. PBMCs from healthy donors were used to generate knockout T‐cell models for interferon‐γ, Cbl proto‐oncogene B (CBLB), Fas cell surface death receptor (Fas) and T‐cell receptor (TCRαβ) genes. The effect of CRISPR/Cas9‐mediated gene editing on T cells was evaluated using apoptosis assays, cytokine bead arrays and ex vivo and in vitro stimulation assays. Our results demonstrate that CRISPR/Cas9‐mediated gene editing of ex vivo T cells is efficient and does not overtly affect T‐cell viability. Cytokine release and T‐cell proliferation were not affected in gene‐edited T cells. Interestingly, memory T cells were more susceptible to CRISPR/Cas9 gene editing than naïve T cells. Ex vivo and in vitro stimulation with antigens resulted in equivalent antigen‐specific T‐cell responses in gene‐edited and untouched control cells, making CRISPR/Cas9‐mediated gene editing compatible with clinical antigen‐specific T‐cell activation and expansion assays. Here, we report an optimized protocol for rapid, viable and highly efficient genetic modification in ex vivo human antigen‐specific T cells, for subsequent functional evaluation and/or expansion. Our platform extends CRISPR/Cas9‐mediated gene editing for use in gold‐standard clinically used immune‐monitoring pipelines and serves as a starting point for development of analogous approaches, such as those including transcriptional activators and/or epigenetic modifiers.
Collapse
Affiliation(s)
- Anneke L Eerkens
- Department of Obstetrics and Gynecology University of Groningen University Medical Center Groningen Groningen The Netherlands
| | - Annegé Vledder
- Department of Obstetrics and Gynecology University of Groningen University Medical Center Groningen Groningen The Netherlands
| | - Nienke Rooij
- Department of Obstetrics and Gynecology University of Groningen University Medical Center Groningen Groningen The Netherlands
| | - Floris Foijer
- European Research Institute for the Biology of Ageing University of Groningen University Medical Center Groningen Groningen The Netherlands
| | - Hans W Nijman
- Department of Obstetrics and Gynecology University of Groningen University Medical Center Groningen Groningen The Netherlands
| | - Marco Bruyn
- Department of Obstetrics and Gynecology University of Groningen University Medical Center Groningen Groningen The Netherlands
| |
Collapse
|
5
|
Chinami M, Iwabuchi K, Muto Y, Uchida Y, Arita R, Shuraim RA, Adra CN. Assessment by miRNA microarray of an autologous cancer antigen-pulsed adoptive immune ensemble cell therapy (AC-ACT) approach; demonstrated induction of anti-oncogenic and anti-PD-L1 miRNAs. Clin Case Rep 2019; 7:2156-2164. [PMID: 31788270 PMCID: PMC6878052 DOI: 10.1002/ccr3.2343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Revised: 06/13/2019] [Accepted: 06/20/2019] [Indexed: 11/11/2022] Open
Abstract
A 60-year-old woman with stage IV rectal cancer received adoptive cell therapy with autologous cancer antigen (AC-ACT) causing induction of anti-oncogenic and anti-PD-L1 miRNAs as assessed by miRNA microarray. More than 1 year after AC-ACT, metastases have been arrested, and the patient reports good quality of life.
Collapse
Affiliation(s)
| | | | - Yoshiteru Muto
- The Research Institute of Health Rehabilitation of TokyoTokyoJapan
| | - Yasuhiko Uchida
- The Research Institute of Health Rehabilitation of TokyoTokyoJapan
| | - Ryu Arita
- Fukuoka MSC Medical ClinicsFukuokaJapan
| | | | - Chaker N. Adra
- BFSR InstituteFukuokaJapan
- The Adra InstituteBoston, MAUSA
| |
Collapse
|
6
|
Park HS, Kwon WS, Park S, Jo E, Lim SJ, Lee CK, Lee JB, Jung M, Kim HS, Beom SH, Park JY, Kim TS, Chung HC, Rha SY. Comprehensive immune profiling and immune-monitoring using body fluid of patients with metastatic gastric cancer. J Immunother Cancer 2019; 7:268. [PMID: 31639056 PMCID: PMC6805480 DOI: 10.1186/s40425-019-0708-8] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Accepted: 08/14/2019] [Indexed: 02/08/2023] Open
Abstract
Background The aim of this study is to profile the cytokines and immune cells of body fluid from metastatic gastric cancer (mGC), and evaluate the potential role as a prognostic factor and the feasibility as a predictive biomarker or monitoring source for immune checkpoint inhibitor. Methods Body fluid including ascites and pleural fluid were obtained from 55 mGC patients and 24 matched blood. VEGF-A, IL-10, and TGF-β1 were measured and immune cells were profiled by fluorescence assisted cell sorting (FACS). Results VEGF-A and IL-10 were significantly higher in body fluid than in plasma of mGC. Proportion of T lymphocytes with CD69 or PD-1, memory T cell marked with CD45RO, and number of Foxp3+ T regulatory cells (Tregs) were significantly higher in body fluid than those in blood of mGC. Proportion of CD8 T lymphocyte with memory marker (CD45RO) and activation marker (HLA-DR), CD3 T lymphocyte with PD-1, and number of FoxP3+ Tregs were identified as independent prognostic factors. When patients were classified by molecular subgroups of primary tumor, VEGF-A was significantly higher in genomically stable (GS)-like group than that in chromosomal instability (CIN)-like group while PD-L1 positive tumor cells (%) showed opposite results. Monitoring immune dynamics using body fluid was also feasible. Early activated T cell marked with CD25 was significantly increased in chemotherapy treated group. Conclusions By analyzing cytokines and proportion of immune cells in body fluid, prognosis of patients with mGC can be predicted. Immune monitoring using body fluid may provide more effective treatment for patients with mGC. Electronic supplementary material The online version of this article (10.1186/s40425-019-0708-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Hyung Soon Park
- Division of Medical Oncology, Department of Internal Medicine, St. Vincent's Hospital, The Catholic University of Korea, Suwon, South Korea.,Songdang Institute for Cancer Research, Yonsei University College of Medicine, Seoul, South Korea
| | - Woo Sun Kwon
- Songdang Institute for Cancer Research, Yonsei University College of Medicine, Seoul, South Korea.,Brain Korea 21 Project for Medical Sciences, Yonsei University College of Medicine, Seoul, South Korea
| | - Sejung Park
- Songdang Institute for Cancer Research, Yonsei University College of Medicine, Seoul, South Korea.,Brain Korea 21 Project for Medical Sciences, Yonsei University College of Medicine, Seoul, South Korea
| | - Eunji Jo
- Songdang Institute for Cancer Research, Yonsei University College of Medicine, Seoul, South Korea.,Brain Korea 21 Project for Medical Sciences, Yonsei University College of Medicine, Seoul, South Korea
| | - So Jung Lim
- Songdang Institute for Cancer Research, Yonsei University College of Medicine, Seoul, South Korea
| | - Choong-Kun Lee
- Division of Medical Oncology, Department of Internal Medicine, Yonsei Cancer Center, Yonsei University College of Medicine, 50-1 Yonsei-Ro, Seodaemun-gu, Seoul, 120-752, South Korea
| | - Jii Bum Lee
- Division of Medical Oncology, Department of Internal Medicine, Yonsei Cancer Center, Yonsei University College of Medicine, 50-1 Yonsei-Ro, Seodaemun-gu, Seoul, 120-752, South Korea
| | - Minkyu Jung
- Songdang Institute for Cancer Research, Yonsei University College of Medicine, Seoul, South Korea.,Division of Medical Oncology, Department of Internal Medicine, Yonsei Cancer Center, Yonsei University College of Medicine, 50-1 Yonsei-Ro, Seodaemun-gu, Seoul, 120-752, South Korea
| | - Hyo Song Kim
- Songdang Institute for Cancer Research, Yonsei University College of Medicine, Seoul, South Korea.,Division of Medical Oncology, Department of Internal Medicine, Yonsei Cancer Center, Yonsei University College of Medicine, 50-1 Yonsei-Ro, Seodaemun-gu, Seoul, 120-752, South Korea
| | - Seung-Hoon Beom
- Songdang Institute for Cancer Research, Yonsei University College of Medicine, Seoul, South Korea.,Division of Medical Oncology, Department of Internal Medicine, Yonsei Cancer Center, Yonsei University College of Medicine, 50-1 Yonsei-Ro, Seodaemun-gu, Seoul, 120-752, South Korea
| | - Jun Yong Park
- Division of Gastroenterology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, South Korea
| | - Tae Soo Kim
- Songdang Institute for Cancer Research, Yonsei University College of Medicine, Seoul, South Korea
| | - Hyun Cheol Chung
- Songdang Institute for Cancer Research, Yonsei University College of Medicine, Seoul, South Korea.,Brain Korea 21 Project for Medical Sciences, Yonsei University College of Medicine, Seoul, South Korea.,Division of Medical Oncology, Department of Internal Medicine, Yonsei Cancer Center, Yonsei University College of Medicine, 50-1 Yonsei-Ro, Seodaemun-gu, Seoul, 120-752, South Korea
| | - Sun Young Rha
- Songdang Institute for Cancer Research, Yonsei University College of Medicine, Seoul, South Korea. .,Brain Korea 21 Project for Medical Sciences, Yonsei University College of Medicine, Seoul, South Korea. .,Division of Medical Oncology, Department of Internal Medicine, Yonsei Cancer Center, Yonsei University College of Medicine, 50-1 Yonsei-Ro, Seodaemun-gu, Seoul, 120-752, South Korea.
| |
Collapse
|
7
|
Weber M, Wehrhan F, Baran C, Agaimy A, Büttner-Herold M, Kesting M, Ries J. Prognostic significance of PD-L2 expression in patients with oral squamous cell carcinoma-A comparison to the PD-L1 expression profile. Cancer Med 2019; 8:1124-1134. [PMID: 30659749 PMCID: PMC6434218 DOI: 10.1002/cam4.1929] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2018] [Revised: 11/01/2018] [Accepted: 11/26/2018] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Despite the observed association of increased PD-L1 expression in peripheral blood of oral squamous cell carcinoma (OSCC) patients with histomorphologic parameters, the role of the PD1 ligands-PD-L1 and PD-L2-is insufficiently understood. Aim of the study was to investigate whether the alterations of PD-L1 and PD-L2 expression in blood are associated with survival and could serve as immune monitoring parameter. Moreover, it should be analyzed if PD-L2 is differentially expressed in tissue and blood samples of OSCC patients compared to healthy controls and if there is an association of PD-L2 expression with histomorphologic and prognostic tumor parameters. METHODS PD-L2 mRNA expression was analyzed in tumors and healthy oral mucosa specimens and in corresponding peripheral blood samples of 48 OSCC patients and 26 healthy controls using RT-qPCR. A cutoff point (COP) was determined and a chi-square test (χ2 test) was carried out. Survival analysis of PD-L2 and previously reported PD-L1 expression data was performed using Kaplan-Meier analysis (Log-rank test). RESULTS PD-L2 expression in tissue samples was significantly (P < 0.001) higher in OSCC patients compared to healthy controls. A significant association of PD-L2 expression above the COP (positive) with malignancy was ascertained (P < 0.001). A significant (P = 0.01) association of previously reported PD-L1 expression rates in peripheral blood with survival could be shown. CONCLUSION Peripheral blood PD-L1 expression might be a prognostic marker for OSCC patients and a possible parameter to monitor immune dysfunction in malign diseases. In the peripheral blood, PD-L1 might be more relevant for immune tolerance than PD-L2. Local PD-L2 expression in tissue samples might be useful as a diagnostic parameter for malignancy and could contribute to the immunosuppressive local microenvironment in OSCC.
Collapse
Affiliation(s)
- Manuel Weber
- Department of Oral and Maxillofacial Surgery, Friedrich-Alexander University Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Falk Wehrhan
- Department of Oral and Maxillofacial Surgery, Friedrich-Alexander University Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Christoph Baran
- Department of Oral and Maxillofacial Surgery, Friedrich-Alexander University Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Abbas Agaimy
- Institute of Pathology, Friedrich-Alexander University Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Maike Büttner-Herold
- Institute of Pathology, Department of Nephropathology, Friedrich-Alexander University Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Marco Kesting
- Department of Oral and Maxillofacial Surgery, Friedrich-Alexander University Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Jutta Ries
- Department of Oral and Maxillofacial Surgery, Friedrich-Alexander University Erlangen-Nürnberg (FAU), Erlangen, Germany
| |
Collapse
|
8
|
Bonomi M, Ahmed T, Addo S, Kooshki M, Palmieri D, Levine BJ, Ruiz J, Grant S, Petty WJ, Triozzi PL. Circulating immune biomarkers as predictors of the response to pembrolizumab and weekly low dose carboplatin and paclitaxel in NSCLC and poor PS: An interim analysis. Oncol Lett 2018; 17:1349-1356. [PMID: 30655905 DOI: 10.3892/ol.2018.9724] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Accepted: 10/26/2018] [Indexed: 12/19/2022] Open
Abstract
The combination of standard-dose chemotherapy and immunotherapy has been shown to be beneficial for patients with non-small cell lung cancer (NSCLC) with good performance status (PS). However, treatment options for patients with poor PS are limited. In the present study, the feasibility and immunological effects of low-dose chemotherapy with carboplatin and paclitaxel combined with immunotherapy with pembrolizumab were examined in patients with metastatic NSCLC and a poor PS. Patients with advanced NSCLC and a PS of 2 were randomized to single-agent pembrolizumab at 200 mg every 3 weeks or pembrolizumab combined with weekly carboplatin area under the curve 1 and paclitaxel 25 mg/m2. Blood for circulating immune cell phenotyping, soluble program death ligand 1 (sPD-L1) and immune-modulatory microRNAs (miRNAs) was collected prior to treatment and at weeks 4 and 7. Ten patients were randomized to the combination arm and 10 to the single-agent arm. Therapy was well tolerated. Four patients discontinued carboplatin due to hypersensitivity reactions but continued pembrolizumab and paclitaxel treatments. Increases in activated CD4+ T cells and in immune-regulatory miRNA, and decreases in myeloid derived suppressor cells were observed in the blood of patients in the combination arm and not in the single-agent arm. Changes in circulating regulatory T cells and sPD-L1 were not observed. Seven patients in the combination arm manifested a partial response compared with only two in the single-agent arm. Weekly low-dose chemotherapy carboplatin and paclitaxel was well tolerated and immunologically active when combined with pembrolizumab in patients with advanced NSCLC and a PS of 2. This combination merits further study in this patient population.
Collapse
Affiliation(s)
- Marcelo Bonomi
- Department of Medical Oncology, The Ohio State University, Columbus, OH 43210, USA
| | - Tamjeed Ahmed
- Wake Forest University Comprehensive Cancer Center, Winston-Salem, NC 27157, USA
| | - Safoa Addo
- Department of Internal Medicine, Wake Forest Baptist Medical Center, Winston-Salem, NC 27157, USA
| | - Mitra Kooshki
- Wake Forest University Comprehensive Cancer Center, Winston-Salem, NC 27157, USA
| | - Dario Palmieri
- Solid Tumor Biology Program, Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA
| | - Beverly J Levine
- Division of Public Health Sciences, Wake Forest University Comprehensive Cancer Center, Medical Center Blvd, Winston-Salem, NC 27157, USA
| | - Jimmy Ruiz
- Wake Forest University Comprehensive Cancer Center, Winston-Salem, NC 27157, USA
| | - Stefan Grant
- Wake Forest University Comprehensive Cancer Center, Winston-Salem, NC 27157, USA
| | - William J Petty
- Wake Forest University Comprehensive Cancer Center, Winston-Salem, NC 27157, USA
| | - Pierre L Triozzi
- Wake Forest University Comprehensive Cancer Center, Winston-Salem, NC 27157, USA
| |
Collapse
|
9
|
Santegoets SJAM, de Groot AF, Dijkgraaf EM, Simões AMC, van der Noord VE, van Ham JJ, Welters MJP, Kroep JR, van der Burg SH. The blood mMDSC to DC ratio is a sensitive and easy to assess independent predictive factor for epithelial ovarian cancer survival. Oncoimmunology 2018; 7:e1465166. [PMID: 30221063 PMCID: PMC6136880 DOI: 10.1080/2162402x.2018.1465166] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Revised: 04/09/2018] [Accepted: 04/09/2018] [Indexed: 12/26/2022] Open
Abstract
Epithelial ovarian cancer (EOC) may cause abnormal blood levels of leukocytes. This paraneoplastic manifestation is associated with a worse response to therapy and shorter survival. To understand the complexity and nature of these leukocytes, we dissected the different populations of myeloid cells and analyzed their relation to clinical outcome. Therefore, baseline blood samples of 36 EOC patients treated either with carboplatin/doxorubucin or with gemcitabine were analyzed for different subsets of monocytes/macrophages, myeloid derived suppressor cells (MDSC) and dendritic cells (DC) using multiparameter flow cytometry as well as functional assays for myeloid cell mediated suppression of antigen-specific T cell reactivity. Healthy donor blood served as control. EOC patients displayed an increase in monocytes/macrophages, monocytic MDSC (mMDSC) and CD33-CD11b+CD14-CD15- double-negative MDSC (CD33- dnMDSC) and a decrease in the frequency of DC, across all EOC subtypes. A low frequency of DC and high frequencies of monocytes/macrophages and mMDSC, but not CD33- dnMDSC, were associated with poor overall survival. Patient's monocytes/macrophages and mMDSC, but not CD33- dnMDSC, were shown to suppress T cell reactivity in vitro. The mMDSC and DC frequencies were not altered upon treatment. Importantly, the mMDSC to DC ratio was the strongest independent, highly sensitive and specific, predictive factor for survival. This was irrespective of the type of chemotherapy or disease stage and outperformed classical parameters as WHO status or time from last chemotherapy. Thus, the baseline blood mMDSC to DC ratio is a robust, independent and easy to analyze predictive factor for EOC survival, and may assist patient selection for immunotherapy.
Collapse
Affiliation(s)
- S J A M Santegoets
- Department of Medical Oncology, Leiden University Medical Center, Leiden, The Netherlands
| | - A F de Groot
- Department of Medical Oncology, Leiden University Medical Center, Leiden, The Netherlands
| | - E M Dijkgraaf
- Department of Medical Oncology, Leiden University Medical Center, Leiden, The Netherlands
| | - A M Carnaz Simões
- Department of Medical Oncology, Leiden University Medical Center, Leiden, The Netherlands
| | - V E van der Noord
- Department of Medical Oncology, Leiden University Medical Center, Leiden, The Netherlands
| | - J J van Ham
- Department of Medical Oncology, Leiden University Medical Center, Leiden, The Netherlands
| | - M J P Welters
- Department of Medical Oncology, Leiden University Medical Center, Leiden, The Netherlands
| | - J R Kroep
- Department of Medical Oncology, Leiden University Medical Center, Leiden, The Netherlands
| | - S H van der Burg
- Department of Medical Oncology, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
10
|
Zhou Y, Huang J, Bai Y, Li C, Lu X. Effects of preemptive analgesia with flurbiprofen ester on lymphocytes and natural killer cells in patients undergoing esophagectomy: A randomized controlled pilot study. Thorac Cancer 2017; 8:649-654. [PMID: 28892265 PMCID: PMC5668486 DOI: 10.1111/1759-7714.12502] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Revised: 08/03/2017] [Accepted: 08/03/2017] [Indexed: 02/02/2023] Open
Abstract
BACKGROUND Tumors may induce systemic immune dysfunction, which can be aggravated by surgery and anesthesia/analgesia. Data on the effect of flurbiprofen preemptive analgesia on immune dysfunction is limited. The aim of this study was to investigate the effect of flurbiprofen preemptive analgesia on lymphocytes and natural killer (NK) cells in patients undergoing thoracotomy and thoracoscopy radical esophagectomy, and to explore the analgesic methods suitable for tumor patients. METHODS This was a randomized controlled pilot study of 89 patients with esophageal cancer treated with surgery at the Henan Cancer Hospital between January 1, 2015 and December 31, 2016. The patients were divided into three groups: group 1, thoracotomy; group 2, thoracoscopy and laparoscopic surgery; and group 3, flurbiprofen, thoracoscopy, and laparoscopic surgery. CD3+, CD19+, NK, CD4+, and CD8+ cells in whole blood were measured by flow cytometry 30 minutes before surgery (T0), at the end of the thoracic section of the procedure (T1), and at the end of the operation (T2). RESULTS There were no significant differences in CD3+, CD19+, CD8+, NK, and CD4+ cells between the three groups or regarding the time points during the procedure (all P > 0.05). Thoracotomy and thoracoscopy surgery resulted in similar immunological outcomes. CONCLUSION Flurbiprofen ester preemptive analgesia did not suppress the immune function in patients and could be a safe analgesic method for patients with esophageal cancer undergoing surgery.
Collapse
Affiliation(s)
- Yi Zhou
- Department of AnesthesiologyThe Affiliated Cancer Hospital of Zhengzhou UniversityZhengzhouChina
| | - Jinxi Huang
- Department of General SurgeryThe Affiliated Cancer Hospital of Zhengzhou UniversityZhengzhouChina
| | - Yu Bai
- Department of AnesthesiologyThe Affiliated Cancer Hospital of Zhengzhou UniversityZhengzhouChina
| | - Changsheng Li
- Department of AnesthesiologyThe Affiliated Cancer Hospital of Zhengzhou UniversityZhengzhouChina
| | - Xihua Lu
- Department of AnesthesiologyThe Affiliated Cancer Hospital of Zhengzhou UniversityZhengzhouChina
| |
Collapse
|
11
|
Rodríguez-Cerdeira C, Carnero Gregorio M, López-Barcenas A, Sánchez-Blanco E, Sánchez-Blanco B, Fabbrocini G, Bardhi B, Sinani A, Guzman RA. Advances in Immunotherapy for Melanoma: A Comprehensive Review. Mediators Inflamm 2017; 2017:3264217. [PMID: 28848246 PMCID: PMC5564072 DOI: 10.1155/2017/3264217] [Citation(s) in RCA: 79] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Revised: 03/21/2017] [Accepted: 04/03/2017] [Indexed: 12/18/2022] Open
Abstract
Melanomas are tumors originating from melanocytes and tend to show early metastasis secondary to the loss of cellular adhesion in the primary tumor, resulting in high mortality rates. Cancer-specific active immunotherapy is an experimental form of treatment that stimulates the immune system to recognize antigens on the surface of cancer cells. Current experimental approaches in immunotherapy include vaccines, biochemotherapy, and the transfer of adoptive T cells and dendritic cells. Several types of vaccines, including peptide, viral, and dendritic cell vaccines, are currently under investigation for the treatment of melanoma. These treatments have the same goal as drugs that are already used to stimulate the proliferation of T lymphocytes in order to destroy tumor cells; however, immunotherapies aim to selectively attack the tumor cells of each patient. In this comprehensive review, we describe recent advancements in the development of immunotherapies for melanoma, with a specific focus on the identification of neoantigens for the prediction of their elicited immune responses. This review is expected to provide important insights into the future of immunotherapy for melanoma.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Ardiana Sinani
- Dermatology Service, Military Medical Unit, University Trauma Hospital, Tirana, Albania
| | | |
Collapse
|
12
|
Fang WF, Chen YM, Lin CY, Huang KT, Kao HC, Fang YT, Huang CH, Chang YT, Wang YH, Wang CC, Lin MC. Immune profiles and clinical outcomes between sepsis patients with or without active cancer requiring admission to intensive care units. PLoS One 2017; 12:e0179749. [PMID: 28692671 PMCID: PMC5503229 DOI: 10.1371/journal.pone.0179749] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2017] [Accepted: 06/02/2017] [Indexed: 12/15/2022] Open
Abstract
Background Immunoparalysis was observed in both patients with cancer and sepsis. In cancer patients, Cytotoxic T lymphocyte antigen-4 and programmed cell death protein 1/programmed death-ligand 1 axis are two key components of immunoparalysis. Several emerging therapies against these two axes gained significant clinical benefit. In severe sepsis patients, immunoparalysis was known as compensatory anti-inflammatory response syndrome and this has been suggested as an important cause of death in patients with sepsis. It would be interesting to see if immune status was different in severe sepsis patients with or without active cancer. The aim of this study was to assess the differences in immune profiles, and clinical outcomes between severe sepsis patients with or without cancer admitted to ICU. Methods A combined retrospective and prospective observational study from a cohort of adult sepsis patients admitted to three medical ICUs at Kaohsiung Chang Gung Memorial Hospital in Taiwan between August 2013 and June 2016. Results Of the 2744 patients admitted to the ICU, 532 patients with sepsis were included. Patients were divided into those with or without active cancer according to their medical history. Of the 532 patients, 95 (17.9%) patients had active cancer, and 437 (82.1%) patients had no active cancer history. Patients with active cancer were younger (p = 0.001) and were less likely to have diabetes mellitus (p < 0.001), hypertension (p < 0.001), coronary artery disease (p = 0.004), chronic obstructive pulmonary disease (p = 0.002) or stroke (p = 0.002) compared to patients without active cancer. Patients with active cancer also exhibited higher baseline lactate levels (p = 0.038), and higher baseline plasma interleukin (IL)-10 levels (p = 0.040), higher trend of granulocyte colony-stimulating factor (G-CSF) (p = 0.004) compared to patients without active cancer. The 14-day, 28-day and 90-day mortality rates were higher for patients with active cancer than those without active cancer (P < 0.001 for all intervals). Conclusions Among patients admitted to the ICU with sepsis, those with underling active cancer had higher baseline levels of plasma IL-10, higher trend of G-CSF and higher mortality rate than those without active cancer.
Collapse
Affiliation(s)
- Wen-Feng Fang
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Niaosung, Kaohsiung, Taiwan
- Department of Respiratory Therapy, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung, Taiwan
- Department of Respiratory Care, Chang Gung University of Science and Technology, Chiayi, Taiwan
- * E-mail: (MCL); (WFF)
| | - Yu-Mu Chen
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Niaosung, Kaohsiung, Taiwan
| | - Chiung-Yu Lin
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Niaosung, Kaohsiung, Taiwan
| | - Kuo-Tung Huang
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Niaosung, Kaohsiung, Taiwan
| | - Hsu-Ching Kao
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Niaosung, Kaohsiung, Taiwan
| | - Ying-Tang Fang
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Niaosung, Kaohsiung, Taiwan
| | - Chi-Han Huang
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Niaosung, Kaohsiung, Taiwan
| | - Ya-Ting Chang
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Niaosung, Kaohsiung, Taiwan
| | - Yi-His Wang
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Niaosung, Kaohsiung, Taiwan
- Department of Respiratory Therapy, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Chin-Chou Wang
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Niaosung, Kaohsiung, Taiwan
- Department of Respiratory Therapy, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung, Taiwan
- Department of Respiratory Care, Chang Gung University of Science and Technology, Chiayi, Taiwan
| | - Meng-Chih Lin
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Niaosung, Kaohsiung, Taiwan
- Department of Respiratory Therapy, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung, Taiwan
- * E-mail: (MCL); (WFF)
| |
Collapse
|
13
|
Abstract
PURPOSE OF REVIEW The growing awareness that the immune system is a key player in the antitumoral response and the excellent clinical results achieved in some settings with anti-programmed cell death 1 (PD1)/programmed death ligand 1 (PDL1) and anti-cytotoxic T-lymphocyte-associated protein 4 (CTLA4) drugs has led to the rise of immunotherapy as a supplement or an alternative to conventional cancer treatment. The high costs associated with these therapies, their significant toxicity and the need to understand and circumvent immune escape mechanisms raise the urgent need for immunological assessment of therapy response. The study of the immunological parameters before, during and after treatment is referred to as immunomonitoring. This review discusses the current knowledge of immunomonitoring markers in gastrointestinal cancers. RECENT FINDINGS The last decade has seen a collaborative effort to standardize the assays performed in clinical trials to assess response to immunotherapy. Since then, multiple studies have been conducted on blood samples, biopsies and surgical specimens to determine their immunological profiles leading to the identification of several immunological markers possessing a predictive value of response to treatment. SUMMARY Future research will focus on detangling the predictive value of immune markers in different therapeutic models, and also to develop new noninvasive means to monitor the immune response of patients. VIDEO ABSTRACT: http://links.lww.com/COON/A20.
Collapse
|