1
|
McKechnie T, Talwar G, Grewal S, Wang A, Eskicioglu C, Parvez E. The impact of statins on melanoma survival: a systematic review and meta-analysis. Melanoma Res 2024; 34:475-486. [PMID: 39264579 DOI: 10.1097/cmr.0000000000001001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/13/2024]
Abstract
Statin use may decrease recurrence and improve survival in patients with melanoma. In this systematic review and meta-analysis, we examine the current body of literature concerning the use of statins as an adjunctive therapy in melanoma, Medline, EMBASE, CENTRAL, and PubMed were systematically searched from inception through to April 2023. Studies were included if they compared patients with melanoma receiving and not receiving statin therapy concurrently with their oncologic treatment in terms of long-term oncologic outcomes. The primary outcome was 5-year overall survival (OS). Meta-analyses was performed with DerSimonian and Laird random effects. Risk of bias was assessed with the ROBINS-I and GRADE was used to assess certainty of evidence. From 952 citations, eight non-randomized studies were identified. Included studies were conducted between 2007 and 2022. Random effects meta-analysis of adjusted hazard ratios from three studies suggested an improvement in 5-year OS with statin use with wide 95% confidence intervals (CIs) crossing the line of no effect (hazard ratio 0.87, 95% CI: 0.73-1.04, P = 0.12, I2 = 95%, very-low certainty). Outcome reporting was heterogeneous across all other oncologic outcomes such that pooling of data was not possible. Risk of bias was serious for seven studies and moderate for one study. This systematic review of studies evaluating the impact of statin use on survival in patients with melanoma found a 13% reduction in risk of death at 5 years from diagnosis - a point estimate suggesting benefit. However, the wide 95% CIs and resultant type II error risk create significant uncertainty.
Collapse
Affiliation(s)
- Tyler McKechnie
- Division of General Surgery, Department of Surgery, McMaster University
| | - Gaurav Talwar
- Division of General Surgery, Department of Surgery, McMaster University
| | - Shan Grewal
- Department of Health Sciences, Michael G. DeGroote School of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Austine Wang
- Department of Health Sciences, Michael G. DeGroote School of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Cagla Eskicioglu
- Division of General Surgery, Department of Surgery, McMaster University
- Department of Health Sciences, Michael G. DeGroote School of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Elena Parvez
- Division of General Surgery, Department of Surgery, McMaster University
- Department of Health Sciences, Michael G. DeGroote School of Medicine, McMaster University, Hamilton, Ontario, Canada
| |
Collapse
|
2
|
Dong Y, Chen Z, Yang F, Wei J, Huang J, Long X. Prediction of immunotherapy responsiveness in melanoma through single-cell sequencing-based characterization of the tumor immune microenvironment. Transl Oncol 2024; 43:101910. [PMID: 38417293 PMCID: PMC10907870 DOI: 10.1016/j.tranon.2024.101910] [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: 11/27/2023] [Revised: 01/13/2024] [Accepted: 02/08/2024] [Indexed: 03/01/2024] Open
Abstract
Immune checkpoint inhibitors (ICB) therapy have emerged as effective treatments for melanomas. However, the response of melanoma patients to ICB has been highly heterogenous. Here, by analyzing integrated scRNA-seq datasets from melanoma patients, we revealed significant differences in the TiME composition between ICB-resistant and responsive tissues, with resistant or responsive tissues characterized by an abundance of myeloid cells and CD8+ T cells or CD4+ T cell predominance, respectively. Among CD4+ T cells, CD4+ CXCL13+ Tfh-like cells were associated with an immunosuppressive phenotype linked to immune escape-related genes and negative regulation of T cell activation. We also develop an immunotherapy response prediction model based on the composition of the immune compartment. Our predictive model was validated using CIBERSORTx on bulk RNA-seq datasets from melanoma patients pre- and post-ICB treatment and showed a better performance than other existing models. Our study presents an effective immunotherapy response prediction model with potential for further translation, as well as underscores the critical role of the TiME in influencing the response of melanomas to immunotherapy.
Collapse
Affiliation(s)
- Yucheng Dong
- Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China
| | - Zhizhuo Chen
- Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China
| | - Fan Yang
- Department of Liver Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Jiaxin Wei
- Department of Emergency Department, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Jiuzuo Huang
- Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China.
| | - Xiao Long
- Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China.
| |
Collapse
|
3
|
Patinote C, Raevens S, Baumann A, Pellegrin E, Bonnet PA, Deleuze-Masquéfa C. [1,2,4]triazolo[4,3- a]quinoxaline as Novel Scaffold in the Imiqualines Family: Candidates with Cytotoxic Activities on Melanoma Cell Lines. Molecules 2023; 28:5478. [PMID: 37513350 PMCID: PMC10384284 DOI: 10.3390/molecules28145478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 07/13/2023] [Accepted: 07/16/2023] [Indexed: 07/30/2023] Open
Abstract
Cutaneous melanoma is one of the most aggressive human cancers and is the deadliest form of skin cancer, essentially due to metastases. Novel therapies are always required, since cutaneous melanoma develop resistance to oncogenic pathway inhibition treatment. The Imiqualine family is composed of heterocycles diversely substituted around imidazo[1,2-a]quinoxaline, imidazo[1,2-a]pyrazine, imidazo[1,5-a]quinoxaline, and pyrazolo[1,5-a]quinoxaline scaffolds, which display interesting activities on a panel of cancer cell lines, especially melanoma cell lines. We have designed and prepared novel compounds based on the [1,2,4]triazolo[4,3-a]quinoxaline scaffold through a common synthetic route, using 1-chloro-2-hydrazinoquinoxaline and an appropriate aldehyde. Cyclization is ensured by an oxidation-reduction mechanism using chloranil. The substituents on positions 1 and 8 were chosen based on previous structure-activity relationship (SAR) studies conducted within our heterocyclic Imiqualine family. Physicochemical parameters of all compounds have also been predicted. A375 melanoma cell line viability has been evaluated for 16 compounds. Among them, three novel [1,2,4]triazolo[4,3-a]quinoxalines display cytotoxic activities. Compounds 16a and 16b demonstrate relative activities in the micromolar range (respectively, 3158 nM and 3527 nM). Compound 17a shows the best EC50 of the novel series (365 nM), even if EAPB02303 remains the lead of the entire Imiqualine family (3 nM).
Collapse
Affiliation(s)
- Cindy Patinote
- Institut des Biomolécules Max Mousseron (IBMM), UMR 5247 F16, (CNRS, ENSCM, Université de Montpellier), 1919 Route de Mende, 34090 Montpellier, France
| | - Sandy Raevens
- Institut des Biomolécules Max Mousseron (IBMM), UMR 5247 F16, (CNRS, ENSCM, Université de Montpellier), 1919 Route de Mende, 34090 Montpellier, France
| | - Amélie Baumann
- Institut des Biomolécules Max Mousseron (IBMM), UMR 5247 F16, (CNRS, ENSCM, Université de Montpellier), 1919 Route de Mende, 34090 Montpellier, France
| | - Eloise Pellegrin
- Institut des Biomolécules Max Mousseron (IBMM), UMR 5247 F16, (CNRS, ENSCM, Université de Montpellier), 1919 Route de Mende, 34090 Montpellier, France
| | - Pierre-Antoine Bonnet
- Institut des Biomolécules Max Mousseron (IBMM), UMR 5247 F16, (CNRS, ENSCM, Université de Montpellier), 1919 Route de Mende, 34090 Montpellier, France
| | - Carine Deleuze-Masquéfa
- Institut des Biomolécules Max Mousseron (IBMM), UMR 5247 F16, (CNRS, ENSCM, Université de Montpellier), 1919 Route de Mende, 34090 Montpellier, France
| |
Collapse
|
4
|
Zhang Y, Li Y, Yan H. Low Expression of CLEC2B Indicates Poor Prognosis in Melanoma. Clin Cosmet Investig Dermatol 2023; 16:463-477. [PMID: 36851951 PMCID: PMC9961593 DOI: 10.2147/ccid.s395854] [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: 11/18/2022] [Accepted: 02/11/2023] [Indexed: 02/22/2023]
Abstract
Background Melanoma is a highly malignant skin tumor with a poor prognosis. Identification of novel biomarkers might potentially reveal the underlying mechanisms of melanoma progression. Methods We demonstrated the relationship between pan-cancer CLEC2B expression and melanoma samples in The Cancer Genome Atlas (TCGA) database. Next, the Kaplan-Meier plot and Cox regression analysis determined the prognostic value of CLEC2B in melanoma. Biological pathway enrichment was screened by Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG), and Gene Set Enrichment Analysis (GSEA), enabling the correlation analysis between the immune infiltration level and CLEC2B expression in melanoma. Our final claim was validated using qPCR, immunohistochemistry, Western blot, cell colony formation assays, ethynyldeoxyuridine (Edu) analysis, and cell Invasion assays. Results Our study revealed that the high CLEC2B expression correlates with poor overall survival of melanoma patients. Moreover, a low expression of CLEC2B was found in the A375 cell line. In addition, CLEC2B has significant prognostic value in melanoma diagnosis, with an AUC of 0.896. Prognostic analysis showed the low expression of CLEC2B to be independently associated with melanoma patients. Moreover, the expression of CLEC2B was significantly correlated with B cells, eosinophils, macrophages, neutrophils, NK cells, T helper cells, Tregs, Th1 cells, Th17 cells, and Th2 cells. PCR and immunohistochemistry indicated CLEC2B to be significantly downregulated in melanoma. The cell colony formation assay showed CLEC2B knockout increased the proliferation of A375 cells. Conclusion Our study established low levels of CLEC2B to be poor prognostic markers, enabling immunosuppressive cell infiltration in melanoma.
Collapse
Affiliation(s)
- Yanqiu Zhang
- Department of Dermatology, The First Hospital of China Medical University, Shenyang, 110001, People's Republic of China
| | - Yaling Li
- Department of Dermatology, The First Hospital of China Medical University, Shenyang, 110001, People's Republic of China
| | - Hongwei Yan
- Department of Dermatology, The First Hospital of China Medical University, Shenyang, 110001, People's Republic of China
| |
Collapse
|
5
|
Jahani V, Yazdani M, Badiee A, Jaafari MR, Arabi L. Liposomal celecoxib combined with dendritic cell therapy enhances antitumor efficacy in melanoma. J Control Release 2023; 354:453-464. [PMID: 36649743 DOI: 10.1016/j.jconrel.2023.01.034] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2022] [Revised: 01/11/2023] [Accepted: 01/12/2023] [Indexed: 01/19/2023]
Abstract
Cancer vaccine efficacy is limited by the immunosuppressive nature of the tumor microenvironment created by inflammation, immune inhibitory factors, and regulatory T cells (Tregs). Inspired by the role of cyclooxygenase-2 (COX-2) in inflammation in the tumor site, we proposed that normalization of the tumor microenvironment by celecoxib as a COX-2 inhibitor might improve the efficacy of Dendritic Cell (DC) therapy in a melanoma model. In the present study, liposomal celecoxib (Lip-CLX) was combined with ex vivo generated DC vaccines pulsed with gp100 peptide (in liposomal and non-liposomal forms) for prophylactic and therapeutic evaluation in the B16F10 melanoma model. Tumor site analysis by flow cytometry demonstrated that intravenous administration of Lip-CLX at a dose of 1 mg/kg in four doses effectively normalized the tumor microenvironment by reducing Tregs and IL-10 production. Furthermore, in combination with DC vaccination (DC + Lip-peptide+Lip-CLX), it significantly increased tumor-infiltrating CD4+ and CD8+ T cells and secretion of IFN-γ. This combinatorial strategy produced an effective prophylactic and therapeutic antitumor response, which reduced tumor growth and prolonged the overall survival. In conclusion, our findings suggest that the liposomal celecoxib targets the inhibitory mechanisms of the tumor microenvironment and broadens the impact of DC therapy to improve the outcome of immunotherapy in solid tumors.
Collapse
Affiliation(s)
- Vajiheh Jahani
- School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mona Yazdani
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Ali Badiee
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmaceutical Nanotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mahmoud Reza Jaafari
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmaceutical Nanotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran; Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Leila Arabi
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmaceutical Nanotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
6
|
Wang MS, Wang ZZ, Li ZL, Gong Y, Duan CX, Cheng QH, Huang W, Yang GF. Discovery of Macrocycle-Based HPK1 Inhibitors for T-Cell-Based Immunotherapy. J Med Chem 2023; 66:611-626. [PMID: 36542759 DOI: 10.1021/acs.jmedchem.2c01551] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Hematopoietic progenitor kinase 1 (HPK1) is a negative regulator of T-cell activation, and targeting HPK1 is considered a promising strategy for improving responses to antitumor immune therapies. The biggest challenge of HPK1 inhibitor design is to achieve a higher selectivity to GLK, an HPK1 homology protein as a positive regulator of T-cell activation. Herein, we report the design of a series of macrocycle-based HPK1 inhibitors via a conformational constraint strategy. The identified candidate compound 5i exhibited HPK1 inhibition with an IC50 value of 0.8 nM and 101.3-fold selectivity against GLK. Compound 5i also displayed good oral bioavailability (F = 27-49%) in mice and beagles and favorable metabolic stability (T1/2 > 186.4 min) in human liver microsomes. More importantly, compound 5i demonstrated a clear synergistic effect with anti-PD-1 in both MC38 (MSI) and CT26 (MSS) syngeneic tumor mouse models. These results showed that compound 5i has a great potential in immunotherapy.
Collapse
Affiliation(s)
- Ming-Shu Wang
- Key Laboratory of Pesticide & Chemical Biology of Ministry of Education, International Joint Research Center for Intelligent Biosensor Technology and Health, College of Chemistry, Central China Normal University, Wuhan 430079, P. R. of China
| | - Zhi-Zheng Wang
- Key Laboratory of Pesticide & Chemical Biology of Ministry of Education, International Joint Research Center for Intelligent Biosensor Technology and Health, College of Chemistry, Central China Normal University, Wuhan 430079, P. R. of China
| | - Zi-Long Li
- Key Laboratory of Pesticide & Chemical Biology of Ministry of Education, International Joint Research Center for Intelligent Biosensor Technology and Health, College of Chemistry, Central China Normal University, Wuhan 430079, P. R. of China
| | - Yi Gong
- Key Laboratory of Pesticide & Chemical Biology of Ministry of Education, International Joint Research Center for Intelligent Biosensor Technology and Health, College of Chemistry, Central China Normal University, Wuhan 430079, P. R. of China
| | - Cheng-Xiang Duan
- Key Laboratory of Pesticide & Chemical Biology of Ministry of Education, International Joint Research Center for Intelligent Biosensor Technology and Health, College of Chemistry, Central China Normal University, Wuhan 430079, P. R. of China
| | - Qian-Hui Cheng
- Key Laboratory of Pesticide & Chemical Biology of Ministry of Education, International Joint Research Center for Intelligent Biosensor Technology and Health, College of Chemistry, Central China Normal University, Wuhan 430079, P. R. of China
| | - Wei Huang
- Key Laboratory of Pesticide & Chemical Biology of Ministry of Education, International Joint Research Center for Intelligent Biosensor Technology and Health, College of Chemistry, Central China Normal University, Wuhan 430079, P. R. of China
| | - Guang-Fu Yang
- Key Laboratory of Pesticide & Chemical Biology of Ministry of Education, International Joint Research Center for Intelligent Biosensor Technology and Health, College of Chemistry, Central China Normal University, Wuhan 430079, P. R. of China
| |
Collapse
|
7
|
Liu S, Liu T, Jiang J, Guo H, Yang R. p53 mutation and deletion contribute to tumor immune evasion. Front Genet 2023; 14:1088455. [PMID: 36891151 PMCID: PMC9986462 DOI: 10.3389/fgene.2023.1088455] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Accepted: 01/11/2023] [Indexed: 02/22/2023] Open
Abstract
TP53 (or p53) is widely accepted to be a tumor suppressor. Upon various cellular stresses, p53 mediates cell cycle arrest and apoptosis to maintain genomic stability. p53 is also discovered to suppress tumor growth through regulating metabolism and ferroptosis. However, p53 is always lost or mutated in human and the loss or mutation of p53 is related to a high risk of tumors. Although the link between p53 and cancer has been well established, how the different p53 status of tumor cells help themselves evade immune response remains largely elusive. Understanding the molecular mechanisms of different status of p53 and tumor immune evasion can help optimize the currently used therapies. In this context, we discussed the how the antigen presentation and tumor antigen expression mode altered and described how the tumor cells shape a suppressive tumor immune microenvironment to facilitate its proliferation and metastasis.
Collapse
Affiliation(s)
- Siyang Liu
- Department of Urology, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China
| | - Tianyao Liu
- Department of Urology, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China
| | - Jiaxuan Jiang
- Department of Endocrinology, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China
| | - Hongqian Guo
- Department of Urology, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China
| | - Rong Yang
- Department of Urology, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China
| |
Collapse
|
8
|
Hwang M, Chia YL, Zheng Y, Chen CCK, He J, Song X, Zhou D, Goldberg SB, Siu LL, Planchard D, Peters S, Mann H, Krug L, Even C. Population pharmacokinetic modelling of tremelimumab in patients with advanced solid tumours and the impact of disease status on time-varying clearance. Br J Clin Pharmacol 2022; 89:1601-1616. [PMID: 36454221 DOI: 10.1111/bcp.15622] [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: 07/01/2022] [Revised: 10/24/2022] [Accepted: 11/22/2022] [Indexed: 12/03/2022] Open
Abstract
AIMS Tremelimumab, a cytotoxic T-lymphocyte-associated protein 4 human monoclonal antibody of the immunoglobulin G2 κ isotype, has been studied in oncology clinical trials as both monotherapy and in combination with durvalumab. This study characterized the pharmacokinetics of tremelimumab as monotherapy and in combination with durvalumab and evaluated the impact of patient covariates on pharmacokinetics. METHODS A pooled-analysis population pharmacokinetics model was built using NONMEM methodology. Pharmacokinetic data from 5 studies spanning different tumour types and therapy regimens were pooled for model development (956 patients). A dataset pooled from 4 additional studies was used for external validation (554 patients). Demographic and relevant clinical covariates were explored during model development. RESULTS Tremelimumab exhibited linear pharmacokinetics, well described by a 2-compartment model, with time-varying clearance (0.276 L/day at baseline) associated primarily with therapy regimen and linked with changes in disease status. As monotherapy and combination therapy, tremelimumab clearance over 1 year increased by ~16% and decreased by ~17%, respectively. Pharmacokinetic behaviour was consistent across patient demographics and cancer subtypes. Patients with higher bodyweight and lower albumin levels at baseline had significantly higher clearance; however, no dosage adjustments are warranted. A flat dose (75 mg) was projected to provide comparable exposure to weight-based dosing (1 mg/kg) in adults. CONCLUSION Tremelimumab exhibited linear pharmacokinetics but consistently opposite trends of time-varying clearance as monotherapy and in combination with durvalumab. Baseline bodyweight and albumin were significant covariates, but conversion from weight-based dosing at 1 mg/kg to flat dosing at 75 mg had no clinically relevant impact.
Collapse
Affiliation(s)
- Michael Hwang
- Clinical Pharmacology and Quantitative Pharmacology, Biopharmaceuticals R&D, AstraZeneca, South San Francisco, CA, USA
| | - Yen Lin Chia
- Clinical Pharmacology and Quantitative Pharmacology, Biopharmaceuticals R&D, AstraZeneca, South San Francisco, CA, USA
| | - Yanan Zheng
- Clinical Pharmacology and Quantitative Pharmacology, Biopharmaceuticals R&D, AstraZeneca, South San Francisco, CA, USA
| | - Cecil Chi-Keung Chen
- Clinical Pharmacology and Quantitative Pharmacology, Biopharmaceuticals R&D, AstraZeneca, South San Francisco, CA, USA
| | - Jimmy He
- Clinical Pharmacology and Quantitative Pharmacology, Biopharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Xuyang Song
- Clinical Pharmacology and Quantitative Pharmacology, Biopharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Diansong Zhou
- Clinical Pharmacology and Quantitative Pharmacology, Biopharmaceuticals R&D, AstraZeneca, Boston, MA, USA
| | - Sarah B Goldberg
- Department of Medicine (Section of Medical Oncology), Yale School of Medicine, New Haven, CT, USA
| | - Lillian L Siu
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - David Planchard
- Department of Medical Oncology, Thoracic Unit, Institut Gustave-Roussy, Villejuif, France
| | - Solange Peters
- Department of Oncology CHUV-UNIL, University Hospital Lausanne, Lausanne, Switzerland
| | - Helen Mann
- Oncology Biometrics, Oncology R&D, AstraZeneca, Cambridge, UK
| | - Lee Krug
- Late Development Oncology R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Caroline Even
- Head and Neck Department, Institut Gustave-Roussy, Villejuif, France
| |
Collapse
|
9
|
Ramiscal JAB, Stern SL, Wilson AK, Lorimer PD, Lee NA, Goldfarb MR, Foshag LJ, Fischer TD. Does Residual Invasive Disease in Wide Local Excision after Diagnosis with Partial Biopsy Technique Influence Survival in Melanoma? Matched-Pair Analysis of Multicenter Selective Lymphadenectomy Trial I and II. J Am Coll Surg 2022; 235:49-59. [PMID: 35703962 DOI: 10.1097/xcs.0000000000000263] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
BACKGROUND Current guidelines recommend excisional/complete biopsy for melanoma diagnosis, owing to high rates of residual disease found at wide local excision (WLE) after partial biopsy techniques. We sought to determine any survival disadvantage associated with the presence of residual invasive melanoma in the WLE after diagnosis with a partial biopsy technique. STUDY DESIGN Data were examined from Multicenter Selective Lymphadenectomy Trials I and II (MSLT-I and -II), 2 large melanoma trials. Patients diagnosed with excisional/complete biopsy were excluded. Clinicopathologic characteristics, melanoma-specific survival (MSS), distant disease-free survival (DDFS), and disease-free survival (DFS) of those with residual invasive melanoma in the definitive WLE and those with no residual melanoma were compared. Matched pairing was used to reduce variability between groups. RESULTS From 1994 through 2014, 3,939 patients were enrolled in these trials and 874 (22%) were diagnosed using partial biopsy techniques. Of these, 399 (46%) had residual tumor in the WLE. Only 6 patients had residual tumor in their WLE resulting in T-upstaging of their tumor. Match-pairing formed two cohorts (1:1) of patients with and without residual invasive tumor after WLE. A total of 514 patients were paired; 288 (56%) males, 148 (28.8%) aged 60 or older, 192 (37.4%) with truncal melanomas, 214 (41.6%) had Breslow thickness 2 mm or greater, and 376 (73.2%) had positive sentinel nodes. Kaplan-Meier analysis showed no statistical difference in 10-year MSS (73.6% ± 3.3% vs 73.9% ± 3.7%, p = 0.891), DDFS (68.7% ± 3.4% vs 65.3% ± 4.0%, p = 0.548), or DFS (59.6% ± 3.7% vs 59.4% ± 3.9%, p = 0.783). CONCLUSIONS Survival in patients with primary melanoma does not appear to be worse in patients who undergo a partial biopsy technique and are later found to have residual invasive tumor in the WLE specimen.
Collapse
Affiliation(s)
- Judi Anne B Ramiscal
- From Saint John's Cancer Institute at Providence Saint John's Health Center, Santa Monica, CA
| | | | | | | | | | | | | | | |
Collapse
|
10
|
Wang L, Hu D, Xie B, Xie L. Blockade of Myd88 signaling by a novel MyD88 inhibitor prevents colitis-associated colorectal cancer development by impairing myeloid-derived suppressor cells. Invest New Drugs 2022; 40:506-518. [PMID: 35089465 PMCID: PMC9098617 DOI: 10.1007/s10637-022-01218-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Accepted: 01/21/2022] [Indexed: 01/04/2023]
Abstract
Background. In cancer, myeloid-derived suppressor cells (MDSCs) are known to escape the host immune system by developing a highly suppressive environment. However, little is known about the molecular mechanism behind MDSC-mediated tumor cell evasion of the immune system. Toll-like receptor (TLR) signaling elicited in the tumor microenvironment has the potential to induce MDSC differentiations in different organs. Therefore, MDSC elimination by blocking the action of myeloid differentiation factor 88 (MyD88), which is a key adaptor-signaling molecule that affects TLR activity, seems to be an ideal tumor immunotherapy. Previous studies have proven that blocking MyD88 signaling with a novel MyD88 inhibitor (TJ-M2010-5, synthesized by Zhou’s group) completely prevented colitis-associated colorectal cancer (CAC) development in mice. Methods. In the present study, we investigated the impact of the novel MyD88 inhibitor on the number, phenotype, and function of MDSC in the mice model of CAC. Results. We showed that CAC growth inhibition was involved in diminished MDSC generation, expansion, and suppressive function and that MDSC-mediated immune escape was dependent on MyD88 signaling pathway activation. MyD88 inhibitor treatment decreased the accumulation of CD11b+Gr1+ MDSCs in mice with CAC, thereby reducing cytokine (GM-CSF, G-CSF, IL-1β, IL-6 and TGF-β) secretion associated with MDSC accumulation, and reducing the expression of molecules (iNOS, Arg-1 and IDO) associated with the suppressive capacity of MDSCs. In addition, MyD88 inhibitor treatment reduced the differentiation of MDSCs from myeloid cells and the suppressive capacity of MDSCs on the proliferation of activated CD4+ T cells in vitro. Conclusion. MDSCs are primary cellular targets of a novel MyD88 inhibitor during CAC development. Our findings prove that MyD88 signaling is involved in the regulation of the immunosuppressive functions of MDSCs. The novel MyD88 inhibitor TJ-M2010-5 is a new and effective agent that modulates MyD88 signaling to overcome MDSC suppressive functions, enabling the development of successful antitumor immunotherapy.
Collapse
Affiliation(s)
- Lu Wang
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology; Key Laboratory of Organ Transplantation, Ministry of Education, China; NHC Key Laboratory of Organ Transplantation, China; Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, China
| | - Dan Hu
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Bin Xie
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology; Key Laboratory of Organ Transplantation, Ministry of Education, China; NHC Key Laboratory of Organ Transplantation, China; Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, China
| | - Lin Xie
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology; Key Laboratory of Organ Transplantation, Ministry of Education, China; NHC Key Laboratory of Organ Transplantation, China; Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, China.
| |
Collapse
|
11
|
Newcomer K, Robbins KJ, Perone J, Hinojosa FL, Chen D, Jones S, Kaufman CK, Weiser R, Fields RC, Tyler DS. Malignant melanoma: evolving practice management in an era of increasingly effective systemic therapies. Curr Probl Surg 2022; 59:101030. [PMID: 35033317 PMCID: PMC9798450 DOI: 10.1016/j.cpsurg.2021.101030] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Accepted: 05/12/2021] [Indexed: 01/03/2023]
Affiliation(s)
- Ken Newcomer
- Department of Surgery, Barnes-Jewish Hospital, Washington University, St. Louis, MO
| | | | - Jennifer Perone
- Department of Surgery, University of Texas Medical Branch, Galveston, TX
| | | | - David Chen
- e. Department of Medicine, Washington University, St. Louis, MO
| | - Susan Jones
- f. Department of Pediatrics, Washington University, St. Louis, MO
| | | | - Roi Weiser
- University of Texas Medical Branch, Galveston, TX
| | - Ryan C Fields
- Department of Surgery, Washington University, St. Louis, MO
| | - Douglas S Tyler
- Department of Surgery, University of Texas Medical Branch, Galveston, TX.
| |
Collapse
|
12
|
Abstract
Melanoma is the most lethal skin cancer that originates from the malignant transformation of melanocytes. Although melanoma has long been regarded as a cancerous malignancy with few therapeutic options, increased biological understanding and unprecedented innovations in therapies targeting mutated driver genes and immune checkpoints have substantially improved the prognosis of patients. However, the low response rate and inevitable occurrence of resistance to currently available targeted therapies have posed the obstacle in the path of melanoma management to obtain further amelioration. Therefore, it is necessary to understand the mechanisms underlying melanoma pathogenesis more comprehensively, which might lead to more substantial progress in therapeutic approaches and expand clinical options for melanoma therapy. In this review, we firstly make a brief introduction to melanoma epidemiology, clinical subtypes, risk factors, and current therapies. Then, the signal pathways orchestrating melanoma pathogenesis, including genetic mutations, key transcriptional regulators, epigenetic dysregulations, metabolic reprogramming, crucial metastasis-related signals, tumor-promoting inflammatory pathways, and pro-angiogenic factors, have been systemically reviewed and discussed. Subsequently, we outline current progresses in therapies targeting mutated driver genes and immune checkpoints, as well as the mechanisms underlying the treatment resistance. Finally, the prospects and challenges in the development of melanoma therapy, especially immunotherapy and related ongoing clinical trials, are summarized and discussed.
Collapse
Affiliation(s)
- Weinan Guo
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, No. 127 of West Changle Road, 710032, Xi'an, Shaanxi, China
| | - Huina Wang
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, No. 127 of West Changle Road, 710032, Xi'an, Shaanxi, China
| | - Chunying Li
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, No. 127 of West Changle Road, 710032, Xi'an, Shaanxi, China.
| |
Collapse
|
13
|
Meng J, Huang X, Qiu Y, Yu M, Lu J, Yao J. Characterization of m6A-Related Genes Landscape in Skin Cutaneous Melanoma to Aid Immunotherapy and Assess Prognosis. Int J Gen Med 2021; 14:5345-5361. [PMID: 34522131 PMCID: PMC8434882 DOI: 10.2147/ijgm.s328522] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Accepted: 08/24/2021] [Indexed: 01/21/2023] Open
Abstract
Background Skin cutaneous melanoma (SKCM) is the most malignant tumor among skin cancers. Immunotherapy has shown a great role in the advantageous prognosis of SKCM. However, only a small percentage of people can benefit from immunotherapy. To date, there has been insufficient evidence to reveal the prognostic value of m6A in SKCM and its relationship with the infiltration of immune cells and the efficacy of immunotherapy. Methods Here, we synthetically analyzed 23 m6A regulators from SKCM samples collected from the TCGA and GEO databases. We defined three m6A modification patterns and constructed m6A scores using principal component analysis (PCA). Results We found significant differences in overall survival (OS) and immune infiltration between different m6A subclusters. Besides, m6A score was positively correlated with regulatory T-cell and helper T-cell content, which may account for the association of high m6A scores with superior prognosis. Multivariate Cox regression analysis revealed that the m6A score was an independent prognostic indicator. Moreover, patients with high m6A scores showed a better response to immunotherapy, and this result was further validated in two independent immunotherapy cohorts receiving anti-PD-1/PD-L1 therapy. Conclusion The findings suggested the m6A score can screen suitable candidates for immunotherapy and can predict immunotherapy response. This analysis of different m6A patterns in a large sample of SKCM expanded our understanding of TME and provided new ideas for prognostic assessment and personalized immunotherapy strategies for SKCM patients.
Collapse
Affiliation(s)
- Jinzhi Meng
- Bone and Joint Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, People's Republic of China
| | - Xing Huang
- Bone and Joint Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, People's Republic of China
| | - Yue Qiu
- Bone and Joint Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, People's Republic of China
| | - Miao Yu
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Nanning, People's Republic of China
| | - Jinfeng Lu
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Nanning, People's Republic of China
| | - Jun Yao
- Bone and Joint Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, People's Republic of China
| |
Collapse
|
14
|
Pang B, Hao Y. Integrated Analysis of the Transcriptome Profile Reveals the Potential Roles Played by Long Noncoding RNAs in Immunotherapy for Sarcoma. Front Oncol 2021; 11:690486. [PMID: 34178688 PMCID: PMC8226247 DOI: 10.3389/fonc.2021.690486] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2021] [Accepted: 04/29/2021] [Indexed: 12/17/2022] Open
Abstract
Background Long-term survival is still low for high-risk patients with soft tissue sarcoma treated with standard management options, including surgery, radiation, and chemotherapy. Immunotherapy is a promising new potential treatment paradigm. However, the application of immune checkpoint inhibitors for the treatment of patients with sarcoma did not yield promising results in a clinical trial. Therefore, there is a considerable need to identify factors that may lead to immune checkpoint inhibitor resistance. Methods In this study, we performed a bioinformatic analysis of The Cancer Genome Atlas (TCGA) to detect key long noncoding RNAs (lncRNAs) that were correlated with immune checkpoint inhibitory molecules in sarcoma. The expression levels of these lncRNAs and their correlation with patient prognosis were explored. The upstream long noncoding RNAs were also examined via 450K array data from the TCGA. The potential roles of these lncRNAs were further examined via KEGG and GO analysis using DAVID online software. Finally, the relationship between these lncRNAs and immune cell infiltration in tumors and their effect on immune checkpoint inhibitors were further explored. Results We identified lncRNAs correlated with tumor cell immune evasion in sarcoma. The expression of these lncRNAs was upregulated and correlated with worse prognosis in sarcoma and other human cancer types. Moreover, low DNA methylation occupation of these lncRNA loci was detected. Negative correlations between DNA methylation and lncRNA expression were also found in sarcoma and other human cancer types. KEGG and GO analyses indicated that these lncRNAs correlated with immune evasion and negative regulation of the immune response in sarcoma. Finally, high expression of these lncRNAs correlated with more suppressive immune cell infiltration and reduced sensitivity to immune checkpoint inhibitors in sarcoma and other human cancer types. Conclusion Our results suggest that long noncoding RNAs confer immune checkpoint inhibitor resistance in human cancer. Further characterization of these lncRNAs may help to elucidate the mechanisms underlying immune checkpoint inhibitor resistance and uncover a novel therapeutic intervention point for immunotherapy.
Collapse
Affiliation(s)
- Boran Pang
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Clinical and Translational Research Center for 3D Printing Technology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yongqiang Hao
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Clinical and Translational Research Center for 3D Printing Technology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
15
|
Yang J, Zhao S, Wang J, Sheng Q, Liu Q, Shyr Y. Immu-Mela: An open resource for exploring immunotherapy-related multidimensional genomic profiles in melanoma. J Genet Genomics 2021; 48:361-368. [PMID: 34127402 PMCID: PMC8349898 DOI: 10.1016/j.jgg.2021.03.016] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 03/15/2021] [Accepted: 03/17/2021] [Indexed: 11/22/2022]
Abstract
There are increasing studies aimed to reveal genomic hallmarks predictive of immune checkpoint blockade (ICB) treatment response, which generated a large number of data and provided an unprecedented opportunity to identify response-related features and evaluate their robustness across cohorts. However, those valuable data sets are not easily accessible to the research community. To take full advantage of existing large-scale immuno-genomic profiles, we developed Immu-Mela (http://bioinfo.vanderbilt.edu/database/Immu-Mela/), a multidimensional immuno-genomic portal that provides interactive exploration of associations between ICB responsiveness and multi-omics features in melanoma, including genetic, transcriptomics, immune cells, and single-cell populations. Immu-Mela also enables integrative analysis of any two genomic features. We demonstrated the value of Immu-Mela by identifying known and novel genomic features associated with ICB response. In addition, Immu-Mela allows users to upload their data sets (unrestricted to any cancer types) and co-analyze with existing data to identify and validate signatures of interest. Immu-Mela reduces barriers between researchers and complex genomic data, facilitating discoveries in cancer immunotherapy.
Collapse
Affiliation(s)
- Jing Yang
- Center for Quantitative Sciences, Vanderbilt University Medical Center, Nashville TN 37203, USA; Department of Biostatistics, Vanderbilt University Medical Center, Nashville TN 37203, USA
| | - Shilin Zhao
- Center for Quantitative Sciences, Vanderbilt University Medical Center, Nashville TN 37203, USA; Department of Biostatistics, Vanderbilt University Medical Center, Nashville TN 37203, USA
| | - Jing Wang
- Center for Quantitative Sciences, Vanderbilt University Medical Center, Nashville TN 37203, USA; Department of Biostatistics, Vanderbilt University Medical Center, Nashville TN 37203, USA
| | - Quanhu Sheng
- Center for Quantitative Sciences, Vanderbilt University Medical Center, Nashville TN 37203, USA; Department of Biostatistics, Vanderbilt University Medical Center, Nashville TN 37203, USA
| | - Qi Liu
- Center for Quantitative Sciences, Vanderbilt University Medical Center, Nashville TN 37203, USA; Department of Biostatistics, Vanderbilt University Medical Center, Nashville TN 37203, USA.
| | - Yu Shyr
- Center for Quantitative Sciences, Vanderbilt University Medical Center, Nashville TN 37203, USA; Department of Biostatistics, Vanderbilt University Medical Center, Nashville TN 37203, USA.
| |
Collapse
|
16
|
Klarquist JS, Janssen EM. Melanoma-infiltrating dendritic cells: Limitations and opportunities of mouse models. Oncoimmunology 2021; 1:1584-1593. [PMID: 23264904 PMCID: PMC3525613 DOI: 10.4161/onci.22660] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
The infiltration of melanoma lesions by dendritic cells (DCs) has been suggested to play a tumorigenic role due to the capacity of DCs to induce tumor tolerance and promote angiogenesis as well as metastasis. However, it has also been shown that tumor-infiltrating DCs (TIDCs) induce antitumor responses and hence may be targeted in cost-effective therapeutic approaches to obtain patient-specific DCs that present relevant tumor antigens, without the need for ex vivo DC expansion or tumor antigen identification. Unfortunately, little is known about the composition, nature and function of TIDCs found in human melanoma. The development of mouse melanoma models has greatly contributed to the molecular understanding of melanoma immunology in mice, but many questions on TIDCs remain unanswered. Here, we discuss current knowledge about melanoma TIDCs in various mouse models with regard to their translational potential and clinical relevance.
Collapse
Affiliation(s)
- Jared S Klarquist
- Division of Cellular and Molecular Immunology; Cincinnati Children's Hospital Research Foundation; University of Cincinnati College of Medicine; Cincinnati, OH USA
| | | |
Collapse
|
17
|
Patinote C, Deleuze-Masquéfa C, Kaddour KH, Vincent LA, Larive R, Zghaib Z, Guichou JF, Assaf MD, Cuq P, Bonnet PA. Imidazo[1,2-a]quinoxalines for melanoma treatment with original mechanism of action. Eur J Med Chem 2020; 212:113031. [PMID: 33309473 DOI: 10.1016/j.ejmech.2020.113031] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Revised: 11/05/2020] [Accepted: 11/14/2020] [Indexed: 10/22/2022]
Abstract
The malignant transformation of melanocytes causes several thousand deaths each year, making melanoma an important public health concern. Melanoma is the most aggressive skin cancer, which incidence has regularly increased over the past decades. We described here the preparation of new compounds based on the 1-(3,4-dihydroxyphenyl)imidazo[1,2-a]quinoxaline structure. Different positions of the quinoxaline moiety were screened to introduce novel substituents in order to study their influence on the biological activity. Several alkylamino or alkyloxy groups were also considered to replace the methylamine of our first generation of Imiqualines. Imidazo[1,2-a]pyrazine derivatives were also designed as potential minimal structure. The investigation on A375 melanoma cells displayed interesting in vitro low nanomolar cytotoxic activity. Among them, 9d (EAPB02303) is particularly remarkable since it is 20 times more potent than vemurafenib, the reference clinical therapy used on BRAF mutant melanoma. Contrary to the first generation, EAPB02303 does not inhibit tubulin polymerization, as confirmed by an in vitro assay and a molecular modelisation study. The mechanism of action for EAPB02303 highlighted by a transcriptomic analysis is clearly different from a panel of 12 well-known anticancer drugs. In vivoEAPB02303 treatment reduced tumor size and weight of the A375 human melanoma xenografts in a dose-dependent manner, correlated with a low mitotic index but not with necrosis.
Collapse
Affiliation(s)
- Cindy Patinote
- Institut des Biomolécules Max Mousseron (IBMM), UMR 5247, CNRS, Université de Montpellier, Faculté de Pharmacie, 15 avenue Charles Flahault, BP 14491, 34093, Montpellier Cedex 5, France; Société d'Accélération du Transfert de Technologies (SATT AxLR), CSU, 950 rue Saint Priest, 34090, Montpellier, France
| | - Carine Deleuze-Masquéfa
- Institut des Biomolécules Max Mousseron (IBMM), UMR 5247, CNRS, Université de Montpellier, Faculté de Pharmacie, 15 avenue Charles Flahault, BP 14491, 34093, Montpellier Cedex 5, France.
| | - Kamel Hadj Kaddour
- Institut des Biomolécules Max Mousseron (IBMM), UMR 5247, CNRS, Université de Montpellier, Faculté de Pharmacie, 15 avenue Charles Flahault, BP 14491, 34093, Montpellier Cedex 5, France
| | - Laure-Anaïs Vincent
- Institut des Biomolécules Max Mousseron (IBMM), UMR 5247, CNRS, Université de Montpellier, Faculté de Pharmacie, 15 avenue Charles Flahault, BP 14491, 34093, Montpellier Cedex 5, France
| | - Romain Larive
- Institut des Biomolécules Max Mousseron (IBMM), UMR 5247, CNRS, Université de Montpellier, Faculté de Pharmacie, 15 avenue Charles Flahault, BP 14491, 34093, Montpellier Cedex 5, France
| | - Zahraa Zghaib
- Institut des Biomolécules Max Mousseron (IBMM), UMR 5247, CNRS, Université de Montpellier, Faculté de Pharmacie, 15 avenue Charles Flahault, BP 14491, 34093, Montpellier Cedex 5, France; Tumorigenèse et Pharmacologie Antitumorale, Lebanese University, EDST, BP 90656, Fanar Jdeideh, Lebanon
| | - Jean-François Guichou
- CNRS, UMR 5048, INSERM, U105, Université de Montpellier, Centre de Biochimie Structurale, 34090, Montpellier, France
| | - Mona Diab Assaf
- Tumorigenèse et Pharmacologie Antitumorale, Lebanese University, EDST, BP 90656, Fanar Jdeideh, Lebanon
| | - Pierre Cuq
- Institut des Biomolécules Max Mousseron (IBMM), UMR 5247, CNRS, Université de Montpellier, Faculté de Pharmacie, 15 avenue Charles Flahault, BP 14491, 34093, Montpellier Cedex 5, France
| | - Pierre-Antoine Bonnet
- Institut des Biomolécules Max Mousseron (IBMM), UMR 5247, CNRS, Université de Montpellier, Faculté de Pharmacie, 15 avenue Charles Flahault, BP 14491, 34093, Montpellier Cedex 5, France
| |
Collapse
|
18
|
Bozcuk H, Artaç M, Mutlu H, Sever Ö, Yıldırım M. Programmed death-1 or programmed death ligand-1 inhibitors? A meta-analysis of differential efficacy as compared to chemotherapy in advanced non-small cell lung cancer. J Oncol Pharm Pract 2020; 27:405-413. [PMID: 33040676 DOI: 10.1177/1078155220964903] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Programmed Death-1 (PD-1) and Programmed Death Ligand-1 (PDL-1) inhibitors have improved survival over chemotherapy in advanced Non- Small Cell Lung Cancer (NSCLC). However, it is unclear if there are class specific differences in the efficacy of Checkpoint Inhibitors (CPIs) in NSCLC, and this paper is designed to answer these clinical questions. METHODS For this Meta-analysis, we searched PubMed, Science of Web, "Clinicaltrials.gov" and online sources for trials comparing PD-1 and PDL-1 CPIs in advanced NSCLC. The data for Hazard Ratio (HR) and their Confidence Intervals (CI) for Overall Survival (OS) was extracted. RESULTS A sum of 9739 patients from 16 trials were included in the efficacy evaluation. For the OS endpoint, both PD-1 inhibitors (HR = 0.76, 95%CI = 0.69-0.83, P < 0.001) and PDL-1 inhibitors (HR = 0.84, 95%CI = 0.74-0.95, P < 0.001) were superior to chemotherapy in treatment naïve (upfront) patients, the results were similar in treatment refractory patients (PD-1 inhibitors (HR = 0.67, 95%CI = 0.60-0.75, P < 0.001) and PDL-1 inhibitors (HR = 0.78, 95%CI = 0.69-0.88, P < 0.001) were superior to chemotherapy). There was no difference in the effect of PD-1 and PDL-1 classes of CPIs over chemotherapy in treatment naïve and treatment refractory settings (Q = 1.88, df = 1, P = 0.017, and, Q = 3.27, df = 1, P = 0.070, respectively). CONCLUSION Efficacy of PD-1 and PDL-1 class of CPIs was not different, although differences among individual CPIs or their combinations cannot be excluded. We were also able to compute pooled efficacy data, as compared to chemotherapy alone, for trials where these groups of CPIs were utilized.
Collapse
Affiliation(s)
- Hakan Bozcuk
- Department of Medical Oncology, Medical Park Hospital, Antalya, Turkey
| | - Mehmet Artaç
- Department of Medical Oncology, Necmettin Erbakan University, Konya, Turkey
| | - Hasan Mutlu
- Department of Medical Oncology, Medical Park Hospital, Antalya, Turkey
| | - Özlem Sever
- Department of Internal Medicine, Sanko University School of Medicine, Gaziantep, Turkey
| | - Mustafa Yıldırım
- Department of Medical Oncology, Medical Park Hospital, Gaziantep, Turkey
| |
Collapse
|
19
|
Yu F, Zhu C, Xie Q, Wang Y. Adenosine A 2A Receptor Antagonists for Cancer Immunotherapy. J Med Chem 2020; 63:12196-12212. [PMID: 32667814 DOI: 10.1021/acs.jmedchem.0c00237] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Currently, the most promising therapeutic modality for cancer treatment is the blockade of immune checkpoint pathways, which has revolutionized cancer therapy in the past 15 years. Strategies targeting and modulating adenosine A2A receptor (A2AR), an emerging alternative immune checkpoint, have shown the potential to produce significant therapeutic effects. In this review, we describe the immunosuppressive activities of A2AR and A2BR in the tumor microenvironment (TME), followed by a summary and discussion of the structure-activity relationship (SAR) of the A2AR (and dual A2AR/A2BR) antagonists that have been experimentally confirmed to exert oncoimmunological effects. This review also provides an update on the compounds under clinical evaluation and insights into the ligand binding modes of the receptor.
Collapse
Affiliation(s)
- Fazhi Yu
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, 826 Zhangheng Road, Shanghai 201203, China
| | - Chenyu Zhu
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, 826 Zhangheng Road, Shanghai 201203, China
| | - Qiong Xie
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, 826 Zhangheng Road, Shanghai 201203, China
| | - Yonghui Wang
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, 826 Zhangheng Road, Shanghai 201203, China
| |
Collapse
|
20
|
Huang Y, Zeng J. Recent development and applications of nanomaterials for cancer
immunotherapy. NANOTECHNOLOGY REVIEWS 2020; 9:367-384. [DOI: 10.1515/ntrev-2020-0027] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/30/2023]
Abstract
Abstract
Immunotherapy, which utilizes the patient’s own immune system to fight against
cancer, further results in durable antitumor responses and reduces metastasis and
recurrence, has become one of the most effective and important cancer therapies along
with surgery, radiotherapy, and chemotherapy. Nanomaterials with the advantages of
large specific surface, delivery function, and controllable surface chemistry are
used to deliver antigens or adjuvants, or both, help to boost immune responses with
the imaging function or just act as adjuvants themselves and modulate tumor
microenvironment (TME). In this review, recent development and applications of
nanomaterials for cancer immunotherapy including delivery systems based on
nanomaterials, uniting imaging, self-adjuvants, targeting functions, artificial
antigen presenting cells, and TME modulation are focused and discussed.
Collapse
Affiliation(s)
- Yao Huang
- Liver Disease Center, The First Affiliated Hospital of Fujian Medical University, Fuzhou 350005 , China
| | - Jinhua Zeng
- Liver Disease Center, The First Affiliated Hospital of Fujian Medical University, Fuzhou 350005 , China
| |
Collapse
|
21
|
Ramesh A, Natarajan SK, Nandi D, Kulkarni A. Dual Inhibitors-Loaded Nanotherapeutics that Target Kinase Signaling Pathways Synergize with Immune Checkpoint Inhibitor. Cell Mol Bioeng 2019; 12:357-373. [PMID: 31719920 DOI: 10.1007/s12195-019-00576-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Accepted: 05/15/2019] [Indexed: 12/21/2022] Open
Abstract
Introduction Immune checkpoint inhibitors that boost cytotoxic T cell-based immune responses have emerged as one of the most promising approaches in cancer treatment. However, it is increasingly being realized that T cell activation needs to be rationally combined with molecularly targeted therapeutics for a maximal anti-tumor outcome. Currently, two oncogenic drivers, MAPK and PI3K-mTOR have emerged as the two main molecular targets for combining with immunotherapy. However, there are major challenges in enabling such combinations: first, such combinations can result in high rates of toxicity. Second, while, these molecular targets could be driving tumor progression, they are essential for activation of the immune cells. So, the kinase inhibitors and immunotherapy can antagonize each other. Objectives We rationalized that the synergistic combination of kinase inhibitors and immunotherapy could be enabled by dual inhibitors-loaded supramolecular nanotherapeutics (DiLN) that can co-deliver PI3K- and MAPK-inhibitors to the cancer cells and activate immune response by T cell-modulating immunotherapy, resulting in greater anti-tumor efficacy while minimizing toxicity. Methods We engineered DiLNs by designing the amphiphilic building blocks (both drugs and co-lipids) that enables supramolecular nanoassembly. DiLNs were tested for their physiochemical properties including size, morphology, stability and drug release kinetics profiles. The efficacy of DiLNs was tested in drug-resistant cells such as BRAFV600E melanoma (D4M), Clear cell ovarian carcinoma (TOV21G) cells. The tumor inhibition efficiency of DiLNs in combination with immune checkpoint inhibitor antibody was studied in syngeneic D4M animal model. Results DiLNs were stable for over a month and released the drugs in a sustained manner. In vitro cytotoxicity studies in D4M and TOV21G cells showed that DiLNs were significantly more effective than free drugs. In vivo studies showed that the combination of DiLNs with anti PD-L1 antibody resulted in superior antitumor effect and survival. Conclusion This study shows that the rational combination of DiLNs that target multiple oncogenic signaling pathways with immune checkpoint inhibitors could emerge as an effective strategy to improve immunotherapeutic response against drug resistant tumors.
Collapse
Affiliation(s)
- Anujan Ramesh
- Department of Chemical Engineering, University of Massachusetts, Amherst, MA USA
| | - Siva Kumar Natarajan
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA USA
| | - Dipika Nandi
- Department of Veterinary and Animal Science, University of Massachusetts, Amherst, MA USA
| | - Ashish Kulkarni
- Department of Chemical Engineering, University of Massachusetts, Amherst, MA USA.,Department of Veterinary and Animal Science, University of Massachusetts, Amherst, MA USA.,Center for Bioactive Delivery, Institute for Applied Life Sciences, University of Massachusetts, Amherst, MA USA.,Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA USA
| |
Collapse
|
22
|
Efficient co-delivery of neo-epitopes using dispersion-stable layered double hydroxide nanoparticles for enhanced melanoma immunotherapy. Biomaterials 2018; 174:54-66. [DOI: 10.1016/j.biomaterials.2018.05.015] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Revised: 04/30/2018] [Accepted: 05/10/2018] [Indexed: 12/29/2022]
|
23
|
Pasquali S, Hadjinicolaou AV, Chiarion Sileni V, Rossi CR, Mocellin S. Systemic treatments for metastatic cutaneous melanoma. Cochrane Database Syst Rev 2018; 2:CD011123. [PMID: 29405038 PMCID: PMC6491081 DOI: 10.1002/14651858.cd011123.pub2] [Citation(s) in RCA: 99] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
BACKGROUND The prognosis of people with metastatic cutaneous melanoma, a skin cancer, is generally poor. Recently, new classes of drugs (e.g. immune checkpoint inhibitors and small-molecule targeted drugs) have significantly improved patient prognosis, which has drastically changed the landscape of melanoma therapeutic management. This is an update of a Cochrane Review published in 2000. OBJECTIVES To assess the beneficial and harmful effects of systemic treatments for metastatic cutaneous melanoma. SEARCH METHODS We searched the following databases up to October 2017: the Cochrane Skin Group Specialised Register, CENTRAL, MEDLINE, Embase and LILACS. We also searched five trials registers and the ASCO database in February 2017, and checked the reference lists of included studies for further references to relevant randomised controlled trials (RCTs). SELECTION CRITERIA We considered RCTs of systemic therapies for people with unresectable lymph node metastasis and distant metastatic cutaneous melanoma compared to any other treatment. We checked the reference lists of selected articles to identify further references to relevant trials. DATA COLLECTION AND ANALYSIS Two review authors extracted data, and a third review author independently verified extracted data. We implemented a network meta-analysis approach to make indirect comparisons and rank treatments according to their effectiveness (as measured by the impact on survival) and harm (as measured by occurrence of high-grade toxicity). The same two review authors independently assessed the risk of bias of eligible studies according to Cochrane standards and assessed evidence quality based on the GRADE criteria. MAIN RESULTS We included 122 RCTs (28,561 participants). Of these, 83 RCTs, encompassing 21 different comparisons, were included in meta-analyses. Included participants were men and women with a mean age of 57.5 years who were recruited from hospital settings. Twenty-nine studies included people whose cancer had spread to their brains. Interventions were categorised into five groups: conventional chemotherapy (including single agent and polychemotherapy), biochemotherapy (combining chemotherapy with cytokines such as interleukin-2 and interferon-alpha), immune checkpoint inhibitors (such as anti-CTLA4 and anti-PD1 monoclonal antibodies), small-molecule targeted drugs used for melanomas with specific gene changes (such as BRAF inhibitors and MEK inhibitors), and other agents (such as anti-angiogenic drugs). Most interventions were compared with chemotherapy. In many cases, trials were sponsored by pharmaceutical companies producing the tested drug: this was especially true for new classes of drugs, such as immune checkpoint inhibitors and small-molecule targeted drugs.When compared to single agent chemotherapy, the combination of multiple chemotherapeutic agents (polychemotherapy) did not translate into significantly better survival (overall survival: HR 0.99, 95% CI 0.85 to 1.16, 6 studies, 594 participants; high-quality evidence; progression-free survival: HR 1.07, 95% CI 0.91 to 1.25, 5 studies, 398 participants; high-quality evidence. Those who received combined treatment are probably burdened by higher toxicity rates (RR 1.97, 95% CI 1.44 to 2.71, 3 studies, 390 participants; moderate-quality evidence). (We defined toxicity as the occurrence of grade 3 (G3) or higher adverse events according to the World Health Organization scale.)Compared to chemotherapy, biochemotherapy (chemotherapy combined with both interferon-alpha and interleukin-2) improved progression-free survival (HR 0.90, 95% CI 0.83 to 0.99, 6 studies, 964 participants; high-quality evidence), but did not significantly improve overall survival (HR 0.94, 95% CI 0.84 to 1.06, 7 studies, 1317 participants; high-quality evidence). Biochemotherapy had higher toxicity rates (RR 1.35, 95% CI 1.14 to 1.61, 2 studies, 631 participants; high-quality evidence).With regard to immune checkpoint inhibitors, anti-CTLA4 monoclonal antibodies plus chemotherapy probably increased the chance of progression-free survival compared to chemotherapy alone (HR 0.76, 95% CI 0.63 to 0.92, 1 study, 502 participants; moderate-quality evidence), but may not significantly improve overall survival (HR 0.81, 95% CI 0.65 to 1.01, 2 studies, 1157 participants; low-quality evidence). Compared to chemotherapy alone, anti-CTLA4 monoclonal antibodies is likely to be associated with higher toxicity rates (RR 1.69, 95% CI 1.19 to 2.42, 2 studies, 1142 participants; moderate-quality evidence).Compared to chemotherapy, anti-PD1 monoclonal antibodies (immune checkpoint inhibitors) improved overall survival (HR 0.42, 95% CI 0.37 to 0.48, 1 study, 418 participants; high-quality evidence) and probably improved progression-free survival (HR 0.49, 95% CI 0.39 to 0.61, 2 studies, 957 participants; moderate-quality evidence). Anti-PD1 monoclonal antibodies may also result in less toxicity than chemotherapy (RR 0.55, 95% CI 0.31 to 0.97, 3 studies, 1360 participants; low-quality evidence).Anti-PD1 monoclonal antibodies performed better than anti-CTLA4 monoclonal antibodies in terms of overall survival (HR 0.63, 95% CI 0.60 to 0.66, 1 study, 764 participants; high-quality evidence) and progression-free survival (HR 0.54, 95% CI 0.50 to 0.60, 2 studies, 1465 participants; high-quality evidence). Anti-PD1 monoclonal antibodies may result in better toxicity outcomes than anti-CTLA4 monoclonal antibodies (RR 0.70, 95% CI 0.54 to 0.91, 2 studies, 1465 participants; low-quality evidence).Compared to anti-CTLA4 monoclonal antibodies alone, the combination of anti-CTLA4 plus anti-PD1 monoclonal antibodies was associated with better progression-free survival (HR 0.40, 95% CI 0.35 to 0.46, 2 studies, 738 participants; high-quality evidence). There may be no significant difference in toxicity outcomes (RR 1.57, 95% CI 0.85 to 2.92, 2 studies, 764 participants; low-quality evidence) (no data for overall survival were available).The class of small-molecule targeted drugs, BRAF inhibitors (which are active exclusively against BRAF-mutated melanoma), performed better than chemotherapy in terms of overall survival (HR 0.40, 95% CI 0.28 to 0.57, 2 studies, 925 participants; high-quality evidence) and progression-free survival (HR 0.27, 95% CI 0.21 to 0.34, 2 studies, 925 participants; high-quality evidence), and there may be no significant difference in toxicity (RR 1.27, 95% CI 0.48 to 3.33, 2 studies, 408 participants; low-quality evidence).Compared to chemotherapy, MEK inhibitors (which are active exclusively against BRAF-mutated melanoma) may not significantly improve overall survival (HR 0.85, 95% CI 0.58 to 1.25, 3 studies, 496 participants; low-quality evidence), but they probably lead to better progression-free survival (HR 0.58, 95% CI 0.42 to 0.80, 3 studies, 496 participants; moderate-quality evidence). However, MEK inhibitors probably have higher toxicity rates (RR 1.61, 95% CI 1.08 to 2.41, 1 study, 91 participants; moderate-quality evidence).Compared to BRAF inhibitors, the combination of BRAF plus MEK inhibitors was associated with better overall survival (HR 0.70, 95% CI 0.59 to 0.82, 4 studies, 1784 participants; high-quality evidence). BRAF plus MEK inhibitors was also probably better in terms of progression-free survival (HR 0.56, 95% CI 0.44 to 0.71, 4 studies, 1784 participants; moderate-quality evidence), and there appears likely to be no significant difference in toxicity (RR 1.01, 95% CI 0.85 to 1.20, 4 studies, 1774 participants; moderate-quality evidence).Compared to chemotherapy, the combination of chemotherapy plus anti-angiogenic drugs was probably associated with better overall survival (HR 0.60, 95% CI 0.45 to 0.81; moderate-quality evidence) and progression-free survival (HR 0.69, 95% CI 0.52 to 0.92; moderate-quality evidence). There may be no difference in terms of toxicity (RR 0.68, 95% CI 0.09 to 5.32; low-quality evidence). All results for this comparison were based on 324 participants from 2 studies.Network meta-analysis focused on chemotherapy as the common comparator and currently approved treatments for which high- to moderate-quality evidence of efficacy (as represented by treatment effect on progression-free survival) was available (based on the above results) for: biochemotherapy (with both interferon-alpha and interleukin-2); anti-CTLA4 monoclonal antibodies; anti-PD1 monoclonal antibodies; anti-CTLA4 plus anti-PD1 monoclonal antibodies; BRAF inhibitors; MEK inhibitors, and BRAF plus MEK inhibitors. Analysis (which included 19 RCTs and 7632 participants) generated 21 indirect comparisons.The best evidence (moderate-quality evidence) for progression-free survival was found for the following indirect comparisons:• both combinations of immune checkpoint inhibitors (HR 0.30, 95% CI 0.17 to 0.51) and small-molecule targeted drugs (HR 0.17, 95% CI 0.11 to 0.26) probably improved progression-free survival compared to chemotherapy;• both BRAF inhibitors (HR 0.40, 95% CI 0.23 to 0.68) and combinations of small-molecule targeted drugs (HR 0.22, 95% CI 0.12 to 0.39) were probably associated with better progression-free survival compared to anti-CTLA4 monoclonal antibodies;• biochemotherapy (HR 2.81, 95% CI 1.76 to 4.51) probably lead to worse progression-free survival compared to BRAF inhibitors;• the combination of small-molecule targeted drugs probably improved progression-free survival (HR 0.38, 95% CI 0.21 to 0.68) compared to anti-PD1 monoclonal antibodies;• both biochemotherapy (HR 5.05, 95% CI 3.01 to 8.45) and MEK inhibitors (HR 3.16, 95% CI 1.77 to 5.65) were probably associated with worse progression-free survival compared to the combination of small-molecule targeted drugs; and• biochemotherapy was probably associated with worse progression-free survival (HR 2.81, 95% CI 1.54 to 5.11) compared to the combination of immune checkpoint inhibitors.The best evidence (moderate-quality evidence) for toxicity was found for the following indirect comparisons:• combination of immune checkpoint inhibitors (RR 3.49, 95% CI 2.12 to 5.77) probably increased toxicity compared to chemotherapy;• combination of immune checkpoint inhibitors probably increased toxicity (RR 2.50, 95% CI 1.20 to 5.20) compared to BRAF inhibitors;• the combination of immune checkpoint inhibitors probably increased toxicity (RR 3.83, 95% CI 2.59 to 5.68) compared to anti-PD1 monoclonal antibodies; and• biochemotherapy was probably associated with lower toxicity (RR 0.41, 95% CI 0.24 to 0.71) compared to the combination of immune checkpoint inhibitors.Network meta-analysis-based ranking suggested that the combination of BRAF plus MEK inhibitors is the most effective strategy in terms of progression-free survival, whereas anti-PD1 monoclonal antibodies are associated with the lowest toxicity.Overall, the risk of bias of the included trials can be considered as limited. When considering the 122 trials included in this review and the seven types of bias we assessed, we performed 854 evaluations only seven of which (< 1%) assigned high risk to six trials. AUTHORS' CONCLUSIONS We found high-quality evidence that many treatments offer better efficacy than chemotherapy, especially recently implemented treatments, such as small-molecule targeted drugs, which are used to treat melanoma with specific gene mutations. Compared with chemotherapy, biochemotherapy (in this case, chemotherapy combined with both interferon-alpha and interleukin-2) and BRAF inhibitors improved progression-free survival; BRAF inhibitors (for BRAF-mutated melanoma) and anti-PD1 monoclonal antibodies improved overall survival. However, there was no difference between polychemotherapy and monochemotherapy in terms of achieving progression-free survival and overall survival. Biochemotherapy did not significantly improve overall survival and has higher toxicity rates compared with chemotherapy.There was some evidence that combined treatments worked better than single treatments: anti-PD1 monoclonal antibodies, alone or with anti-CTLA4, improved progression-free survival compared with anti-CTLA4 monoclonal antibodies alone. Anti-PD1 monoclonal antibodies performed better than anti-CTLA4 monoclonal antibodies in terms of overall survival, and a combination of BRAF plus MEK inhibitors was associated with better overall survival for BRAF-mutated melanoma, compared to BRAF inhibitors alone.The combination of BRAF plus MEK inhibitors (which can only be administered to people with BRAF-mutated melanoma) appeared to be the most effective treatment (based on results for progression-free survival), whereas anti-PD1 monoclonal antibodies appeared to be the least toxic, and most acceptable, treatment.Evidence quality was reduced due to imprecision, between-study heterogeneity, and substandard reporting of trials. Future research should ensure that those diminishing influences are addressed. Clinical areas of future investigation should include the longer-term effect of new therapeutic agents (i.e. immune checkpoint inhibitors and targeted therapies) on overall survival, as well as the combination of drugs used in melanoma treatment; research should also investigate the potential influence of biomarkers.
Collapse
Affiliation(s)
- Sandro Pasquali
- Sarcoma Service, Fondazione IRCCS 'Istituto Nazionale Tumori', Via G. Venezian 1, Milano, Italy, 20133
| | | | | | | | | |
Collapse
|
24
|
Therapeutic Antibodies in Cancer Therapy. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 917:95-120. [PMID: 27236554 DOI: 10.1007/978-3-319-32805-8_6] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
The therapeutic arsenal in solid tumors comprises different anticancer strategies with diverse chemotherapeutic agents and a growing number of biological substances. Large clinical study-based chemotherapeutic protocols combined with biologicals have become an important component in (neo-) adjuvant therapy alongside surgery in solid cancers as well as radiation therapy in some instances. In recent years, monoclonal antibodies have entered the mainstream of cancer therapy. Their first use was as antagonists of oncogenic receptor tyrosine kinases, but today monoclonal antibodies have emerged as long-sought vehicles for the targeted delivery of potent chemotherapeutic agents and as powerful tools to manipulate anticancer immune responses. There is a growing number of FDA approved monoclonal antibodies and small molecules targeting specific types of cancer suggestive of the clinical relevance of this approach.Targeted cancer therapies , also referred to as personalized medicine, are being studied for use alone, in combination with other targeted therapies, and in combination with chemotherapy. The use of monoclonal antibodies in colorectal and gastric cancer for example have shown best outcome when combined with chemotherapy, even though single agent anti-EGFR antibodies seem to be active in particular setting of metastatic colorectal cancer patients. However, it is not well defined whether the addition of anti-VEGF - and anti-EGFR strategies to chemotherapy could improve outcome in those patients susceptible to colorectal cancer-related metastases resection. Among the most promising approaches to activating therapeutic antitumor immunity is the blockade of immune checkpoints, exemplified by the recently FDA-approved agent, Ipilimumab, an antibody that blocks the coinhibitory receptor CTLA-4. Capitalizing on the success of Ipilimumab, agents that target a second coinhibitory receptor, PD-1, or its ligand, PD-L1, are in clinical development. This section attempts to discuss recent progress of targeted agents and in tackling a more general target applicable to gastrointestinal cancer .
Collapse
|
25
|
Not all immune-checkpoint inhibitors are created equal: Meta-analysis and systematic review of immune-related adverse events in cancer trials. Crit Rev Oncol Hematol 2017; 119:1-12. [DOI: 10.1016/j.critrevonc.2017.09.002] [Citation(s) in RCA: 117] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2017] [Revised: 08/13/2017] [Accepted: 09/07/2017] [Indexed: 02/07/2023] Open
|
26
|
Fusi A, Dalgleish A. The importance for immunoregulation for long-term cancer control. Future Oncol 2017; 13:1619-1632. [DOI: 10.2217/fon-2017-0085] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Immune checkpoint blockades have recently emerged as a breakthrough treatment for solid tumors showing high response rates and long durability. In melanoma, the combination of ipilimumab with nivolumab showed high efficacy. However, still half the patients do not respond to this treatment. In order to increase the therapeutic ratio in melanoma and other cancers, different approaches are under evaluation. Three relevant questions are at the moment driving the research community: how to maximize benefit while minimizing toxicity; how to better identify patients who are more likely to benefit from immunotherapy; how to convert nonresponders into responders. In this review we summarize the most recent findings and we outline the most likely future challenges.
Collapse
Affiliation(s)
- Alberto Fusi
- Infection & Immunity Research Center – St George’s, University of London, Cranmer Terrace, London, SW17 0RE, UK
- Charité Comprehensive Cancer Center – Charité-Invalidenstr. 80, 10115 Berlin, Germany
| | - Angus Dalgleish
- Infection & Immunity Research Center – St George’s, University of London, Cranmer Terrace, London, SW17 0RE, UK
| |
Collapse
|
27
|
Bamias G, Delladetsima I, Perdiki M, Siakavellas SI, Goukos D, Papatheodoridis GV, Daikos GL, Gogas H. Immunological Characteristics of Colitis Associated with Anti-CTLA-4 Antibody Therapy. Cancer Invest 2017; 35:443-455. [PMID: 28548891 DOI: 10.1080/07357907.2017.1324032] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Anti-CTL4-A therapy is associated with development of colitis. We characterized ipilimumab-associated colitis in nine melanoma patients (6 male, mean age: 55.3-yrs). Median value for diarrhea grade was 2, number of ipilimumab doses 2, and interval since last administration 3-wks. Endoscopic characteristics resembled inflammatory bowel disease and histology revealed predominance of plasmacytes or CD4+ T-cells. We observed significant upregulation of Th1 and Th17 effector pathways (>10-fold increase for IFN-γmRNA, >5-fold for IL-17A, p < 0.01 vs. controls). Significant elevation of FoxP3 was also detected. In conclusion, ipilimumab administration results in elevations of effector lymphocytes and pro-inflammatory mediators in the gut lamina propria.
Collapse
Affiliation(s)
- Giorgos Bamias
- a Academic Department of Gastroenterology, School of Medicine , National and Kapodistrian University of Athens, Laiko General Hospital , Athens , Greece
| | - Ioanna Delladetsima
- b First Department of Pathology, School of Medicine , National and Kapodistrian University of Athens , Athens , Greece
| | - Marina Perdiki
- b First Department of Pathology, School of Medicine , National and Kapodistrian University of Athens , Athens , Greece
| | - Spyros I Siakavellas
- a Academic Department of Gastroenterology, School of Medicine , National and Kapodistrian University of Athens, Laiko General Hospital , Athens , Greece
| | - Dimitrios Goukos
- c First Department of Internal Medicine, School of Medicine , National and Kapodistrian University of Athens, Laiko General Hospital , Athens , Greece
| | - George V Papatheodoridis
- a Academic Department of Gastroenterology, School of Medicine , National and Kapodistrian University of Athens, Laiko General Hospital , Athens , Greece
| | - George L Daikos
- c First Department of Internal Medicine, School of Medicine , National and Kapodistrian University of Athens, Laiko General Hospital , Athens , Greece
| | - Helen Gogas
- c First Department of Internal Medicine, School of Medicine , National and Kapodistrian University of Athens, Laiko General Hospital , Athens , Greece
| |
Collapse
|
28
|
Enewold L, Sharon E, Harlan LC. Metastatic Melanoma: Treatment and Survival in the US after the Introduction of Ipilimumab and Vemurafenib. Oncol Res Treat 2017; 40:174-183. [PMID: 28376479 PMCID: PMC5383086 DOI: 10.1159/000456014] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2016] [Accepted: 11/28/2016] [Indexed: 01/07/2023]
Abstract
INTRODUCTION The 5-year survival of metastatic melanoma is < 18%. Historically, treatment options were limited. In 2011, 2 new agents were approved. METHODS We re-abstracted the medical records of a random sample (n = 520) of metastatic melanoma patients who had been diagnosed in 2011 and reported to population-based registries in the U.S. We also queried their treating physicians. Factors associated with treatment and survival were assessed using logistic and Cox proportional hazards regressions, respectively. RESULTS 21.4% of patients received no treatment, 20.8% received ipilimumab and 57.5% of patients with BRAF-positive tumors received vemurafenib/dabrafenib. Receipt of ipilimumab was less likely among patients of 75 years or older (vs. < 55 years: odds ratio (OR) 0.32; 95% confidence interval (CI) 0.15-0.66) and patients without private/military insurance. 46.8% of patients received BRAF testing. Receipt of BRAF testing was less likely among patients of 65 years or more and uninsured patients (OR 0.22; 95% CI 0.07-0.65). Receipt of ipilimumab was associated with better survival during the first 18 months after diagnosis (hazard ratio (HR) 0.66; 95% CI 0.51-0.84) and vemurafenib/dabrafenib with better survival during the first 10 months after diagnosis (HR 0.51; 95% CI 0.36-0.73). CONCLUSION The initial dissemination of ipilimumab and vemurafenib/dabrafenib was limited. Additional research is needed to investigate the apparent lack of long-term survival benefit from these agents.
Collapse
Affiliation(s)
- Lindsey Enewold
- National Cancer Institute, 9609 Medical Center Drive, Bethesda, MD 20892-9762
| | - Elad Sharon
- National Cancer Institute, 9609 Medical Center Drive, Bethesda, MD 20892-9762
| | - Linda C Harlan
- National Cancer Institute, 9609 Medical Center Drive, Bethesda, MD 20892-9762
| |
Collapse
|
29
|
Kalinski P, Talmadge JE. Tumor Immuno-Environment in Cancer Progression and Therapy. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 1036:1-18. [PMID: 29275461 DOI: 10.1007/978-3-319-67577-0_1] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The approvals of Provenge (Sipuleucel-T), Ipilimumab (Yervoy/anti-CTLA-4) and blockers of the PD-1 - PD-L1/PD-L2 pathway, such as nivolumab (Opdivo), pembrolizumab (Keytruda), or atezolizumab (Tecentriq), have established immunotherapy as a key component of comprehensive cancer care. Further, murine mechanistic studies and studies in immunocompromised patients have documented the critical role of immunity in effectiveness of radio- and chemotherapy. However, in addition to the ability of the immune system to control cancer progression, it can also promote tumor growth, via regulatory T cells (Tregs), myeloid-derived dendritic cells (MDSCs) and tumor associated macrophages (TAM), which can enhance survival of cancer cells directly or via the regulation of the tumor stroma.An increasing body of evidence supports a central role for the tumor microenvironment (TME) and the interactions between tumor stroma, infiltrating immune cells and cancer cells during the induction and effector phase of anti-cancer immunity, and the overall effectiveness of immunotherapy and other forms of cancer treatment. In this chapter, we discuss the roles of key TME components during tumor progression, metastatic process and cancer therapy-induced tumor regression, as well as opportunities for their modulation to enhance the overall therapeutic benefit.
Collapse
Affiliation(s)
- Pawel Kalinski
- Department of Medicine and Center for Immunotherapy, Roswell Park Cancer Institute, Buffalo, NY, USA.
| | - James E Talmadge
- University of Nebraska Medical Center, 986495 Nebraska Medical Center, Omaha, NE, USA
| |
Collapse
|
30
|
Abstract
This review focuses on summarizing the existing work about nanomaterial-based cancer immunotherapy in detail.
Collapse
Affiliation(s)
- Lijia Luo
- Key Laboratory of Magnetic Materials and Devices
- CAS & Key Laboratory of Additive Manufacturing Materials of Zhejiang Province, & Division of Functional Materials and Nanodevices
- Ningbo Institute of Materials Technology and Engineering
- Chinese Academy of Sciences
- Ningbo
| | - Rui Shu
- University of Chinese Academy of Sciences
- Beijing 100049
- China
- Key Laboratory of Marine Materials and Related Technology
- CAS & Ningbo Institute of Materials Technology and Engineering
| | - Aiguo Wu
- Key Laboratory of Magnetic Materials and Devices
- CAS & Key Laboratory of Additive Manufacturing Materials of Zhejiang Province, & Division of Functional Materials and Nanodevices
- Ningbo Institute of Materials Technology and Engineering
- Chinese Academy of Sciences
- Ningbo
| |
Collapse
|
31
|
Zhang LX, Liu DQ, Wang SW, Yu XL, Ji M, Xie XX, Liu SY, Liu RT. MgAl-layered double hydroxide nanoparticles co-delivering siIDO and Trp2 peptide effectively reduce IDO expression and induce cytotoxic T-lymphocyte responses against melanoma tumor in mice. J Mater Chem B 2017; 5:6266-6276. [DOI: 10.1039/c7tb00819h] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The co-delivery of Trp2 and siIDO by LDH nanoparticles alleviates immune tolerance and promotes CTL responses in vivo.
Collapse
Affiliation(s)
- Ling-xiao Zhang
- State Key Laboratory of Biochemical Engineering
- Institute of Process Engineering
- Chinese Academy of Sciences
- Beijing
- China
| | - Dong-qun Liu
- State Key Laboratory of Biochemical Engineering
- Institute of Process Engineering
- Chinese Academy of Sciences
- Beijing
- China
| | - Shao-wei Wang
- State Key Laboratory of Biochemical Engineering
- Institute of Process Engineering
- Chinese Academy of Sciences
- Beijing
- China
| | - Xiao-lin Yu
- State Key Laboratory of Biochemical Engineering
- Institute of Process Engineering
- Chinese Academy of Sciences
- Beijing
- China
| | - Mei Ji
- State Key Laboratory of Biochemical Engineering
- Institute of Process Engineering
- Chinese Academy of Sciences
- Beijing
- China
| | - Xi-xiu Xie
- State Key Laboratory of Biochemical Engineering
- Institute of Process Engineering
- Chinese Academy of Sciences
- Beijing
- China
| | - Shu-ying Liu
- State Key Laboratory of Biochemical Engineering
- Institute of Process Engineering
- Chinese Academy of Sciences
- Beijing
- China
| | - Rui-tian Liu
- State Key Laboratory of Biochemical Engineering
- Institute of Process Engineering
- Chinese Academy of Sciences
- Beijing
- China
| |
Collapse
|
32
|
Masarova L, Kantarjian H, Garcia-Mannero G, Ravandi F, Sharma P, Daver N. Harnessing the Immune System Against Leukemia: Monoclonal Antibodies and Checkpoint Strategies for AML. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 995:73-95. [PMID: 28321813 DOI: 10.1007/978-3-319-53156-4_4] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Acute myeloid leukemia (AML) is the most common leukemia among adults and is associated with a poor prognosis, especially in patients with adverse prognostic factors, older age, or relapsed disease. The last decade has seen a surge in successful immune-based therapies in various solid tumors; however, the role of immune therapies in AML remains poorly defined. This chapter describes the rationale, clinical data, and toxicity profiles of immune-based therapeutic modalities in AML including naked and conjugated monoclonal antibodies, bispecific T-cell engager antibodies, chimeric antigen receptor (CAR)-T cells, and checkpoint blockade via blockade of PD1/PDL1 or CTLA4. Monoclonal antibodies commonly used in AML therapy target highly expressed "leukemia" surface antigens and include (1) naked antibodies against common myeloid markers such as anti-CD33 (e.g., lintuzumab), (2) antibody-drug conjugates linked to either, (a) a highly potent toxin such as calicheamicin, pyrrolobenzodiazepine, maytansine, or others in various anti-CD33 (gemtuzumab ozogamicin, SGN 33A), anti-123 (SL-401), and anti-CD56 (lorvotuzumab mertansine) formulations, or (b) radioactive particles, such as 131I, 213Bi, or 225Ac-labeled anti-CD33 or CD45 antibodies. Novel monoclonal antibodies that recruit and promote proximity-induced cytotoxicity of tumor cells by T cells (bispecific T-cell engager [BiTE] such as anti CD33/CD3, e.g., AMG 330) or block immune checkpoint pathways such as CTLA4 (e.g., ipilimumab) or PD1/PD-L1 (e.g., nivolumab) unleashing the patients T cells to fight leukemic cells are being evaluated in clinical trials in patients with AML. The numerous ongoing clinical trials with immunotherapies in AML will improve our understanding of the biology of AML and allow us to determine the best approaches to immunotherapy in AML.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal/adverse effects
- Antibodies, Monoclonal/therapeutic use
- Antibody Specificity
- Antineoplastic Agents/adverse effects
- Antineoplastic Agents/therapeutic use
- Biomarkers, Tumor/antagonists & inhibitors
- Biomarkers, Tumor/immunology
- Biomarkers, Tumor/metabolism
- Humans
- Immunotherapy/methods
- Immunotherapy, Adoptive
- Leukemia, Myeloid, Acute/drug therapy
- Leukemia, Myeloid, Acute/immunology
- Leukemia, Myeloid, Acute/metabolism
- Leukemia, Myeloid, Acute/pathology
- Molecular Targeted Therapy
- Signal Transduction/drug effects
- T-Lymphocytes/microbiology
- T-Lymphocytes/transplantation
- Tumor Microenvironment
Collapse
Affiliation(s)
- Lucia Masarova
- Department of Leukemia, MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX, 77030, USA
| | - Hagop Kantarjian
- Department of Leukemia, MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX, 77030, USA
| | | | - Farhad Ravandi
- Department of Leukemia, MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX, 77030, USA
| | - Padmanee Sharma
- Immunotherapy Platform, MD Anderson Cancer Center, Houston, TX, USA
| | - Naval Daver
- Department of Leukemia, MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX, 77030, USA.
| |
Collapse
|
33
|
[Systemic treatment of inoperable metastasized malignant melanoma]. Hautarzt 2016; 67:529-35. [PMID: 27164828 DOI: 10.1007/s00105-016-3795-1] [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: 10/21/2022]
Abstract
BACKGROUND The medical therapy of inoperable malignant melanoma has changed dramatically over the last few years. OBJECTIVES The purpose of this article is to summarize the current state of systemic medical treatment of malignant melanoma. MATERIALS AND METHODS Clinical studies and guidelines in the therapy of malignant melanoma are reviewed. RESULTS Medical therapy of inoperable melanoma changed due to developments in immunotherapies (checkpoint inhibitors) and molecular-targeted therapies (BRAF and MEK inhibitors). Checkpoint inhibitors are antibodies administered as infusions every 2-3 weeks, blocking the checkpoints PD-1 or CTLA-4, thus, preventing downregulation of the immune system. BRAF and MEK inhibitors are small molecules, they are given orally and block a certain signaling pathway in tumor cells. The activation of this pathway has to be demonstrated by molecular analysis of tumor tissue first. This strategy is currently registered for 40-50 % of melanomas harboring a BRAF V600 mutation, while the combination of a BRAF plus MEK inhibitor has been proven more efficient than a BRAF inhibitor alone. DISCUSSION A fascinating development has started in the melanoma field due to immunotherapeutic and molecular-targeted treatment strategies. The continuation of this development needs further clinical and translational studies. This includes particular clinical studies with the new substances in the adjuvant situation, and sequences and combinations in the metastatic setting. Translational studies are needed to develop biomarkers for response and side effects.
Collapse
|
34
|
Staser K, Chen D, Solus J, Rosman IS, Schaffer A, Cornelius L, Linette GP, Fields RC. Extensive tumoral melanosis associated with ipilimumab-treated melanoma. Br J Dermatol 2016; 175:391-3. [PMID: 26877232 DOI: 10.1111/bjd.14474] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/04/2016] [Indexed: 11/29/2022]
Abstract
Tumoral melanosis describes a pigmented lesion clinically similar to melanoma but on histology reveals dense aggregates of melanin-laden, benign macrophages without malignant cells. In the few reported cases so far, tumoral melanosis has arisen in the skin or lymph node of a patient with a regressed melanoma or an epithelioid tumour. As a marker of regressed primary melanoma, its discovery may prompt investigation and surveillance for undiagnosed local or metastatic disease. Here, we present a unique case of extensive tumoral melanosis arising during ipilimumab treatment of in-transit metastases from a previously excised melanoma.
Collapse
Affiliation(s)
- K Staser
- Division of Dermatology, Washington University in St. Louis, St. Louis, MO, U.S.A
| | - D Chen
- Division of Dermatology, Washington University in St. Louis, St. Louis, MO, U.S.A
| | - J Solus
- Division of Dermatology, Washington University in St. Louis, St. Louis, MO, U.S.A.,Department of Pathology and Immunology, Washington University in St. Louis, St. Louis, MO, U.S.A
| | - I S Rosman
- Division of Dermatology, Washington University in St. Louis, St. Louis, MO, U.S.A.,Department of Pathology and Immunology, Washington University in St. Louis, St. Louis, MO, U.S.A
| | - A Schaffer
- Division of Dermatology, Washington University in St. Louis, St. Louis, MO, U.S.A.,Department of Pathology and Immunology, Washington University in St. Louis, St. Louis, MO, U.S.A
| | - L Cornelius
- Division of Dermatology, Washington University in St. Louis, St. Louis, MO, U.S.A
| | - G P Linette
- Division of Hematology and Oncology, Washington University in St. Louis, St. Louis, MO, U.S.A
| | - R C Fields
- Department of Surgery, Washington University in St. Louis, St. Louis, MO, U.S.A
| |
Collapse
|
35
|
Antonioli L, Yegutkin GG, Pacher P, Blandizzi C, Haskó G. Anti-CD73 in cancer immunotherapy: awakening new opportunities. Trends Cancer 2016; 2:95-109. [PMID: 27014745 PMCID: PMC4800751 DOI: 10.1016/j.trecan.2016.01.003] [Citation(s) in RCA: 160] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
In recent years, cancer immunotherapy made significant advances due to a better understanding of the principles underlying tumor biology and immunology. In this context, CD73 is a key molecule, since via degradation of adenosine monophosphate into adenosine, endorses the generation of an immunosuppressed and pro-angiogenic niche within the tumor microenvironment that promotes the onset and progression of cancer. Targeting CD73 results in favorable antitumor effects in pre-clinical models and combined treatments of CD73 blockade with other immune-modulating agents (i.e. anti-CTLA-4 mAb or anti-PD1 mAb) is particularly attractive. Although there is still a long way to go, anti-CD73 therapy, through the development of CD73 monoclonal antibodies, can potentially constitute a new biologic therapy for cancer patients. In this review, we discuss the link between CD73 and the onset, development and spread of tumors, highlighting the potential value of this molecule as a target and as a novel biomarker in the context of personalized cancer therapy.
Collapse
Affiliation(s)
- Luca Antonioli
- Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy; Department of Surgery and Center for Immunity and Inflammation, Rutgers-New Jersey Medical School, Newark, NJ 07103, USA
| | - Gennady G Yegutkin
- Medicity Research Laboratory, Department of Medical Microbiology and Immunology, University of Turku, Finland
| | - Pál Pacher
- Section on Oxidative Stress Tissue Injury, Laboratories of Physiological Studies, National Institutes of Health/NIAAA, Bethesda, MD 20892, USA
| | - Corrado Blandizzi
- Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy
| | - György Haskó
- Department of Surgery and Center for Immunity and Inflammation, Rutgers-New Jersey Medical School, Newark, NJ 07103, USA
| |
Collapse
|
36
|
Ye C, Geng Z, Dominguez D, Chen S, Fan J, Qin L, Long A, Zhang Y, Kuzel TM, Zhang B. Targeting Ornithine Decarboxylase by α-Difluoromethylornithine Inhibits Tumor Growth by Impairing Myeloid-Derived Suppressor Cells. THE JOURNAL OF IMMUNOLOGY 2015; 196:915-23. [PMID: 26663722 DOI: 10.4049/jimmunol.1500729] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/30/2015] [Accepted: 11/04/2015] [Indexed: 01/04/2023]
Abstract
α-Difluoromethylornithine (DFMO) is currently used in chemopreventive regimens primarily for its conventional direct anticarcinogenesic activity. However, little is known about the effect of ornithine decarboxylase (ODC) inhibition by DFMO on antitumor immune responses. We showed in this study that pharmacologic blockade of ODC by DFMO inhibited tumor growth in intact immunocompetent mice, but abrogated in the immunodeficient Rag1(-/-) mice, suggesting that antitumor effect of DFMO is dependent on the induction of adaptive antitumor T cell immune responses. Depletion of CD8(+) T cells impeded the tumor-inhibiting advantage of DFMO. Moreover, DFMO treatment enhanced antitumor CD8(+) T cell infiltration and IFN-γ production and augmented the efficacy of adoptive T cell therapy. Importantly, DFMO impaired Gr1(+)CD11b(+) myeloid-derived suppressor cells (MDSCs) suppressive activity through at least two mechanisms, including reducing arginase expression and activity and inhibiting the CD39/CD73-mediated pathway. MDSCs were one primary cellular target of DFMO as indicated by both adoptive transfer and MDSC-depletion analyses. Our findings establish a new role of ODC inhibition by DFMO as a viable and effective immunological adjunct in effective cancer treatment, thereby adding to the growing list of chemoimmunotherapeutic applications of these agents.
Collapse
Affiliation(s)
- Cong Ye
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China; Robert H. Lurie Comprehensive Cancer Center, Division of Hematology/Oncology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611; Department of Rheumatology and Immunology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; and
| | - Zhe Geng
- Robert H. Lurie Comprehensive Cancer Center, Division of Hematology/Oncology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611; Hubei Maternity and Child Health Hospital, Wuhan 430070, China
| | - Donye Dominguez
- Robert H. Lurie Comprehensive Cancer Center, Division of Hematology/Oncology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611
| | - Siqi Chen
- Robert H. Lurie Comprehensive Cancer Center, Division of Hematology/Oncology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611
| | - Jie Fan
- Robert H. Lurie Comprehensive Cancer Center, Division of Hematology/Oncology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611
| | - Lei Qin
- Robert H. Lurie Comprehensive Cancer Center, Division of Hematology/Oncology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611
| | - Alan Long
- Robert H. Lurie Comprehensive Cancer Center, Division of Hematology/Oncology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611
| | - Yi Zhang
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China
| | - Timothy M Kuzel
- Robert H. Lurie Comprehensive Cancer Center, Division of Hematology/Oncology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611
| | - Bin Zhang
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China; Robert H. Lurie Comprehensive Cancer Center, Division of Hematology/Oncology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611;
| |
Collapse
|
37
|
Lin M, Yu HP. Dexamethasone decreases IL-29 expression in house dust mite-stimulated human bronchial epithelial cells. JOURNAL OF HUAZHONG UNIVERSITY OF SCIENCE AND TECHNOLOGY. MEDICAL SCIENCES = HUA ZHONG KE JI DA XUE XUE BAO. YI XUE YING DE WEN BAN = HUAZHONG KEJI DAXUE XUEBAO. YIXUE YINGDEWEN BAN 2015; 35:823-827. [PMID: 26670431 DOI: 10.1007/s11596-015-1513-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/20/2015] [Revised: 11/02/2015] [Indexed: 12/12/2022]
Abstract
The aim of this study was to explore the effect of IL-29 on the progression of airway allergic disease by detecting the level of IL-29 in airway allergic cell models stimulated by house dust mite (HDM) in the presence or absence of dexamethasone (DEX). The same batch of human bronchial epithelial cells in exponential growth phase was randomly divided into five groups: blank group (A), 300 ng/mL HDM group (B), 1000 ng/mL HDM group (C), 3000 ng/mL HDM group (D), and 300 ng/mL HDM+100 ng/mL DEX group (E). The IL-29 mRNA expression was detected by quantitative reverse transcription polymerase chain reaction (qRT-PCR). The IL-29 protein expression in cell suspension was detected by ELISA. The results showed that after stimulation with HDM for 24 h, the expression of IL-29 was increased significantly, and after co-stimulation with HDM and DEX for 24 h, the expression of IL-29 in group E was significantly lower than that in the groups stimulated by HDM alone but higher than that in the group A. The differences between the different groups were significant (F=132.957, P<0.01). Additionally, the higher the concentration of HDM was, the more significant the increase in the IL-29 expression was. In conclusion, IL-29 may play a role in the progression of airway allergic disease including asthma.
Collapse
Affiliation(s)
- Mei Lin
- Southern Medical University, Guangzhou, 510515, China
- Wuhan Third Hospital, Wuhan, 430060, China
| | - Hua-Peng Yu
- Southern Medical University, Guangzhou, 510515, China.
| |
Collapse
|
38
|
Michielin O, Hoeller C. Gaining momentum: New options and opportunities for the treatment of advanced melanoma. Cancer Treat Rev 2015; 41:660-70. [DOI: 10.1016/j.ctrv.2015.05.012] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2015] [Revised: 05/26/2015] [Accepted: 05/27/2015] [Indexed: 12/31/2022]
|
39
|
Abstract
Advances in the understanding of the immunogenicity of tumors have provided the basis for immuno-oncology, the development of immunotherapeutic agents that augment the patient's antitumor immunity and disrupt the immune-regulatory circuits that allow tumors to evade the immune system. Two immunomodulatory agents recently have been introduced for the treatment of malignancy: sipuleucel-T and ipilimumab. Unlike cytotoxic chemotherapy, immunotherapies stimulate the patient's immune system to mount or augment existing endogenous antitumor immune responses. Both agents have demonstrated significant improvements in long-term overall survival in patients. Like other immunotherapies, sipuleucel-T and ipilimumab also are characterized by adverse events that manifest as immune-related inflammatory conditions that typically are low grade. Management guidelines have been developed and emphasize early recognition of the signs and symptoms of immune-related adverse events and treatment with corticosteroids. Because these events can manifest even after the cessation of therapy, patients treated with immunotherapies should continue to be followed by their oncology team and other healthcare providers.
Collapse
Affiliation(s)
- Rajni Kannan
- Laura and Isaac Perlmutter Cancer Center, New York University Medical Center in New York City
| | - Kathleen Madden
- Laura and Isaac Perlmutter Cancer Center, New York University Medical Center in New York City
| | | |
Collapse
|
40
|
Gibney GT, Kudchadkar RR, DeConti RC, Thebeau MS, Czupryn MP, Tetteh L, Eysmans C, Richards A, Schell MJ, Fisher KJ, Horak CE, Inzunza HD, Yu B, Martinez AJ, Younos I, Weber JS. Safety, correlative markers, and clinical results of adjuvant nivolumab in combination with vaccine in resected high-risk metastatic melanoma. Clin Cancer Res 2015; 21:712-20. [PMID: 25524312 PMCID: PMC4620684 DOI: 10.1158/1078-0432.ccr-14-2468] [Citation(s) in RCA: 196] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
PURPOSE The anti-programmed death-1 (PD-1) antibody nivolumab (BMS-936558) has clinical activity in patients with metastatic melanoma. Nivolumab plus vaccine was investigated as adjuvant therapy in resected stage IIIC and IV melanoma patients. EXPERIMENTAL DESIGN HLA-A*0201 positive patients with HMB-45, NY-ESO-1, and/or MART-1 positive resected tumors received nivolumab (1 mg/kg, 3 mg/kg, or 10 mg/kg i.v.) with a multi-peptide vaccine (gp100, MART-1, and NY-ESO-1 with Montanide ISA 51 VG) every 2 weeks for 12 doses followed by nivolumab maintenance every 12 weeks for 8 doses. Primary objective was safety and determination of a maximum tolerated dose (MTD). Secondary objectives included relapse-free survival (RFS), overall survival (OS), and immunologic correlative studies. RESULTS Thirty-three patients were enrolled. Median age was 47 years; 55% were male. Two patients had stage IIIC disease; 31 patients had stage IV disease. Median follow-up was 32.1 months. MTD was not reached. Most common related adverse events (>40%) were vaccine injection site reaction, fatigue, rash, pruritus, nausea, and arthralgias. Five related grade 3 adverse events [hypokalemia (1), rash (1), enteritis (1), and colitis (2)] were observed. Ten of 33 patients relapsed. Estimated median RFS was 47.1 months; median OS was not reached. Increases in CTLA-4(+)/CD4(+), CD25(+)Treg/CD4(+), and tetramer specific CD8(+) T-cell populations were observed with treatment (P < 0.05). Trends for lower baseline myeloid-derived suppressor cell and CD25(+)Treg/CD4(+) populations were seen in nonrelapsing patients; PD-L1 tumor status was not significantly associated with RFS. CONCLUSIONS Nivolumab with vaccine is well tolerated as adjuvant therapy and demonstrates immunologic activity with promising survival in high-risk resected melanoma, justifying further study.
Collapse
Affiliation(s)
- Geoffrey T Gibney
- Department of Cutaneous Oncology and Comprehensive Melanoma Research Center, Moffitt Cancer Center, Tampa, Florida. Department of Oncologic Sciences, University of South Florida, Tampa, Florida.
| | - Ragini R Kudchadkar
- Division of Oncology, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia
| | - Ronald C DeConti
- Department of Cutaneous Oncology and Comprehensive Melanoma Research Center, Moffitt Cancer Center, Tampa, Florida. Department of Oncologic Sciences, University of South Florida, Tampa, Florida
| | - Melissa S Thebeau
- Department of Cutaneous Oncology and Comprehensive Melanoma Research Center, Moffitt Cancer Center, Tampa, Florida
| | - Maria P Czupryn
- Department of Cutaneous Oncology and Comprehensive Melanoma Research Center, Moffitt Cancer Center, Tampa, Florida
| | - Leticia Tetteh
- Department of Cutaneous Oncology and Comprehensive Melanoma Research Center, Moffitt Cancer Center, Tampa, Florida
| | - Cabell Eysmans
- Division of Oncology, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia
| | - Allison Richards
- Department of Cutaneous Oncology and Comprehensive Melanoma Research Center, Moffitt Cancer Center, Tampa, Florida
| | - Michael J Schell
- Department of Biostatistics and Bioinformatics, Moffitt Cancer Center, Tampa, Florida
| | - Kate J Fisher
- Department of Biostatistics and Bioinformatics, Moffitt Cancer Center, Tampa, Florida
| | - Christine E Horak
- Bristol-Myers Squibb, Early Clinical and Translational Research, Princeton, New Jersey
| | - H David Inzunza
- Bristol-Myers Squibb, Early Clinical and Translational Research, Princeton, New Jersey
| | - Bin Yu
- Department of Cutaneous Oncology and Comprehensive Melanoma Research Center, Moffitt Cancer Center, Tampa, Florida
| | - Alberto J Martinez
- Department of Cutaneous Oncology and Comprehensive Melanoma Research Center, Moffitt Cancer Center, Tampa, Florida
| | - Ibrahim Younos
- Department of Clinical Pharmacology, College of Medicine, Minufiya University, Minufiya, Egypt. Department of Pharmacology and Clinical Pharmacy, College of Medicine and Health Sciences, Sultan Qaboos University, Muscat, Oman
| | - Jeffrey S Weber
- Department of Cutaneous Oncology and Comprehensive Melanoma Research Center, Moffitt Cancer Center, Tampa, Florida. Department of Oncologic Sciences, University of South Florida, Tampa, Florida
| |
Collapse
|
41
|
Kato Y, Yoshino I, Egusa C, Maeda T, Pili R, Tsuboi R. Combination of HDAC inhibitor MS-275 and IL-2 increased anti-tumor effect in a melanoma model via activated cytotoxic T cells. J Dermatol Sci 2014; 75:140-7. [DOI: 10.1016/j.jdermsci.2014.04.014] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2014] [Revised: 04/14/2014] [Accepted: 04/28/2014] [Indexed: 01/30/2023]
|
42
|
RUSSO ANGELA, FICILI BARTOLOMEA, CANDIDO SAVERIO, PEZZINO FRANCAMARIA, GUARNERI CLAUDIO, BIONDI ANTONIO, TRAVALI SALVATORE, McCUBREY JAMESA, SPANDIDOS DEMETRIOSA, LIBRA MASSIMO. Emerging targeted therapies for melanoma treatment (review). Int J Oncol 2014; 45:516-24. [PMID: 24899250 PMCID: PMC4091965 DOI: 10.3892/ijo.2014.2481] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2014] [Accepted: 06/03/2014] [Indexed: 12/14/2022] Open
Abstract
Cutaneous melanoma is an aggressive cancer with a poor prognosis for patients with advanced disease. The identification of several key molecular pathways implicated in the pathogenesis of melanoma has led to the development of novel therapies for this devastating disease. In melanoma, both the Ras/Raf/MEK/ERK (MAPK) and the PI3K/AKT (AKT) signalling pathways are constitutively activated through multiple mechanisms. Targeting various effectors of these pathways with pharmacologic inhibitors may inhibit melanoma cell growth and angiogenesis. Ongoing clinical trials provide hope to improve progression-free survival of patients with advanced melanoma. This review summarizes the most relevant studies focused on the specific action of these new molecular targeted agents. Mechanisms of resistance to therapy are also discussed.
Collapse
Affiliation(s)
- ANGELA RUSSO
- Laboratory of Translational Oncology and Functional Genomics, Section of General Pathology and Oncology, Department of Biomedical Sciences, University of Catania, I-95124 Catania, Italy
| | - BARTOLOMEA FICILI
- Laboratory of Translational Oncology and Functional Genomics, Section of General Pathology and Oncology, Department of Biomedical Sciences, University of Catania, I-95124 Catania, Italy
| | - SAVERIO CANDIDO
- Laboratory of Translational Oncology and Functional Genomics, Section of General Pathology and Oncology, Department of Biomedical Sciences, University of Catania, I-95124 Catania, Italy
| | - FRANCA MARIA PEZZINO
- Laboratory of Translational Oncology and Functional Genomics, Section of General Pathology and Oncology, Department of Biomedical Sciences, University of Catania, I-95124 Catania, Italy
| | - CLAUDIO GUARNERI
- Department of Social Territorial Medicine, Section of Dermatology, University of Messina, I-98125 Messina, Italy
| | - ANTONIO BIONDI
- Department of Surgery, University of Catania, I-95124 Catania, Italy
| | - SALVATORE TRAVALI
- Laboratory of Translational Oncology and Functional Genomics, Section of General Pathology and Oncology, Department of Biomedical Sciences, University of Catania, I-95124 Catania, Italy
| | - JAMES A. McCUBREY
- Department of Microbiology and Immunology, Brody School of Medicine at East Carolina University, Greenville, NC, USA
| | - DEMETRIOS A. SPANDIDOS
- Department of Virology, Medical School, University of Crete, Heraklion 71003, Crete, Greece
| | - MASSIMO LIBRA
- Laboratory of Translational Oncology and Functional Genomics, Section of General Pathology and Oncology, Department of Biomedical Sciences, University of Catania, I-95124 Catania, Italy
| |
Collapse
|
43
|
Pasquali S, Kefford R, Chiarion Sileni V, Nitti D, Rossi CR, Pilati P, Mocellin S. Systemic treatments for metastatic cutaneous melanoma. THE COCHRANE DATABASE OF SYSTEMATIC REVIEWS 2014. [DOI: 10.1002/14651858.cd011123] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Sandro Pasquali
- Veneto Institute of Oncology - IRCCS; Surgical Oncology Unit; Via Gattamelata 64 Padova Italy 35128
| | - Richard Kefford
- The University of Sydney; Discipline of Medicine; Clinical Sciences Block, Westmead Hospital Westmead Australia 2145
| | - Vanna Chiarion Sileni
- Veneto Region Oncology Research Institute; Medical Oncology Unit 2; Via Gattamelata 64 Padova Italy 35128
| | - Donato Nitti
- University of Padova; Clinica Chirurgica II; Via Giustiniani 2 Padova Italy 35128
| | - Carlo Riccardo Rossi
- Veneto Institute of Oncology; Melanoma and Sarcomas Unit; Via Gattamelata 64 Padova Italy 35128
| | - Pierluigi Pilati
- University of Padova; Meta-Analysis Unit, Department of Surgery, Oncology and Gastroenterology; via Giustiniani 2 Padova Italy 35128
| | - Simone Mocellin
- University of Padova; Dept. Surgery Oncology and Gastroenterology; Via Giustiniani 2 Padova Veneto Italy 35128
- IOV-IRCCS; Istituto Oncologico Veneto; Padova Italy 35100
| |
Collapse
|
44
|
Jin CH, Krishnaiah M, Sreenu D, Subrahmanyam VB, Rao KS, Lee HJ, Park SJ, Park HJ, Lee K, Sheen YY, Kim DK. Discovery of N-((4-([1,2,4]triazolo[1,5-a]pyridin-6-yl)-5-(6-methylpyridin-2-yl)-1H-imidazol-2-yl)methyl)-2-fluoroaniline (EW-7197): a highly potent, selective, and orally bioavailable inhibitor of TGF-β type I receptor kinase as cancer immunotherapeutic/antifibrotic agent. J Med Chem 2014; 57:4213-38. [PMID: 24786585 DOI: 10.1021/jm500115w] [Citation(s) in RCA: 102] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
A series of 2-substituted-4-([1,2,4]triazolo[1,5-a]pyridin-6-yl)-5-(6-methylpyridin-2-yl)imidazoles was synthesized and evaluated to optimize a prototype inhibitor of TGF-β type I receptor kinase (ALK5), 6. Combination of replacement of a quinoxalin-6-yl moiety of 6 with a [1,2,4]triazolo[1,5-a]pyridin-6-yl moiety, insertion of a methyleneamino linker, and a o-F substituent in the phenyl ring markedly increased ALK5 inhibitory activity, kinase selectivity, and oral bioavailability. The 12b (EW-7197) inhibited ALK5 with IC50 value of 0.013 μM in a kinase assay and with IC50 values of 0.0165 and 0.0121 μM in HaCaT (3TP-luc) stable cells and 4T1 (3TP-luc) stable cells, respectively, in a luciferase assay. Selectivity profiling of 12b using a panel of 320 protein kinases revealed that it is a highly selective ALK5/ALK4 inhibitor. Pharmacokinetic study with 12b·HCl in rats showed an oral bioavailability of 51% with high systemic exposure (AUC) of 1426 ng × h/mL and maximum plasma concentration (Cmax) of 1620 ng/mL. Rational optimization of 6 has led to the identification of a highly potent, selective, and orally bioavailable ALK5 inhibitor 12b.
Collapse
Affiliation(s)
- Cheng Hua Jin
- Graduate School of Pharmaceutical Sciences, College of Pharmacy, Ewha Womans University , 11-1 Daehyun-dong, Seodaemun-gu, Seoul 120-750, Korea
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Roddie C, Peggs KS. Emerging options for the treatment of melanoma - focus on ipilimumab. Immunotargets Ther 2014; 3:67-78. [PMID: 27482517 PMCID: PMC4918235 DOI: 10.2147/itt.s43522] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Ipilimumab is a fully human immunoglobulin subclass G1 anticytotoxic-T-lymphocyte-antigen-4 monoclonal antibody. It has been approved by the US Food and Drug Administration (FDA) and the European Medicines Agency for use in advanced melanoma following clear evidence of survival benefit in randomized Phase III studies. It is also under investigation as a treatment for other solid tumors such as renal cell, lung, and prostate cancers. The purported mechanism of antitumor activity of ipilimumab is through T-cell activation, and the side effect profile reflects this. Immune-related adverse events (irAEs) affect 60% of treated patients and 15% are defined as severe. Fortunately, most irAEs are reversible with early diagnosis and correct management. FDA approval of ipilimumab is dependent on the careful execution of a risk evaluation and mitigation strategy, with the aim of increasing awareness amongst patients and clinicians of the immunological risks of treatment, and providing algorithms for management of irAEs as they develop. Ipilimumab is one of the first immunotherapies to become widely available in the setting of solid tumors, and ongoing research aims to elucidate optimal dosing, optimal scheduling, and expanded access to ipilimumab as an adjuvant or maintenance therapy where appropriate. The identification of clinical correlates or biomarkers to identify those likely to benefit from this high-cost therapy is a top priority.
Collapse
Affiliation(s)
- Claire Roddie
- UCL Cancer Institute, Department of Hematology, London, UK
| | - Karl S Peggs
- UCL Cancer Institute, Department of Hematology, London, UK
| |
Collapse
|
46
|
Sim GC, Martin-Orozco N, Jin L, Yang Y, Wu S, Washington E, Sanders D, Lacey C, Wang Y, Vence L, Hwu P, Radvanyi L. IL-2 therapy promotes suppressive ICOS+ Treg expansion in melanoma patients. J Clin Invest 2014; 124:99-110. [PMID: 24292706 DOI: 10.1172/jci46266] [Citation(s) in RCA: 183] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2013] [Accepted: 09/26/2013] [Indexed: 12/11/2022] Open
Abstract
High-dose (HD) IL-2 therapy in patients with cancer increases the general population of Tregs, which are positive for CD4, CD25, and the Treg-specific marker Foxp3. It is unknown whether specific subsets of Tregs are activated and expanded during HD IL-2 therapy or whether activation of any particular Treg subset correlates with clinical outcome. Here, we evaluated Treg population subsets that were induced in patients with melanoma following HD IL-2 therapy. We identified a Treg population that was positive for CD4, CD25, Foxp3, and the inducible T cell costimulator (ICOS). This Treg population increased more than any other lymphocyte subset during HD IL-2 therapy and had an activated Treg phenotype, as indicated by high levels of CD39, CD73, and TGF-β. ICOS(+) Tregs were the most proliferative lymphocyte population in the blood after IL-2 therapy. Patients with melanoma with enhanced expansion of ICOS(+) Tregs in blood following the first cycle of HD IL-2 therapy had worse clinical outcomes than patients with fewer ICOS(+) Tregs. However, there was no difference in total Treg expansion between HD IL-2 responders and nonresponders. These data suggest that increased expansion of the ICOS(+) Treg population following the first cycle of HD IL-2 therapy may be predictive of clinical outcome.
Collapse
|
47
|
Alibek K, Baiken Y, Kakpenova A, Mussabekova A, Zhussupbekova S, Akan M, Sultankulov B. Implication of human herpesviruses in oncogenesis through immune evasion and supression. Infect Agent Cancer 2014; 9:3. [PMID: 24438207 PMCID: PMC3904197 DOI: 10.1186/1750-9378-9-3] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2013] [Accepted: 01/07/2014] [Indexed: 12/20/2022] Open
Abstract
All human herpesviruses (HHVs) have been implicated in immune system evasion and suppression. Moreover, two HHV family members, i.e. EBV and KSHV, are recognised as oncogenic viruses. Our literature review summarises additional examples of possible oncogenic mechanisms that have been attributed to other HHVs. In general, HHVs affect almost every cancer-implicated branch of the immune system, namely tumour-promoting inflammation, immune evasion, and immunosuppression. Some HHVs accomplish these effects by inhibiting apoptotic pathways and by promoting proliferation. Mechanisms related to immunosupression and low grade chronic inflammation could eventually result in the initiation and progression of cancer. In this article we open a discussion on the members of Herpesviridae, their immune evasion and suppression mechanisms, and their possible role in cancer development. We conclude that discerning the mechanisms of interplay between HHV, immune system, and cancer is essential for the development of novel preventative and therapeutic approaches for cancer treatment and prophylaxis.
Collapse
Affiliation(s)
| | | | - Ainur Kakpenova
- Nazarbayev University, 53 Kabanbay Batyr Avenue, Astana 010000, Kazakhstan.
| | | | | | | | | |
Collapse
|
48
|
Nicolay HJM, Sigalotti L, Fonsatti E, Covre A, Parisi G, Fratta E, Coral S, Maio M. Epigenetically regulated tumor-associated antigens in melanoma. ACTA ACUST UNITED AC 2014. [DOI: 10.1586/edm.09.6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
|
49
|
Ridolfi L, Ridolfi R. Anti-CTLA-4 therapy in melanoma: role of ipilimumab (MDX-010). ACTA ACUST UNITED AC 2014. [DOI: 10.1586/edm.09.11] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
|
50
|
Cesano A, Spellmeyer D. Immune system functional pathway analysis using single cell network profiling (SCNP): a novel tool in cancer immunotherapy. Methods Mol Biol 2014; 1102:583-99. [PMID: 24259000 DOI: 10.1007/978-1-62703-727-3_31] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
The development of cancer immunotherapies has been ongoing for many years and has shown limited success. Novel biomarkers are needed to identify patients most likely to respond to anticancer immune-therapeutic approaches. Moreover, a systems-level approach is required for comprehensive understanding of the interconnected components, pathways, and cell types associated with an immune response. In this chapter, we describe single cell network profiling (SCNP), a novel method for assessing and measuring immune function/dysfunction at a systems level. SCNP is a multiparametric flow-cytometry-based analysis that can simultaneously measure, at the single cell level, both extracellular surface markers and changes in intracellular signaling proteins in response to extracellular modulators. Measuring changes in signaling proteins following the application of an external modulation informs on the functional capacity of the signaling network which cannot be assessed by the measurement of basal signaling alone. In addition, the simultaneous analysis of multiple pathways in multiple cell subsets can provide insight into the connectivity of both cell signaling networks and immune cell subtypes. The experimental steps associated with an SCNP assay are (1) pre-analytical sample preparation; (2) modulation for functional analysis; (3) staining with antibody cocktail; (4) data acquisition on flow cytometer; and (5) data analysis and metrics. Important considerations for each step of the assay will be discussed, and data demonstrating the utility of SCNP for immune monitoring applications will be summarized.
Collapse
|