1
|
Dia AK, Kolnohuz A, Yolchuyeva S, Tonneau M, Lamaze F, Orain M, Gagné A, Blais F, Coulombe F, Malo J, Belkaid W, Elkrief A, Williamson D, Routy B, Joubert P, Laplante M, Bilodeau S, Manem VS. Computational analysis of whole slide images predicts PD-L1 expression and progression-free survival in immunotherapy-treated non-small cell lung cancer patients. J Transl Med 2025; 23:510. [PMID: 40329352 PMCID: PMC12056990 DOI: 10.1186/s12967-025-06487-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Accepted: 04/13/2025] [Indexed: 05/08/2025] Open
Abstract
BACKGROUND Immune checkpoint inhibitors (ICIs) have revolutionized cancer treatment by significantly improving the efficacy of treatments and tolerability for patients with non-small cell lung cancer (NSCLC). However, even after meticulous selection based on molecular criteria, only 20-30% of the patients respond to ICIs. This highlights the urgent clinical need to develop more precise biomarkers to better identify individuals who will benefit from these expensive therapies. METHODS Data from NSCLC patients treated with immunotherapy were collected from two institutions. From the histological images of tumors, pathomics features were extracted. We employed six machine learning models and seven feature selection methods to predict expression of the programmed death-ligand 1 (PD-L1), a current biomarker used to select patients for immunotherapy, and progression-free survival (PFS). The association between pathomics features and biological pathways was explored to validate pathomics-based signatures. We performed gene set enrichment analysis to identify the pathways enriched with the predictive signatures. RESULTS Handcrafted histological features were extracted from the whole slide images (WSI). The Support Vector Machines model with the SurfStar feature selection method, offered the best results, with an area under the curve (AUC) of around 0.66 for both the training and validation sets to predict PD-L1. For the prediction of PFS, the most effective model was linear discriminant analysis using the Multi Surf feature selection method with an AUC of 0.71 for the training set and 0.62 for the validation set. We found immune pathways to be upregulated in the high PD-L1 and high PFS groups, confirming the utility of image analysis for predicting clinical endpoints in patients treated with immunotherapy. CONCLUSION Our models, based on the analysis of histological images, can serve as predictive biomarkers for PD-L1 and PFS. This approach, focused on histological images, enables the distinction of patients based on treatment response, thus providing clinicians with a valuable tool for patient management. With further validation on external cohorts, these models could enhance clinical decision-making through analysis of routine medical images.
Collapse
Affiliation(s)
- Abdou Khadir Dia
- Centre de Recherche du CHU de Québec - Université Laval, Québec, Canada
| | - Alona Kolnohuz
- Quebec Heart & Lung Institute Research Center, Québec, Canada
- Université Laval, Québec, Canada
| | - Sevinj Yolchuyeva
- Centre de Recherche du CHU de Québec - Université Laval, Québec, Canada
- Department of Molecular Biology, Medical Biochemistry and Pathology, Université Laval, Québec, Canada
| | - Marion Tonneau
- Centre de Recherche du Centre Hospitalier Universitaire de Montréal, Montréal, Canada
- Université de médecine de Lille, Lille, France
| | - Fabien Lamaze
- Quebec Heart & Lung Institute Research Center, Québec, Canada
| | - Michele Orain
- Quebec Heart & Lung Institute Research Center, Québec, Canada
| | | | | | | | - Julie Malo
- Centre de Recherche du Centre Hospitalier Universitaire de Montréal, Montréal, Canada
| | - Wiam Belkaid
- Centre de Recherche du Centre Hospitalier Universitaire de Montréal, Montréal, Canada
| | - Arielle Elkrief
- Centre de Recherche du Centre Hospitalier Universitaire de Montréal, Montréal, Canada
| | - Drew Williamson
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, USA
| | - Bertrand Routy
- Centre de Recherche du Centre Hospitalier Universitaire de Montréal, Montréal, Canada
| | - Philippe Joubert
- Quebec Heart & Lung Institute Research Center, Québec, Canada
- Department of Molecular Biology, Medical Biochemistry and Pathology, Université Laval, Québec, Canada
| | - Mathieu Laplante
- Quebec Heart & Lung Institute Research Center, Québec, Canada
- Université Laval, Québec, Canada
| | - Steve Bilodeau
- Centre de Recherche du CHU de Québec - Université Laval, Québec, Canada
- Department of Molecular Biology, Medical Biochemistry and Pathology, Université Laval, Québec, Canada
- Cancer Research Center, Université Laval, Québec, Canada
- Big Data Research Center, Université Laval, Québec, Canada
| | - Venkata Sk Manem
- Centre de Recherche du CHU de Québec - Université Laval, Québec, Canada.
- Quebec Heart & Lung Institute Research Center, Québec, Canada.
- Department of Molecular Biology, Medical Biochemistry and Pathology, Université Laval, Québec, Canada.
- Cancer Research Center, Université Laval, Québec, Canada.
- Big Data Research Center, Université Laval, Québec, Canada.
| |
Collapse
|
2
|
Duan Q, Zhang Q, Jiang S, Nie K, Feng S, Qiu Y, He P, Xing Y, Liu J, Ma G, Zhang Y, Gao Y, Wang L. Transmission of peripheral blood α-synuclein fibrils exacerbates synucleinopathy and neurodegeneration in Parkinson's disease by endothelial Lag3 endocytosis. Am J Physiol Cell Physiol 2025; 328:C836-C855. [PMID: 39652643 DOI: 10.1152/ajpcell.00639.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Revised: 11/03/2024] [Accepted: 11/04/2024] [Indexed: 02/20/2025]
Abstract
Parkinson's disease (PD) is an age-related neurodegenerative disorder. The pathological feature of PD is abnormal α-synuclein (α-syn) formation and transmission. Recent evidence demonstrates that α-syn preformed fibrils (α-syn PFFs) can be detected in the serum of patients with PD. The peripheral blood α-syn PFF can cross the blood-brain barrier (BBB) and aggravate neuronal damage, but the mechanism remains to be elucidated. We constructed the PD mouse models of different severity: the mild pathology (A53T ONLY) and the severe pathology (A53T + Brain FIB); this was followed by α-syn PFFs intravenous injection. Then, we used endothelium-specific Lag3 knockout mice (Lag3-ECs-CKO) to decrease the blood α-syn PFFs spreading. We observed that intravenous transmission of α-syn PFFs significantly aggravated motor deficits, dopaminergic neuron loss, neuroinflammation, and pathologic α-syn deposition in A53T ONLY, but not in A53T + Brain FIB. Blocking endothelial Lag3 endocytosis by Lag3-ECs-CKO decreased the blood α-syn PFFs spreading and improved the symptoms and pathogenesis of PD mice. Our findings reveal the role of peripheral blood α-syn PFFs transmission in the mild pathology or early-stage PD and the mechanism of endothelial Lag3 endocytosis in the pathology of α-syn transmission. Targeting endothelial Lag3 to prevent α-syn from spreading from the blood to the brain may be a disease-modifying therapy in early-stage PD.NEW & NOTEWORTHY This study highlights the transmission mechanism of peripheral blood α-synuclein preformed fibrils (α-syn PFFs) through endothelial Lag3 endocytosis in the mild pathology or early-stage Parkinson's disease (PD). Targeting endothelial Lag3 as a perspective of decreasing peripheral blood α-syn PFFs transmission may be a disease-modifying therapy in early-stage PD.
Collapse
Affiliation(s)
- Qingrui Duan
- Department of Neurology, Guangdong Neuroscience Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, People's Republic of China
- Guangzhou Key Laboratory of Diagnosis and Treatment for Neurodegenerative Diseases, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, People's Republic of China
- School of Medicine South China University of Technology, Guangzhou, People's Republic of China
| | - Qingxi Zhang
- Department of Neurology, Guangdong Neuroscience Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, People's Republic of China
- Guangzhou Key Laboratory of Diagnosis and Treatment for Neurodegenerative Diseases, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, People's Republic of China
| | - ShuoLin Jiang
- Department of Neurology, Guangdong Neuroscience Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, People's Republic of China
- Guangzhou Key Laboratory of Diagnosis and Treatment for Neurodegenerative Diseases, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, People's Republic of China
- School of Medicine South China University of Technology, Guangzhou, People's Republic of China
| | - Kun Nie
- Department of Neurology, Guangdong Neuroscience Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, People's Republic of China
- Guangzhou Key Laboratory of Diagnosis and Treatment for Neurodegenerative Diseases, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, People's Republic of China
| | - Shujun Feng
- Department of Neurology, Guangdong Neuroscience Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, People's Republic of China
- Guangzhou Key Laboratory of Diagnosis and Treatment for Neurodegenerative Diseases, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, People's Republic of China
| | - Yihui Qiu
- Department of Neurology, Guangdong Neuroscience Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, People's Republic of China
- Guangzhou Key Laboratory of Diagnosis and Treatment for Neurodegenerative Diseases, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, People's Republic of China
| | - Peikun He
- Department of Neurology, Guangdong Neuroscience Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, People's Republic of China
- Guangzhou Key Laboratory of Diagnosis and Treatment for Neurodegenerative Diseases, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, People's Republic of China
| | - Yuxuan Xing
- Department of Neurology, Guangdong Neuroscience Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, People's Republic of China
- Guangzhou Key Laboratory of Diagnosis and Treatment for Neurodegenerative Diseases, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, People's Republic of China
| | - Jiaxuan Liu
- Department of Neurology, Guangdong Neuroscience Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, People's Republic of China
- Guangzhou Key Laboratory of Diagnosis and Treatment for Neurodegenerative Diseases, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, People's Republic of China
| | - Guixian Ma
- Department of Neurology, Guangdong Neuroscience Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, People's Republic of China
- Guangzhou Key Laboratory of Diagnosis and Treatment for Neurodegenerative Diseases, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, People's Republic of China
| | - Yuhu Zhang
- Department of Neurology, Guangdong Neuroscience Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, People's Republic of China
- Guangzhou Key Laboratory of Diagnosis and Treatment for Neurodegenerative Diseases, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, People's Republic of China
| | - Yuyuan Gao
- Department of Neurology, Guangdong Neuroscience Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, People's Republic of China
- Guangzhou Key Laboratory of Diagnosis and Treatment for Neurodegenerative Diseases, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, People's Republic of China
| | - Lijuan Wang
- Department of Neurology, Guangdong Neuroscience Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, People's Republic of China
- Guangzhou Key Laboratory of Diagnosis and Treatment for Neurodegenerative Diseases, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, People's Republic of China
- School of Medicine South China University of Technology, Guangzhou, People's Republic of China
| |
Collapse
|
3
|
Gupta AA, Tinker A, Jonker D, Jamal R, Hirte H, Winquist EW, Chu Q, Kollmannsberger C, Wong R, Alcindor T, Nielsen TO, Tsao M, Cottrell TR, Provencher D, Hilton J, Krzyżanowska MK, Elser C, Hotte S, Sederias J, Zhang S, Tu W, Dancey J. Durvalumab and tremelimumab in patients with advanced rare cancer: a multi-centre, non-blinded, open-label phase II basket trial. EClinicalMedicine 2025; 79:102991. [PMID: 39737219 PMCID: PMC11683278 DOI: 10.1016/j.eclinm.2024.102991] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Revised: 11/04/2024] [Accepted: 11/22/2024] [Indexed: 01/01/2025] Open
Abstract
Background Dual inhibition of cytotoxic T-lymphocyte associated protein 4 (CTLA-4) and programmed death ligand 1 (PD-L1) has been shown to be an effective treatment strategy in many cancers. We sought to determine the objective response rate of combination durvalumab (D) plus tremelimumab (TM) in parallel cohorts of patients with carefully selected rare cancer types in which these agents had not previously been evaluated in phase II trials and for which there was clinical or biological rationale for dual immune checkpoint inhibitor therapy to be active. Methods We designed a multi-centre, non-blinded, open-label phase II basket trial with each of the following 8 rare cancers considered a separate phase II trial: salivary carcinoma, carcinoma of unknown primary (CUP) with tumour infiltrating lymphocytes and/or expressing PD-L1, mucosal melanoma, acral melanoma, osteosarcoma, undifferentiated pleomorphic sarcoma, clear cell carcinoma of the ovary (CCCO) or squamous cell carcinoma of the anal canal (SCCA). The primary objective was to evaluate the response rate of the combination of D and TM, and the secondary objectives were to evaluate the tolerability and safety of D and TM combination. Eligible patients had advanced, metastatic or recurrent, or unresectable cancer with no known life-prolonging treatment option, age ≥16 years, ECOG performance status 0 or 1. Patients received D (1500 mg IV) + TM (75 mg IV) on Day 1 q4 weeks for 4 cycles followed by D q4 weeks until disease progression. This trial is registered with ClinicalTrials.gov, NCT02879162. Findings From December 14th, 2016, to August 14, 2019, 140 patients enrolled into seven cohorts. The rare melanoma cohorts were closed due to lack of accrual. Of the 140 patients enrolled, 138 were eligible, 138 were evaluable for toxicity and 128 (91%) were evaluable for response. Durable responses were noted in all cohorts except for osteosarcoma. The overall response rate for eligible patients was 16% (95% CI: 10-23%). The response rates in each cancer cohort were undifferentiated pleomorphic sarcoma 15% (n = 3/20; 95% CI 3-38%), salivary carcinoma 20% (n = 4/20; 95% CI: 6-44%), CUP 17% (n = 3/18; 95% CI 4-41%), SCCA 10% (n = 2/20; 95% CI 12-32%) and CCCO 21% (n = 8/39; 95% CI 9-37%). Grade 3/4 adverse events were rare, where 4 patients experienced grade 4 related events and39 patients experienced grade 3 events. Interpretation Durvalumab + tremelimumab treatment resulted in meaningful responses in salivary carcinoma and CCCO and deserves further exploration in front-line studies. Funding AstraZeneca and Canadian Cancer Society.
Collapse
Affiliation(s)
- Abha A. Gupta
- University Health Network, Princess Margaret Cancer Centre, Toronto, ON, Canada
| | | | - Derek Jonker
- Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | - Rahma Jamal
- CHUM-Centre Hospitalier de l'Universite de Montreal, Montreal, QC, Canada
| | - Hal Hirte
- Juravinski Cancer Centre at Hamilton Health Sciences, Hamilton, ON, Canada
| | | | - Quincy Chu
- CancerCare Manitoba, Winnipeg, MB, Canada
| | | | - Ralph Wong
- CancerCare Manitoba, Winnipeg, MB, Canada
| | - Thierry Alcindor
- The Research Institute of the McGill University, Montreal, QC, Canada
| | - Torsten O. Nielsen
- BC Cancer and Molecular and Advanced Pathology Centre, University of British Columbia, Vancouver, BC, Canada
| | - Ming Tsao
- University Health Network, Princess Margaret Cancer Centre, Toronto, ON, Canada
| | | | - Diane Provencher
- CHUM-Centre Hospitalier de l'Universite de Montreal, Montreal, QC, Canada
| | - John Hilton
- Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | | | - Christine Elser
- University Health Network, Princess Margaret Cancer Centre, Toronto, ON, Canada
| | - Sebastien Hotte
- Juravinski Cancer Centre at Hamilton Health Sciences, Hamilton, ON, Canada
| | - Joana Sederias
- Canadian Cancer Trials Group, Queen's University, Kingston, ON, Canada
| | - Siwei Zhang
- Canadian Cancer Trials Group, Queen's University, Kingston, ON, Canada
| | - Wei Tu
- Canadian Cancer Trials Group, Queen's University, Kingston, ON, Canada
| | - Janet Dancey
- Canadian Cancer Trials Group, Queen's University, Kingston, ON, Canada
| |
Collapse
|
4
|
Guo X, Bian X, Li Y, Zhu X, Zhou X. The intricate dance of tumor evolution: Exploring immune escape, tumor migration, drug resistance, and treatment strategies. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167098. [PMID: 38412927 DOI: 10.1016/j.bbadis.2024.167098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 01/14/2024] [Accepted: 02/19/2024] [Indexed: 02/29/2024]
Abstract
Recent research has unveiled fascinating insights into the intricate mechanisms governing tumor evolution. These studies have illuminated how tumors adapt and proliferate by exploiting various factors, including immune evasion, resistance to therapeutic drugs, genetic mutations, and their ability to adapt to different environments. Furthermore, investigations into tumor heterogeneity and chromosomal aberrations have revealed the profound complexity that underlies the evolution of cancer. Emerging findings have also underscored the role of viral influences in the development and progression of cancer, introducing an additional layer of complexity to the field of oncology. Tumor evolution is a dynamic and complex process influenced by various factors, including immune evasion, drug resistance, tumor heterogeneity, and viral influences. Understanding these elements is indispensable for developing more effective treatments and advancing cancer therapies. A holistic approach to studying and addressing tumor evolution is crucial in the ongoing battle against cancer. The main goal of this comprehensive review is to explore the intricate relationship between tumor evolution and critical aspects of cancer biology. By delving into this complex interplay, we aim to provide a profound understanding of how tumors evolve, adapt, and respond to treatment strategies. This review underscores the pivotal importance of comprehending tumor evolution in shaping effective approaches to cancer treatment.
Collapse
Affiliation(s)
- Xiaojun Guo
- Department of Immunology, School of Medicine, Nantong University, Nantong, China; The Marine Biomedical Research Institute of Guangdong Zhanjiang, School of Ocean and Tropical Medicine, Guangdong Medical University, Zhanjiang, China
| | - Xiaonan Bian
- Department of Immunology, School of Medicine, Nantong University, Nantong, China
| | - Yitong Li
- The Marine Biomedical Research Institute of Guangdong Zhanjiang, School of Ocean and Tropical Medicine, Guangdong Medical University, Zhanjiang, China
| | - Xiao Zhu
- The Marine Biomedical Research Institute of Guangdong Zhanjiang, School of Ocean and Tropical Medicine, Guangdong Medical University, Zhanjiang, China.
| | - Xiaorong Zhou
- Department of Immunology, School of Medicine, Nantong University, Nantong, China.
| |
Collapse
|
5
|
Zhang J, Wei X, Zhang Q, Jiao X, Li K, Geng M, Cao Y, Wang D, Cheng J, Yang J. Fish Uses CTLA-4 Immune Checkpoint to Suppress mTORC1-Controlled T-Cell Glycolysis and Immunity. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 212:1113-1128. [PMID: 38363204 DOI: 10.4049/jimmunol.2300599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Accepted: 01/26/2024] [Indexed: 02/17/2024]
Abstract
As an immune checkpoint, cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) suppresses the activation, proliferation, and effector function of T cells, thus preventing an overexuberant response and maintaining immune homeostasis. However, whether and how this immune checkpoint functions in early vertebrates remains unknown. In the current study, using a Nile tilapia (Oreochromis niloticus) model, we investigated the suppression of T cell response by CTLA-4 in bony fish. Tilapia CTLA-4 is constitutively expressed in lymphoid tissues, and its mRNA and protein expression in lymphocytes are upregulated following PHA stimulation or Edwardsiella piscicida infection. Blockade of CTLA-4 signaling enhanced T cell activation and proliferation but inhibited activation-induced T cell apoptosis, indicating that CTLA-4 negatively regulated T cell activation. In addition, blocking CTLA-4 signaling in vivo increased the differentiation potential and cytotoxicity of T cells, resulting in an enhanced T cell response during E. piscicida infection. Tilapia CTLA-4 competitively bound the B7.2/CD86 molecule with CD28, thus antagonizing the CD28-mediated costimulatory signal of T cell activation. Furthermore, inhibition of mammalian/mechanistic target of rapamycin complex 1 (mTORC1) signaling, c-Myc, or glycolysis markedly impaired the CTLA-4 blockade-enhanced T cell response, suggesting that CTLA-4 suppressed the T cell response of tilapia by inhibiting mTORC1/c-Myc axis-controlled glycolysis. Overall, the findings indicate a detailed mechanism by which CTLA-4 suppresses T cell immunity in tilapia; therefore, we propose that early vertebrates have evolved sophisticated mechanisms coupling immune checkpoints and metabolic reprogramming to avoid an overexuberant T cell response.
Collapse
Affiliation(s)
- Jiansong Zhang
- State Key Laboratory of Estuarine and Coastal Research, School of Life Sciences, East China Normal University, Shanghai, China
| | - Xiumei Wei
- State Key Laboratory of Estuarine and Coastal Research, School of Life Sciences, East China Normal University, Shanghai, China
| | - Qian Zhang
- State Key Laboratory of Estuarine and Coastal Research, School of Life Sciences, East China Normal University, Shanghai, China
| | - Xinying Jiao
- State Key Laboratory of Estuarine and Coastal Research, School of Life Sciences, East China Normal University, Shanghai, China
| | - Kang Li
- State Key Laboratory of Estuarine and Coastal Research, School of Life Sciences, East China Normal University, Shanghai, China
| | - Ming Geng
- State Key Laboratory of Estuarine and Coastal Research, School of Life Sciences, East China Normal University, Shanghai, China
| | - Yi Cao
- State Key Laboratory of Estuarine and Coastal Research, School of Life Sciences, East China Normal University, Shanghai, China
| | - Ding Wang
- State Key Laboratory of Estuarine and Coastal Research, School of Life Sciences, East China Normal University, Shanghai, China
| | - Jie Cheng
- State Key Laboratory of Estuarine and Coastal Research, School of Life Sciences, East China Normal University, Shanghai, China
| | - Jialong Yang
- State Key Laboratory of Estuarine and Coastal Research, School of Life Sciences, East China Normal University, Shanghai, China
- Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China
| |
Collapse
|
6
|
Otegui N, Houry M, Arozarena I, Serrano D, Redin E, Exposito F, Leon S, Valencia K, Montuenga L, Calvo A. Cancer Cell-Intrinsic Alterations Associated with an Immunosuppressive Tumor Microenvironment and Resistance to Immunotherapy in Lung Cancer. Cancers (Basel) 2023; 15:3076. [PMID: 37370686 PMCID: PMC10295869 DOI: 10.3390/cancers15123076] [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: 05/01/2023] [Revised: 05/24/2023] [Accepted: 06/01/2023] [Indexed: 06/29/2023] Open
Abstract
Despite the great clinical success of immunotherapy in lung cancer patients, only a small percentage of them (<40%) will benefit from this therapy alone or combined with other strategies. Cancer cell-intrinsic and cell-extrinsic mechanisms have been associated with a lack of response to immunotherapy. The present study is focused on cancer cell-intrinsic genetic, epigenetic, transcriptomic and metabolic alterations that reshape the tumor microenvironment (TME) and determine response or refractoriness to immune checkpoint inhibitors (ICIs). Mutations in KRAS, SKT11(LKB1), KEAP1 and TP53 and co-mutations of these genes are the main determinants of ICI response in non-small-cell lung cancer (NSCLC) patients. Recent insights into metabolic changes in cancer cells that impose restrictions on cytotoxic T cells and the efficacy of ICIs indicate that targeting such metabolic restrictions may favor therapeutic responses. Other emerging pathways for therapeutic interventions include epigenetic modulators and DNA damage repair (DDR) pathways, especially in small-cell lung cancer (SCLC). Therefore, the many potential pathways for enhancing the effect of ICIs suggest that, in a few years, we will have much more personalized medicine for lung cancer patients treated with immunotherapy. Such strategies could include vaccines and chimeric antigen receptor (CAR) cells.
Collapse
Affiliation(s)
- Nerea Otegui
- CCUN Cancer Center and Program in Solid Tumors, Center for Applied Medical Research (CIMA), University of Navarra, 31008 Pamplona, Spain; (N.O.); (M.H.); (D.S.); (S.L.); (K.V.); (L.M.)
- Department of Pathology, Anatomy and Physiology, School of Medicine, University of Navarra, 31008 Pamplona, Spain
| | - Maeva Houry
- CCUN Cancer Center and Program in Solid Tumors, Center for Applied Medical Research (CIMA), University of Navarra, 31008 Pamplona, Spain; (N.O.); (M.H.); (D.S.); (S.L.); (K.V.); (L.M.)
- Department of Pathology, Anatomy and Physiology, School of Medicine, University of Navarra, 31008 Pamplona, Spain
| | - Imanol Arozarena
- Instituto de Investigación Sanitaria de Navarra (IDISNA), 31008 Pamplona, Spain;
- Cancer Signaling Unit, Navarrabiomed, University Hospital of Navarra (HUN), Public University of Navarra (UPNA), 31008 Pamplona, Spain
| | - Diego Serrano
- CCUN Cancer Center and Program in Solid Tumors, Center for Applied Medical Research (CIMA), University of Navarra, 31008 Pamplona, Spain; (N.O.); (M.H.); (D.S.); (S.L.); (K.V.); (L.M.)
- Department of Pathology, Anatomy and Physiology, School of Medicine, University of Navarra, 31008 Pamplona, Spain
| | - Esther Redin
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA;
| | - Francisco Exposito
- Yale Cancer Center, New Haven, CT 06519, USA;
- Department of Pathology, Yale School of Medicine, New Haven, CT 06510, USA
| | - Sergio Leon
- CCUN Cancer Center and Program in Solid Tumors, Center for Applied Medical Research (CIMA), University of Navarra, 31008 Pamplona, Spain; (N.O.); (M.H.); (D.S.); (S.L.); (K.V.); (L.M.)
- Department of Pathology, Anatomy and Physiology, School of Medicine, University of Navarra, 31008 Pamplona, Spain
| | - Karmele Valencia
- CCUN Cancer Center and Program in Solid Tumors, Center for Applied Medical Research (CIMA), University of Navarra, 31008 Pamplona, Spain; (N.O.); (M.H.); (D.S.); (S.L.); (K.V.); (L.M.)
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), ISCIII, 28029 Madrid, Spain
| | - Luis Montuenga
- CCUN Cancer Center and Program in Solid Tumors, Center for Applied Medical Research (CIMA), University of Navarra, 31008 Pamplona, Spain; (N.O.); (M.H.); (D.S.); (S.L.); (K.V.); (L.M.)
- Department of Pathology, Anatomy and Physiology, School of Medicine, University of Navarra, 31008 Pamplona, Spain
- Instituto de Investigación Sanitaria de Navarra (IDISNA), 31008 Pamplona, Spain;
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), ISCIII, 28029 Madrid, Spain
| | - Alfonso Calvo
- CCUN Cancer Center and Program in Solid Tumors, Center for Applied Medical Research (CIMA), University of Navarra, 31008 Pamplona, Spain; (N.O.); (M.H.); (D.S.); (S.L.); (K.V.); (L.M.)
- Department of Pathology, Anatomy and Physiology, School of Medicine, University of Navarra, 31008 Pamplona, Spain
- Instituto de Investigación Sanitaria de Navarra (IDISNA), 31008 Pamplona, Spain;
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), ISCIII, 28029 Madrid, Spain
| |
Collapse
|
7
|
Xia C, Yin S, To KKW, Fu L. CD39/CD73/A2AR pathway and cancer immunotherapy. Mol Cancer 2023; 22:44. [PMID: 36859386 PMCID: PMC9979453 DOI: 10.1186/s12943-023-01733-x] [Citation(s) in RCA: 130] [Impact Index Per Article: 65.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Accepted: 01/26/2023] [Indexed: 03/03/2023] Open
Abstract
Cancer development is closely associated with immunosuppressive tumor microenvironment (TME) that attenuates antitumor immune responses and promotes tumor cell immunologic escape. The sequential conversion of extracellular ATP into adenosine by two important cell-surface ectonucleosidases CD39 and CD73 play critical roles in reshaping an immunosuppressive TME. The accumulated extracellular adenosine mediates its regulatory functions by binding to one of four adenosine receptors (A1R, A2AR, A2BR and A3R). The A2AR elicits its profound immunosuppressive function via regulating cAMP signaling. The increasing evidence suggests that CD39, CD73 and A2AR could be used as novel therapeutic targets for manipulating the antitumor immunity. In recent years, monoclonal antibodies or small molecule inhibitors targeting the CD39/CD73/A2AR pathway have been investigated in clinical trials as single agents or in combination with anti-PD-1/PD-L1 therapies. In this review, we provide an updated summary about the pathophysiological function of the adenosinergic pathway in cancer development, metastasis and drug resistance. The targeting of one or more components of the adenosinergic pathway for cancer therapy and circumvention of immunotherapy resistance are also discussed. Emerging biomarkers that may be used to guide the selection of CD39/CD73/A2AR-targeting treatment strategies for individual cancer patients is also deliberated.
Collapse
Affiliation(s)
- Chenglai Xia
- Affiliated Foshan Maternity and Child Healthcare Hospital, Southern Medical University, Foshan, 528000, China. .,School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 515150, China.
| | - Shuanghong Yin
- grid.284723.80000 0000 8877 7471Affiliated Foshan Maternity and Child Healthcare Hospital, Southern Medical University, Foshan, 528000 China ,grid.488530.20000 0004 1803 6191State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Esophageal Cancer Institute, Sun Yat-sen University Cancer Center, Guangzhou, 510060 China
| | - Kenneth K. W. To
- grid.10784.3a0000 0004 1937 0482School of Pharmacy, The Chinese University of Hong Kong, Hong Kong, China
| | - Liwu Fu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Esophageal Cancer Institute, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China.
| |
Collapse
|
8
|
Sarkar M, Nguyen T, Gundre E, Ogunlusi O, El-Sobky M, Giri B, Sarkar TR. Cancer-associated fibroblasts: The chief architect in the tumor microenvironment. Front Cell Dev Biol 2023; 11:1089068. [PMID: 36793444 PMCID: PMC9923123 DOI: 10.3389/fcell.2023.1089068] [Citation(s) in RCA: 66] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Accepted: 01/12/2023] [Indexed: 01/31/2023] Open
Abstract
Stromal heterogeneity of tumor microenvironment (TME) plays a crucial role in malignancy and therapeutic resistance. Cancer-associated fibroblasts (CAFs) are one of the major players in tumor stroma. The heterogeneous sources of origin and subsequent impacts of crosstalk with breast cancer cells flaunt serious challenges before current therapies to cure triple-negative breast cancer (TNBC) and other cancers. The positive and reciprocal feedback of CAFs to induce cancer cells dictates their mutual synergy in establishing malignancy. Their substantial role in creating a tumor-promoting niche has reduced the efficacy of several anti-cancer treatments, including radiation, chemotherapy, immunotherapy, and endocrine therapy. Over the years, there has been an emphasis on understanding CAF-induced therapeutic resistance in order to enhance cancer therapy results. CAFs, in the majority of cases, employ crosstalk, stromal management, and other strategies to generate resilience in surrounding tumor cells. This emphasizes the significance of developing novel strategies that target particular tumor-promoting CAF subpopulations, which will improve treatment sensitivity and impede tumor growth. In this review, we discuss the current understanding of the origin and heterogeneity of CAFs, their role in tumor progression, and altering the tumor response to therapeutic agents in breast cancer. In addition, we also discuss the potential and possible approaches for CAF-mediated therapies.
Collapse
Affiliation(s)
- Mrinmoy Sarkar
- Department of Biology, Texas A&M University, College Station, TX, United States
- Cancer Biology and Inflammatory Disorder Division, CSIR-Indian Institute of Chemical Biology, Kolkata, India
| | - Tristan Nguyen
- Department of Biology, Texas A&M University, College Station, TX, United States
| | - Esheksha Gundre
- Department of Biology, Texas A&M University, College Station, TX, United States
| | - Olajumoke Ogunlusi
- Department of Biology, Texas A&M University, College Station, TX, United States
| | - Mohanad El-Sobky
- Department of Biology, Texas A&M University, College Station, TX, United States
| | - Biplab Giri
- Department of Physiology, University of Gour Banga, English Bazar, India
| | - Tapasree Roy Sarkar
- Department of Biology, Texas A&M University, College Station, TX, United States
| |
Collapse
|
9
|
Liu Z, Liu X, Shen H, Xu X, Zhao X, Fu R. Adenosinergic axis and immune checkpoint combination therapy in tumor: A new perspective for immunotherapy strategy. Front Immunol 2022; 13:978377. [PMID: 36159861 PMCID: PMC9493240 DOI: 10.3389/fimmu.2022.978377] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2022] [Accepted: 08/23/2022] [Indexed: 11/21/2022] Open
Abstract
There are two figures and one table in this review, the review consists of 5823 words, without the description of figures and table, but including references. Tumor cells escape anti-tumor immune responses in various ways, including functionally shaping the microenvironment through the secretion of various chemokines and, cytokines. Adenosine is a powerful immunosuppressive metabolite, that is frequently elevated in the extracellular tumor microenvironment (TME). Thus, it has recently been proposed as a novel antitumor immunoassay for targeting adenosine- generating enzymes, such as CD39, CD73, and adenosine receptors. In recent years, the discovery of the immune checkpoints, such as programmed cell death 1(PD-1) and cytotoxic T lymphocyte antigen 4 (CTLA-4), has also greatly changed treatment methods and ideas for malignant tumors. Malignant tumor immunotherapy has been developed from point-to-point therapy targeting immune checkpoints, combining different points of different pathways to create a therapy based on the macroscopic immune regulatory system network. This article reviews the theoretical basis of the adenosine energy axis and immune checkpoint combined therapy for malignant tumors and the latest advances in malignant tumors.
Collapse
|
10
|
Chen X, Zhang W, Yang W, Zhou M, Liu F. Acquired resistance for immune checkpoint inhibitors in cancer immunotherapy: challenges and prospects. Aging (Albany NY) 2022; 14:1048-1064. [PMID: 35037899 PMCID: PMC8833108 DOI: 10.18632/aging.203833] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Accepted: 12/29/2021] [Indexed: 04/21/2023]
Abstract
Drug resistance has become an obstacle to the further development of immunotherapy in clinical applications and experimental studies. In the current review, the acquired resistance to immunotherapy was examined. The mechanisms of acquired resistance were based on three aspects as follows: The change of the tumor functions, the upregulated expression of inhibitory immune checkpoint proteins, and the effects of the tumor microenvironment. The combined use of immunotherapy and other therapies is performed to delay acquired resistance. A comprehensive understanding of acquired drug resistance may provide ideas for solving this dilemma.
Collapse
Affiliation(s)
- Xunrui Chen
- Department of Oncology, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 201900, China
| | - Wenhui Zhang
- Shanghai Institute of Precision Medicine, Shanghai 200125, China
| | - Wenyan Yang
- Medical Center, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 201900, China
| | - Min Zhou
- Department of Respirtory Medicine, Jinshan Branch of the Sixth People’s Hospital of Shanghai, Shanghai 201599, P.R. China
| | - Feng Liu
- Department of Oncology, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 201900, China
| |
Collapse
|
11
|
Anti-PD-1 therapy activates tumoricidic properties of NKT cells and contributes to the overall deceleration of tumor progression in a model of murine mammary carcinoma. VOJNOSANIT PREGL 2022. [DOI: 10.2298/vsp210126039j] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Background/Aim. Immune checkpoint therapy is a well-established therapeutic approach in the treatment of malignant diseases and is thought to be mostly based on facilitating the adaptive immune response. However, the cells of the innate immune response, such as natural killer T (NKT) cells, might also be important for a successful anti-programmed cell death protein-1 (anti-PD-1) therapy, as they initiate the antitumor immune response. The aim of this study was to investigate the influence of anti-PD-1 therapy on the immune response against tumors. Methods. For tumor induction, 4T1 cells synergic to BALB/c back-ground were used, after which mice underwent anti-PD-1 treatment. After the mice were sacrificed, NKT cells, dendritic cells (DCs), and macrophages derived from spleen and primary tumor tissue were analyzed using flow cytometry. Results. Anti-PD-1 therapy enhanced the expression of activating molecules CD69, NKp46, and NKG2D in NKT cells of the tumor and spleen. This therapy activated NKT cells directly and indirectly via DCs. Activated NKT cells acquired tumoricidic properties directly, by secreting perforin, and indirectly by stimulating M1 macrophages polarization. Conclusion. Anti-PD-1 therapy activates changes in DCs and macrophages of primary tumor tissue towards protumoricidic activity. Since anti-PD-1 therapy induces significant changes in NKT cells, DCs, and macrophages, the efficacy of the overall antitumor response is increased and has significantly decelerated tumor growth.
Collapse
|
12
|
Motofei IG. Nobel Prize for immune checkpoint inhibitors, understanding the immunological switching between immunosuppression and autoimmunity. Expert Opin Drug Saf 2021; 21:599-612. [PMID: 34937484 DOI: 10.1080/14740338.2022.2020243] [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: 10/19/2022]
Abstract
INTRODUCTION Immune checkpoint inhibitors (ICIs) are a revolutionary form of immunotherapy in cancer. However, the percentage of patients responding to therapy is relatively low, while adverse effects occur in a large number of patients. In addition, the therapeutic mechanisms of ICIs are not yet completely described. AREAS COVERED The initial view (articles published in PubMed, Scopus, Web of Science, etc.) was that ICIs increase tumor-specific immunity. Recent data (collected from the same databases) suggest that the ICIs pharmacotherapy actually extends beyond the topic of immune reactivity, including additional immune pathways, such as disrupting immunosuppression and increasing tumor-specific autoimmunity. Unfortunately, there is no clear delimitation between these specific autoimmune reactions that are therapeutically beneficial, and nonspecific autoimmune reactions/toxicity that can be extremely severe side effects. EXPERT OPINION Immune checkpoint mechanisms perform a non-selective immune regulation, maintaining a dynamic balance between immunosuppression and autoimmunity. By blocking these mechanisms, ICIs actually perform an immunological reset, decreasing immunosuppression and increasing tumor-specific immunity and predisposition to autoimmunity. The predisposition to autoimmunity induces both side effects and beneficial autoimmunity. Consequently, further studies are necessary to maximize the beneficial tumor-specific autoimmunity, while reducing the counterproductive effect of associated autoimmune toxicity.
Collapse
Affiliation(s)
- Ion G Motofei
- Department of Surgery/ Oncology, Carol Davila University, Bucharest, Romania.,Department of Surgery/ Oncology, St. Pantelimon Hospital, Bucharest, Romania
| |
Collapse
|
13
|
Precision medicine in clinical oncology: the journey from IgG antibody to IgE. Curr Opin Allergy Clin Immunol 2021; 20:282-289. [PMID: 32349107 DOI: 10.1097/aci.0000000000000637] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
PURPOSE OF REVIEW Cancer is one of the leading causes of death and the incidence rates are constantly rising. The heterogeneity of tumors poses a big challenge for the treatment of the disease and natural antibodies additionally affect disease progression. The introduction of engineered mAbs for anticancer immunotherapies has substantially improved progression-free and overall survival of cancer patients, but little efforts have been made to exploit other antibody isotypes than IgG. RECENT FINDINGS In order to improve these therapies, 'next-generation antibodies' were engineered to enhance a specific feature of classical antibodies and form a group of highly effective and precise therapy compounds. Advanced antibody approaches include among others antibody-drug conjugates, glyco-engineered and Fc-engineered antibodies, antibody fragments, radioimmunotherapy compounds, bispecific antibodies and alternative (non-IgG) immunoglobulin classes, especially IgE. SUMMARY The current review describes solutions for the needs of next-generation antibody therapies through different approaches. Careful selection of the best-suited engineering methodology is a key factor in developing personalized, more specific and more efficient mAbs against cancer to improve the outcomes of cancer patients. We highlight here the large evidence of IgE exploiting a highly cytotoxic effector arm as potential next-generation anticancer immunotherapy.
Collapse
|
14
|
Poma AM, Bruno R, Pietrini I, Alì G, Pasquini G, Proietti A, Vasile E, Cappelli S, Chella A, Fontanini G. Biomarkers and Gene Signatures to Predict Durable Response to Pembrolizumab in Non-Small Cell Lung Cancer. Cancers (Basel) 2021; 13:cancers13153828. [PMID: 34359727 PMCID: PMC8345106 DOI: 10.3390/cancers13153828] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 07/27/2021] [Accepted: 07/28/2021] [Indexed: 12/15/2022] Open
Abstract
Simple Summary Not all patients with advanced or metastatic non-small cell lung cancer (NSCLC) respond to pembrolizumab, even if their tumor expresses PD-L1. This is a monocentric study aimed at identifying potential predictive biomarkers for pembrolizumab first-line treatment. Tumor microenvironment was characterized by gene expression analysis in 46 tumor samples from 25 NSCLC patients with and 21 without durable clinical benefit. As expected, patients achieving clinical benefit had a greater infiltration of immune cells. In particular, CD8 T-cell and NK cell scores were strongly associated with durable benefit. Single immune cell markers such as XCL1/2 showed a high performance in predicting durable response to pembrolizumab with an AUC of 0.85. In the same series PD-L1 expression levels had an AUC equal to 0.61. Identified predictive biomarkers can improve patients’ selection, thus optimizing treatment definition. Abstract Pembrolizumab has been approved as first-line treatment for advanced Non-small cell lung cancer (NSCLC) patients with tumors expressing PD-L1 and in the absence of other targetable alterations. However, not all patients that meet these criteria have a durable benefit. In this monocentric study, we aimed at refining the selection of patients based on the expression of immune genes. Forty-six consecutive advanced NSCLC patients treated with pembrolizumab in first-line setting were enrolled. The expression levels of 770 genes involved in the regulation of the immune system was analysed by the nanoString system. PD-L1 expression was evaluated by immunohistochemistry. Patients with durable clinical benefit had a greater infiltration of cytotoxic cells, exhausted CD8, B-cells, CD45, T-cells, CD8 T-cells and NK cells. Immune cell scores such as CD8 T-cell and NK cell were good predictors of durable response with an AUC of 0.82. Among the immune cell markers, XCL1/2 showed the better performance in predicting durable benefit to pembrolizumab, with an AUC of 0.85. Additionally, CD8A, CD8B and EOMES showed a high specificity (>0.86) in identifying patients with a good response to treatment. In the same series, PD-L1 expression levels had an AUC of 0.61. The characterization of tumor microenvironment, even with the use of single markers, can improve patients’ selection for pembrolizumab treatment.
Collapse
Affiliation(s)
- Anello Marcello Poma
- Department of Surgical, Medical, Molecular Pathology and Critical Area, University of Pisa, via Savi 10, 56126 Pisa, Italy;
| | - Rossella Bruno
- Unit of Pathological Anatomy, University Hospital of Pisa, via Roma 67, 56126 Pisa, Italy; (R.B.); (G.A.); (A.P.)
| | - Iacopo Pietrini
- General Pathology, University of Pisa, via Savi 10, 56126 Pisa, Italy;
| | - Greta Alì
- Unit of Pathological Anatomy, University Hospital of Pisa, via Roma 67, 56126 Pisa, Italy; (R.B.); (G.A.); (A.P.)
| | - Giulia Pasquini
- Unit of Medical Oncology, San Jacopo Hospital of Pistoia, 51100 Pistoia, Italy;
| | - Agnese Proietti
- Unit of Pathological Anatomy, University Hospital of Pisa, via Roma 67, 56126 Pisa, Italy; (R.B.); (G.A.); (A.P.)
| | - Enrico Vasile
- Unit of Pneumology, University Hospital of Pisa, via Paradisa 2, 56126 Pisa, Italy; (E.V.); (S.C.); (A.C.)
| | - Sabrina Cappelli
- Unit of Pneumology, University Hospital of Pisa, via Paradisa 2, 56126 Pisa, Italy; (E.V.); (S.C.); (A.C.)
| | - Antonio Chella
- Unit of Pneumology, University Hospital of Pisa, via Paradisa 2, 56126 Pisa, Italy; (E.V.); (S.C.); (A.C.)
| | - Gabriella Fontanini
- Department of Surgical, Medical, Molecular Pathology and Critical Area, University of Pisa, via Savi 10, 56126 Pisa, Italy;
- Correspondence: ; Tel.: +39-050-992983
| |
Collapse
|
15
|
Tang Y, Wang T, Yu Y, Yan Y, Wu C. Upregulation of HOXC9 generates interferon-gamma resistance in gastric cancer by inhibiting the DAPK1/RIG1/STAT1 axis. Cancer Sci 2021; 112:3455-3468. [PMID: 34159686 PMCID: PMC8409412 DOI: 10.1111/cas.15043] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Revised: 05/28/2021] [Accepted: 06/19/2021] [Indexed: 12/14/2022] Open
Abstract
Clinical reports indicate that gastric cancer (GC) has a high mortality rate, but its pathological mechanism remains poorly understood. This work integrated bioinformatics analysis with experimental verification to explore novel biomarkers of gastric cancer. First, weighted gene coexpression network analysis was applied to screen significant genes correlated with GC development. Gene set enrichment analysis was also used to unearth the most relevant biological functions of significant genes. As a result, we discovered homeobox C9 (HOXC9) as a novel oncogene in GC, primarily through negatively regulating immune response. High expression of HOXC9 predicted a poor prognosis in GC patients, and knocking down HOXC9 efficiently enhanced the interferon‐gamma (IFNγ)‐dependent apoptosis in two GC cell lines as well as organoids from patients. Furthermore, cleaved caspase‐3/7 and phosphorylated signal transducer and activator of transcription 1 (p‐STAT1) were also significantly enhanced in HOXC9 knockdown cells and organoids treated with IFNγ. Mechanistically, we found that HOXC9 inhibited the death‐associated protein kinase 1 (DAPK1) and its downstream retinoic acid‐inducible gene‐I (RIG1) to generate GC IFNγ resistance. In summary, we identified and confirmed that HOXC9 generates IFNγ resistance in GC by inhibiting the DAPK1/RIG1/p‐STAT1 axis.
Collapse
Affiliation(s)
- Yuanxin Tang
- Department of General Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
| | - Taifang Wang
- Department of Radiation Oncology, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
| | - Yue Yu
- Department of Radiation Oncology, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
| | - Yuhao Yan
- Department of General Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
| | - Chunli Wu
- Department of Radiation Oncology, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
| |
Collapse
|
16
|
Calabrese LH, Caporali R, Blank CU, Kirk AD. Modulating the wayward T cell: New horizons with immune checkpoint inhibitor treatments in autoimmunity, transplant, and cancer. J Autoimmun 2020; 115:102546. [PMID: 32980229 DOI: 10.1016/j.jaut.2020.102546] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 09/02/2020] [Accepted: 09/14/2020] [Indexed: 12/20/2022]
Abstract
The T-cell response is regulated by the balance between costimulatory and coinhibitory signals. Immune checkpoints are essential for efficient T-cell activation, but also for maintaining self-tolerance and protecting tissues from damage caused by the immune system, and for providing protective immunity. Modulating immune checkpoints can serve diametric goals, such that blocking a coinhibitory molecule can unleash anti-cancer immunity whereas stimulating the same molecule can reduce an over-reaction in autoimmune disease. The purpose of this review is to examine the regulation of T-cell costimulation and coinhibition, which is central to the processes underpinning autoimmunity, transplant rejection and immune evasion in cancer. We will focus on the immunomodulation agents that regulate these unwanted over- and under-reactions. The use of such agents has led to control of symptoms and slowing of progression in patients with rheumatoid arthritis, reduced rejection rates in transplant patients, and prolonged survival in patients with cancer. The management of immune checkpoint inhibitor treatment in certain challenging patient populations, including patients with pre-existing autoimmune conditions or transplant patients who develop cancer, as well as the management of immune-related adverse events in patients receiving antitumor therapy, is examined. Finally, the future of immune checkpoint inhibitors, including examples of emerging targets that are currently in development, as well as recent insights gained using new molecular techniques, is discussed. T-cell costimulation and coinhibition play vital roles in these diverse therapeutic areas. Targeting immune checkpoints continues to be a powerful avenue for the development of agents suitable for treating autoimmune diseases and cancers and for improving transplant outcomes. Enhanced collaboration between therapy area specialists to share learnings across disciplines will improve our understanding of the opposing effects of treatments for autoimmune disease/transplant rejection versus cancer on immune checkpoints, which has the potential to lead to improved patient outcomes.
Collapse
Affiliation(s)
| | - Roberto Caporali
- University of Milan, Department of Clinical Sciences and Community Health and Rheumatology Division, ASST Pini-CTO Hospital, Milan, Italy
| | | | - Allan D Kirk
- Department of Surgery, Duke University School of Medicine, Durham, NC, United States
| |
Collapse
|
17
|
Han X, Alu A, Xiao Y, Wei Y, Wei X. Hyperprogression: A novel response pattern under immunotherapy. Clin Transl Med 2020; 10:e167. [PMID: 32997401 PMCID: PMC7510779 DOI: 10.1002/ctm2.167] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 08/19/2020] [Accepted: 08/20/2020] [Indexed: 02/05/2023] Open
Abstract
Checkpoint blockade therapy has shown significant therapeutic benefits and resulted in durable responses in patients with various tumors. However, accumulating evidence has demonstrated that 4-29% of all patients with cancers with various histologies may suffer from tumor flare following such therapy. This novel tumor response pattern, termed hyperprogression, is a potentially deleterious side effect of checkpoint blockade therapy that accelerates disease progression in a subset of patients. In this review, we describe possible immune checkpoint blockade biomarkers and the epidemiology, different definitions, and predictors of hyperprogression based on the research findings and further present the available evidence supporting pathophysiological hypotheses that might explain hyperprogression during checkpoint blockade therapy. We also compare hyperprogression and pseudoprogression. Finally, we discuss areas requiring further study.
Collapse
Affiliation(s)
- Xue‐jiao Han
- Laboratory of Aging Research and Cancer Drug TargetState Key Laboratory of BiotherapyNational Clinical Research Center for GeriatricsWest China HospitalSichuan UniversityChengduChina
| | - Aqu Alu
- Laboratory of Aging Research and Cancer Drug TargetState Key Laboratory of BiotherapyNational Clinical Research Center for GeriatricsWest China HospitalSichuan UniversityChengduChina
| | - Yi‐nan Xiao
- West China School of MedicineWest China HospitalSichuan UniversityChengduChina
| | - Yu‐quan Wei
- Laboratory of Aging Research and Cancer Drug TargetState Key Laboratory of BiotherapyNational Clinical Research Center for GeriatricsWest China HospitalSichuan UniversityChengduChina
| | - Xia‐wei Wei
- Laboratory of Aging Research and Cancer Drug TargetState Key Laboratory of BiotherapyNational Clinical Research Center for GeriatricsWest China HospitalSichuan UniversityChengduChina
| |
Collapse
|
18
|
Sepesi B, Cascone T, Chun SG, Altan M, Le X. Emerging Therapies in Thoracic Malignancies-Immunotherapy, Targeted Therapy, and T-Cell Therapy in Non-Small Cell Lung Cancer. Surg Oncol Clin N Am 2020; 29:555-569. [PMID: 32883458 PMCID: PMC7388816 DOI: 10.1016/j.soc.2020.06.009] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Affiliation(s)
- Boris Sepesi
- Department of Thoracic and Cardiovascular Surgery, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA.
| | - Tina Cascone
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA
| | - Stephen G Chun
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA
| | - Mehmet Altan
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA
| | - Xiuning Le
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA
| |
Collapse
|
19
|
De Vitis C, Corleone G, Salvati V, Ascenzi F, Pallocca M, De Nicola F, Fanciulli M, di Martino S, Bruschini S, Napoli C, Ricci A, Bassi M, Venuta F, Rendina EA, Ciliberto G, Mancini R. B4GALT1 Is a New Candidate to Maintain the Stemness of Lung Cancer Stem Cells. J Clin Med 2019; 8:E1928. [PMID: 31717588 PMCID: PMC6912435 DOI: 10.3390/jcm8111928] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Revised: 10/30/2019] [Accepted: 11/05/2019] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND According to the cancer stem cells (CSCs) hypothesis, a population of cancer cells with stem cell properties is responsible for tumor propagation, drug resistance, and disease recurrence. Study of the mechanisms responsible for lung CSCs propagation is expected to provide better understanding of cancer biology and new opportunities for therapy. METHODS The Lung Adenocarcinoma (LUAD) NCI-H460 cell line was grown either as 2D or as 3D cultures. Transcriptomic and genome-wide chromatin accessibility studies of 2D vs. 3D cultures were carried out using RNA-sequencing and Assay for Transposase Accessible Chromatin with high-throughput sequencing (ATAC-seq), respectively. Reverse transcription polymerase chain reaction (RT-PCR) was also carried out on RNA extracted from primary cultures derived from malignant pleural effusions to validate RNA-seq results. RESULTS RNA-seq and ATAC-seq data disentangled transcriptional and genome accessibility variability of 3D vs. 2D cultures in NCI-H460 cells. The examination of genomic landscape of genes upregulated in 3D vs. 2D cultures led to the identification of 2D cultures led to the identification of Beta-1,4-galactosyltranferase 1 (B4GALT1) as the top candidate. B4GALT1 as the top candidate. B4GALT1 was validated as a stemness factor, since its silencing caused strong inhibition of 3D spheroid formation. CONCLUSION Combined transcriptomic and chromatin accessibility study of 3D vs. 2D LUAD cultures led to the identification of B4GALT1 as a new factor involved in the propagation and maintenance of LUAD CSCs.
Collapse
Affiliation(s)
- Claudia De Vitis
- Department of Clinical and Molecular Medicine, Sant’Andrea Hospital, “Sapienza” University of Rome, 00161 Rome, Italy; (C.D.V.); (R.M.)
| | - Giacomo Corleone
- SAFU Laboratory, Department of Research, Advanced Diagnostic, and Technological Innovation, IRCCS “Regina Elena” National Cancer Institute, 00144 Rome, Italy; (G.C.); (M.P.); (F.D.N.); (M.F.)
| | - Valentina Salvati
- Preclinical Models and New Therapeutic Agents Unit, IRCCS-Regina Elena National Cancer Institute, 00144 Rome, Italy;
| | - Francesca Ascenzi
- Tumor Immunology and Immunotherapy Unit, Department of Research, Advanced Diagnostic and Technological Innovation, IRCCS Regina Elena National Cancer Institute, 00144 Rome, Italy;
| | - Matteo Pallocca
- SAFU Laboratory, Department of Research, Advanced Diagnostic, and Technological Innovation, IRCCS “Regina Elena” National Cancer Institute, 00144 Rome, Italy; (G.C.); (M.P.); (F.D.N.); (M.F.)
| | - Francesca De Nicola
- SAFU Laboratory, Department of Research, Advanced Diagnostic, and Technological Innovation, IRCCS “Regina Elena” National Cancer Institute, 00144 Rome, Italy; (G.C.); (M.P.); (F.D.N.); (M.F.)
| | - Maurizio Fanciulli
- SAFU Laboratory, Department of Research, Advanced Diagnostic, and Technological Innovation, IRCCS “Regina Elena” National Cancer Institute, 00144 Rome, Italy; (G.C.); (M.P.); (F.D.N.); (M.F.)
| | - Simona di Martino
- Pathology Unit, IRCSS “Regina Elena” National Cancer Institute, 00144 Rome, Italy;
| | - Sara Bruschini
- Department of Experimental and Clinical Medicine, Magna Graecia University of Catanzaro, 88100 Catanzaro, Italy;
| | - Christian Napoli
- Department of Medical Surgical Sciences and Translational Medicine, Sant’Andrea Hospital, “Sapienza” University of Rome, 00189 Rome, Italy;
| | - Alberto Ricci
- Department of Clinical and Molecular Medicine, Division of Pneumology, Sapienza University of Rome, Sant’Andrea Hospital, 00189 Rome, Italy;
| | - Massimiliano Bassi
- Department of Thoracic Surgery, University of Rome Sapienza, 00161 Rome, Italy; (M.B.); (F.V.)
| | - Federico Venuta
- Department of Thoracic Surgery, University of Rome Sapienza, 00161 Rome, Italy; (M.B.); (F.V.)
| | - Erino Angelo Rendina
- Department of Thoracic Surgery, Sant’Andrea Hospital, “Sapienza” University of Rome, 00189 Rome, Italy
| | - Gennaro Ciliberto
- Scientific Direction, IRCCS “Regina Elena” National Cancer Institute, 00144 Rome, Italy
| | - Rita Mancini
- Department of Clinical and Molecular Medicine, Sant’Andrea Hospital, “Sapienza” University of Rome, 00161 Rome, Italy; (C.D.V.); (R.M.)
| |
Collapse
|
20
|
Stüven AK, Wiedenmann B. Sustained partial remission of a metastatic NEN using off-label immunotherapy with pembrolizumab. Oncotarget 2019; 10:3302-3311. [PMID: 31143376 PMCID: PMC6524935 DOI: 10.18632/oncotarget.26906] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Accepted: 03/23/2019] [Indexed: 12/11/2022] Open
Abstract
Neuroendocrine neoplasms (NEN) are a heterogeneous group of tumors, which can be histologically separated by primary location, proliferation rate and differentiation of tumor cells. The therapeutic options and outcome depend on grading, staging and resectability of the tumor. Established treatment options of neuroendocrine tumors (NET) and carcinomas (NEC) are based especially on surgery, tumor specific medical treatments, peptide guided radioreceptor therapy (PRRT) and locoregional therapies. We report about a patient diagnosed with a pancreatic, non-functional NET/NEC G2/3 with a proliferation rate of 20% at initial immunohistochemical diagnosis. During the course of the disease, the proliferation rate increased up to more than 50% over a period of 5 years. Due to loss of response to established therapies (i.e. systemic chemotherapy, targeted therapy and brachytherapy), an off-label immunotherapy with the PD-1 antibody pembrolizumab was initiated based on a 30% PD-L1 expression in tumor cells. This report is the first demonstrating a partial remission of a pancreatic NEN using pembrolizumab monotherapy with a hepatic tumor volume reduction of at least 66%, combined with an improvement of the Karnofsky score rising from 60% to 100%. This case offers insight into the potential role of immunotherapy in a subgroup of neuroendocrine neoplasms.
Collapse
Affiliation(s)
- Anna Kathrin Stüven
- Department of Hepatology and Gastroenterology, Campus Virchow-Klinikum and Campus Charité Mitte, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Bertram Wiedenmann
- Department of Hepatology and Gastroenterology, Campus Virchow-Klinikum and Campus Charité Mitte, Charité-Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
21
|
Girard P, Charles J, Cluzel C, Degeorges E, Manches O, Plumas J, De Fraipont F, Leccia MT, Mouret S, Chaperot L, Aspord C. The features of circulating and tumor-infiltrating γδ T cells in melanoma patients display critical perturbations with prognostic impact on clinical outcome. Oncoimmunology 2019; 8:1601483. [PMID: 31413911 DOI: 10.1080/2162402x.2019.1601483] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Revised: 03/18/2019] [Accepted: 03/22/2019] [Indexed: 02/08/2023] Open
Abstract
γδT cells hold a pivotal role in tumor immunosurveillance through their prompt activation and cytokine secretion, their ability to kill tumor cells in an Human Leukocyte Antigen (HLA)-unrestricted manner, and their combination of features of both innate and adaptive immunity. These unique properties and functional plasticity render them very attractive both as targets and vectors for cancer immunotherapy. Yet, these potent and fascinating antitumor effectors have not been extensively explored in melanoma. We provided here a detailed investigation of the phenotypic and functional properties of circulating and tumor-infiltrating γδT cells in melanoma patients, and their impact on clinical evolution. High proportions of circulating- and tumor-infiltrating γδT and δ2+ subset were associated with better clinical outcome. We reported however that circulating and tumor-infiltrating γδT cells from melanoma patients displayed an altered expression of NCR, KIR, and immune checkpoints, and identified NKp44, PD1, 41BB/41BBL, TIM3, and LAG3 as crucial checkpoints allowing immune escape and tumor progression. Notably, melanoma drastically impaired the ability of γδT cells to exhibit activation molecules, secrete cytokines, and display cytotoxicity toward melanoma in response to stimulation with phosphoantigens. It drove them toward regulatory and Th17 profiles associated with poor clinical outcomes. Our study highlights that melanoma hijacked γδT cells to escape from immune control, and revealed that circulating and tumor-infiltrating γδT cell features are promising potential biomarkers of clinical evolution. Such understanding of the physiopathology of γδT cells may help designing new therapeutic approaches exploiting the antitumor potential of γδT cells while counteracting their skewing by tumors to improve patient outcomes.
Collapse
Affiliation(s)
- Pauline Girard
- Etablissement Français du Sang Auvergne Rhone-Alpes, R&D-Laboratory, Grenoble, France.,University Grenoble Alpes, EMR EFS-UGA-INSERM U1209- CNRS, Immunobiology & Immunotherapy of Chronic Diseases, Grenoble, France
| | - Julie Charles
- University Grenoble Alpes, EMR EFS-UGA-INSERM U1209- CNRS, Immunobiology & Immunotherapy of Chronic Diseases, Grenoble, France.,Dermatology clinic, Grenoble University Hospital, Grenoble, France
| | - Camille Cluzel
- Etablissement Français du Sang Auvergne Rhone-Alpes, R&D-Laboratory, Grenoble, France.,University Grenoble Alpes, EMR EFS-UGA-INSERM U1209- CNRS, Immunobiology & Immunotherapy of Chronic Diseases, Grenoble, France
| | - Emmanuelle Degeorges
- Etablissement Français du Sang Auvergne Rhone-Alpes, R&D-Laboratory, Grenoble, France.,University Grenoble Alpes, EMR EFS-UGA-INSERM U1209- CNRS, Immunobiology & Immunotherapy of Chronic Diseases, Grenoble, France
| | - Olivier Manches
- Etablissement Français du Sang Auvergne Rhone-Alpes, R&D-Laboratory, Grenoble, France.,University Grenoble Alpes, EMR EFS-UGA-INSERM U1209- CNRS, Immunobiology & Immunotherapy of Chronic Diseases, Grenoble, France
| | - Joel Plumas
- Etablissement Français du Sang Auvergne Rhone-Alpes, R&D-Laboratory, Grenoble, France.,University Grenoble Alpes, EMR EFS-UGA-INSERM U1209- CNRS, Immunobiology & Immunotherapy of Chronic Diseases, Grenoble, France.,pDCline Pharma, Grenoble, France
| | - Florence De Fraipont
- University Grenoble Alpes, EMR EFS-UGA-INSERM U1209- CNRS, Immunobiology & Immunotherapy of Chronic Diseases, Grenoble, France.,Department of Biochemistry of Cancers and Biotherapies, Grenoble University Hospital, Grenoble, France
| | - Marie-Therese Leccia
- University Grenoble Alpes, EMR EFS-UGA-INSERM U1209- CNRS, Immunobiology & Immunotherapy of Chronic Diseases, Grenoble, France.,Dermatology clinic, Grenoble University Hospital, Grenoble, France
| | - Stephane Mouret
- University Grenoble Alpes, EMR EFS-UGA-INSERM U1209- CNRS, Immunobiology & Immunotherapy of Chronic Diseases, Grenoble, France.,Dermatology clinic, Grenoble University Hospital, Grenoble, France
| | - Laurence Chaperot
- Etablissement Français du Sang Auvergne Rhone-Alpes, R&D-Laboratory, Grenoble, France.,University Grenoble Alpes, EMR EFS-UGA-INSERM U1209- CNRS, Immunobiology & Immunotherapy of Chronic Diseases, Grenoble, France
| | - Caroline Aspord
- Etablissement Français du Sang Auvergne Rhone-Alpes, R&D-Laboratory, Grenoble, France.,University Grenoble Alpes, EMR EFS-UGA-INSERM U1209- CNRS, Immunobiology & Immunotherapy of Chronic Diseases, Grenoble, France
| |
Collapse
|
22
|
Hamm CA, Pry K, Lu J, Bacus S. Immune profiling reveals the diverse nature of the immune response in NSCLC and reveals signaling pathways that may influence the anti-tumor immune response. Exp Mol Pathol 2019; 109:1-15. [PMID: 30953647 DOI: 10.1016/j.yexmp.2019.04.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Revised: 02/19/2019] [Accepted: 04/02/2019] [Indexed: 12/18/2022]
Abstract
Recent FDA approvals of immunotherapy for NSCLC provide patients new treatment options, and these approvals also highlight the importance of the immune response in cancer treatment. While immunotherapy provides patients a new treatment option, the therapy is effective in less than half of the treated patients. To attain greater insight into the tumor-immune microenvironment, NSCLC tumors were analyzed by IHC and RNA-seq. IHC was used to identify NSCLC tumors that contain low, moderate, or high levels of CD8+ positive cells as a manifestation of an active anti-tumor immune response. Gene expression analysis identified an emergent gene signature that is associated with high and moderate levels of CD8 in NSCLC. In addition, the NSCLC tumors also express a unique combination of genes that may indicate complex anti-tumor immune responses (INFG-related genes, STATs, CXCL9, OX40, PD-L1, PD-L2, IDO1, and CD47). Several NSCLC tumors also express the immune checkpoint PD-L1 and at least one additional immune inhibitory molecule (IDO1, PD-L2, or others), which may explain the lack of a therapeutic response to treatments that disrupt only one immune checkpoint pathway.
Collapse
Affiliation(s)
- Christopher A Hamm
- GoPath Laboratories, 1351 Barclay Blvd, Buffalo Grove, IL 60089, United States of America.
| | - Karen Pry
- GoPath Laboratories, 1351 Barclay Blvd, Buffalo Grove, IL 60089, United States of America
| | - Jim Lu
- GoPath Laboratories, 1351 Barclay Blvd, Buffalo Grove, IL 60089, United States of America
| | - Sarah Bacus
- GoPath Laboratories, 1351 Barclay Blvd, Buffalo Grove, IL 60089, United States of America
| |
Collapse
|
23
|
Kather JN, Halama N. Harnessing the innate immune system and local immunological microenvironment to treat colorectal cancer. Br J Cancer 2019; 120:871-882. [PMID: 30936499 PMCID: PMC6734657 DOI: 10.1038/s41416-019-0441-6] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Revised: 02/20/2019] [Accepted: 03/05/2019] [Indexed: 12/14/2022] Open
Abstract
Significant progress in the development of new immunotherapies has led to successful clinical trials for malignant melanoma and non-small cell lung cancer; however, for the majority of solid tumours of the gastrointestinal tract, little or no progress has been seen. The efficacy of immunotherapies is limited by the complexities of a diverse set of immune cells, and interactions between the tumour cells and all other cells in the local microenvironment of solid tumours. A large fraction of immune cells present in and around solid tumours derive from the innate arm of the immune system and using these cells against tumours offers an alternative immunotherapeutic option, especially as current strategies largely harness the adaptive arm of the immune system. This option is currently being investigated and attempts at using the innate immune system for gastrointestinal cancers are showing initial results. Several important factors, including cytokines, chemotherapeutics and the microbiome, influence the plasticity and functionality of innate (myeloid) cells in the microenvironment, and this complexity of regulation has limited translation into successful trials so far. In this review, current concepts of the immunobiology of the innate arm in the tumour microenvironment are presented in the context of clinical translation.
Collapse
Affiliation(s)
- Jakob Nikolas Kather
- Department of Medical Oncology and Internal Medicine VI, National Center for Tumor Diseases, University Hospital Heidelberg, Heidelberg, Germany.,German Translational Cancer Consortium (DKTK), Heidelberg, Germany.,Applied Tumor Immunity, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Niels Halama
- Department of Medical Oncology and Internal Medicine VI, National Center for Tumor Diseases, University Hospital Heidelberg, Heidelberg, Germany. .,Institute for Immunology, University Hospital Heidelberg, Heidelberg, Germany. .,Department of Translational Immunotherapy, German Cancer Research Center (DKFZ), Heidelberg, Germany. .,Helmholtz Institute for Translational Oncology (HI-TRON), Mainz, Germany.
| |
Collapse
|
24
|
Englinger B, Pirker C, Heffeter P, Terenzi A, Kowol CR, Keppler BK, Berger W. Metal Drugs and the Anticancer Immune Response. Chem Rev 2018; 119:1519-1624. [DOI: 10.1021/acs.chemrev.8b00396] [Citation(s) in RCA: 174] [Impact Index Per Article: 24.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Bernhard Englinger
- Institute of Cancer Research and Comprehensive Cancer Center, Department of Medicine I, Medical University of Vienna, Borschkegasse 8a, A-1090 Vienna, Austria
| | - Christine Pirker
- Institute of Cancer Research and Comprehensive Cancer Center, Department of Medicine I, Medical University of Vienna, Borschkegasse 8a, A-1090 Vienna, Austria
| | - Petra Heffeter
- Institute of Cancer Research and Comprehensive Cancer Center, Department of Medicine I, Medical University of Vienna, Borschkegasse 8a, A-1090 Vienna, Austria
- Research Cluster “Translational Cancer Therapy Research”, University of Vienna and Medical University of Vienna, Vienna, Austria
| | - Alessio Terenzi
- Research Cluster “Translational Cancer Therapy Research”, University of Vienna and Medical University of Vienna, Vienna, Austria
- Institute of Inorganic Chemistry, Faculty of Chemistry, University of Vienna, Waehringer Strasse 42, A-1090 Vienna, Austria
| | - Christian R. Kowol
- Research Cluster “Translational Cancer Therapy Research”, University of Vienna and Medical University of Vienna, Vienna, Austria
- Institute of Inorganic Chemistry, Faculty of Chemistry, University of Vienna, Waehringer Strasse 42, A-1090 Vienna, Austria
| | - Bernhard K. Keppler
- Research Cluster “Translational Cancer Therapy Research”, University of Vienna and Medical University of Vienna, Vienna, Austria
- Institute of Inorganic Chemistry, Faculty of Chemistry, University of Vienna, Waehringer Strasse 42, A-1090 Vienna, Austria
| | - Walter Berger
- Institute of Cancer Research and Comprehensive Cancer Center, Department of Medicine I, Medical University of Vienna, Borschkegasse 8a, A-1090 Vienna, Austria
- Research Cluster “Translational Cancer Therapy Research”, University of Vienna and Medical University of Vienna, Vienna, Austria
| |
Collapse
|
25
|
Vogt TJ, Gevensleben H, Dietrich J, Kristiansen G, Bootz F, Landsberg J, Goltz D, Dietrich D. Detailed analysis of adenosine A2a receptor ( ADORA2A) and CD73 (5'-nucleotidase, ecto, NT5E) methylation and gene expression in head and neck squamous cell carcinoma patients. Oncoimmunology 2018; 7:e1452579. [PMID: 30221045 PMCID: PMC6136855 DOI: 10.1080/2162402x.2018.1452579] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2017] [Revised: 03/07/2018] [Accepted: 03/09/2018] [Indexed: 12/15/2022] Open
Abstract
Background: The adenosine A2a receptor (A2aR) and the adenosine synthesizing enzyme CD73 have recently evolved as a novel immunotherapeutic target. However, little is known about epigenetic modification of the encoding genes ADORA2A and NT5E. Methods: In the present study, we evaluated methylation at 23 loci of ADORA2A and 17 loci of NT5E with regard to transcriptional activity, human papilloma virus (HPV) status, immune cell infiltration, and outcome in a cohort of 279 head and neck squamous carcinoma (HNSCC) patients obtained from The Cancer Genome Atlas (TCGA). Methylation and mRNA expression were generated by the Infinium HumanMethylation450 BeadChip and Illumina HiSeq 2000 RNA Sequencing Version 2 analysis (Illumina, Inc., San Diego, CA, USA). HPV status was assessed by RNA-Seq data analysis of the viral genes E6 and E7. Results: Thirteen out of 23 ADORA2A loci and 15/17 NT5E loci were significantly correlated with mRNA levels (p < 0.05). Inverse correlations were predominately found in promoter regions, while positive correlations were more profound at intragenic loci. ADORA2A hypermethylation was significantly associated with poor overall survival (OS, p ≤ 0.030), whereas NT5E hypomethylation was associated with decreased OS in HPV-positive tumors (p ≤ 0.024) and increased OS in HPV-negative HNSCC (p ≤ 0.029). Further, we found significant correlations between methylation and immune cell infiltrates. Conclusion: Our data might point towards a significant role of the A2aR/CD73 axis during cancer progression in HNSCC.
Collapse
Affiliation(s)
- Timo J. Vogt
- Department of Otolaryngology, Head and Neck Surgery, University Hospital Bonn, Bonn, Germany
| | | | - Jörn Dietrich
- Department of Otolaryngology, Head and Neck Surgery, University Hospital Bonn, Bonn, Germany
| | | | - Friedrich Bootz
- Department of Otolaryngology, Head and Neck Surgery, University Hospital Bonn, Bonn, Germany
| | | | | | - Dimo Dietrich
- Department of Otolaryngology, Head and Neck Surgery, University Hospital Bonn, Bonn, Germany
| |
Collapse
|
26
|
Wesley EM, Xin G, McAllister D, Malarkannan S, Newman DK, Dwinell MB, Cui W, Johnson BD, Riese MJ. Diacylglycerol kinase ζ (DGKζ) and Casitas b-lineage proto-oncogene b-deficient mice have similar functional outcomes in T cells but DGKζ-deficient mice have increased T cell activation and tumor clearance. Immunohorizons 2018; 2:107-118. [PMID: 30027154 DOI: 10.4049/immunohorizons.1700055] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Targeting negative regulators downstream of the T cell receptor (TCR) represents a novel strategy to improve cancer immunotherapy. Two proteins that serve as critical inhibitory regulators downstream of the TCR are diacylglycerol kinase ζ (DGKζ), a regulator of Ras and PKC-θ signaling, and Casitas b-lineage proto-oncogene b (Cbl-b), an E3 ubiquitin ligase that predominantly regulates PI(3)K signaling. We sought to compare the signaling and functional effects that result from deletion of DGKζ, Cbl-b, or both (double knockout, DKO) in T cells, and to evaluate tumor responses generated in a clinically relevant orthotopic pancreatic tumor model. We found that whereas deletion of Cbl-b primarily served to enhance NF-κB signaling, deletion of DGKζ enhanced TCR-mediated signal transduction downstream of Ras/Erk and NF-κB. Deletion of DGKζ or Cbl-b comparably enhanced CD8+ T cell functional responses, such as proliferation, production of IFNγ, and generation of granzyme B when compared with WT T cells. DKO T cells demonstrated enhanced function above that observed with single knockout T cells after weak, but not strong, stimulation. Deletion of DGKζ, but not Cbl-b, however, resulted in significant increases in numbers of activated (CD44hi) CD8+ T cells in both non-treated and tumor-bearing mice. DGKζ-deficient mice also had enhanced control of pancreatic tumor cell growth compared to Cbl-b-deficient mice. This represents the first direct comparison between mice of these genotypes and suggests that T cell immunotherapies may be better improved by targeting TCR signaling molecules that are regulated by DGKζ as opposed to molecules regulated by Cbl-b.
Collapse
Affiliation(s)
- Erin M Wesley
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI
| | - Gang Xin
- Blood Research Institute, Blood Center of Wisconsin, Milwaukee, WI
| | - Donna McAllister
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI
| | - Subramaniam Malarkannan
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI.,Blood Research Institute, Blood Center of Wisconsin, Milwaukee, WI.,Division of Hematology/Oncology, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI.,Division of Hematology/Oncology, Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI
| | - Debra K Newman
- Blood Research Institute, Blood Center of Wisconsin, Milwaukee, WI.,Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, WI
| | - Michael B Dwinell
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI
| | - Weiguo Cui
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI.,Blood Research Institute, Blood Center of Wisconsin, Milwaukee, WI
| | - Bryon D Johnson
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI.,Division of Hematology/Oncology, Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI
| | - Matthew J Riese
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI.,Blood Research Institute, Blood Center of Wisconsin, Milwaukee, WI.,Division of Hematology/Oncology, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI
| |
Collapse
|
27
|
Optimizing the Targeting of Mouse Parvovirus 1 to Murine Melanoma Selects for Recombinant Genomes and Novel Mutations in the Viral Capsid Gene. Viruses 2018; 10:v10020054. [PMID: 29385689 PMCID: PMC5850361 DOI: 10.3390/v10020054] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2017] [Revised: 01/23/2018] [Accepted: 01/27/2018] [Indexed: 12/15/2022] Open
Abstract
Combining virus-enhanced immunogenicity with direct delivery of immunomodulatory molecules would represent a novel treatment modality for melanoma, and would require development of new viral vectors capable of targeting melanoma cells preferentially. Here we explore the use of rodent protoparvoviruses targeting cells of the murine melanoma model B16F10. An uncloned stock of mouse parvovirus 1 (MPV1) showed some efficacy, which was substantially enhanced following serial passage in the target cell. Molecular cloning of the genes of both starter and selected virus pools revealed considerable sequence diversity. Chimera analysis mapped the majority of the improved infectivity to the product of the major coat protein gene, VP2, in which linked blocks of amino acid changes and one or other of two apparently spontaneous mutations were selected. Intragenic chimeras showed that these represented separable components, both contributing to enhanced infection. Comparison of biochemical parameters of infection by clonal viruses indicated that the enhancement due to changes in VP2 operates after the virus has bound to the cell surface and penetrated into the cell. Construction of an in silico homology model for MPV1 allowed placement of these changes within the capsid shell, and revealed aspects of the capsid involved in infection initiation that had not been previously recognized.
Collapse
|
28
|
Abstract
As of May 1, 2017, 74 antibody-based molecules have been approved by a regulatory authority in a major market. Additionally, there are 70 and 575 antibody-based molecules in phase III and phase I/II clinical trials, respectively. These total 719 antibody-based clinical stage molecules include 493 naked IgGs, 87 antibody-drug conjugates, 61 bispecific antibodies, 37 total Fc fusion proteins, 17 radioimmunoglobulins, 13 antibody fragments, and 11 immunocytokines. New uses for these antibodies are being discovered each year. For oncology, many of the exciting new approaches involve antibody modulation of T-cells. There are over 80 antibodies in clinical trials targeting T cell checkpoints, 26 T-cell-redirected bispecific antibodies, and 145 chimeric antigen receptor (CAR) cell-based candidates (all currently in phase I or II clinical trials), totaling more than 250 T cell interacting clinical stage antibody-based candidates. Finally, significant progress has been made recently on routes of delivery, including delivery of proteins across the blood-brain barrier, oral delivery to the gut, delivery to the cellular cytosol, and gene- and viral-based delivery of antibodies. Thus, there are currently at least 864 antibody-based clinical stage molecules or cells, with incredible diversity in how they are constructed and what activities they impart. These are followed by a next wave of novel molecules, approaches, and new methods and routes of delivery, demonstrating that the field of antibody-based biologics is very innovative and diverse in its approaches to fulfill their promise to treat unmet medical needs.
Collapse
|
29
|
Téglási V, Reiniger L, Fábián K, Pipek O, Csala I, Bagó AG, Várallyai P, Vízkeleti L, Rojkó L, Tímár J, Döme B, Szállási Z, Swanton C, Moldvay J. Evaluating the significance of density, localization, and PD-1/PD-L1 immunopositivity of mononuclear cells in the clinical course of lung adenocarcinoma patients with brain metastasis. Neuro Oncol 2017; 19:1058-1067. [PMID: 28201746 PMCID: PMC5570158 DOI: 10.1093/neuonc/now309] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Management of lung cancer patients who suffer from brain metastases represents a major challenge. Considering the promising results with immune checkpoint inhibitor treatment, evaluating the status of immune cell (IC) infiltrates in the prognosis of brain metastasis may lead to better therapeutic strategies with these agents. The aim of this study was to characterize the distribution of ICs and determine the expression of the checkpoint molecules programmed death protein 1 (PD-1) and its ligand, PD-L1, in brain metastasis of lung adenocarcinoma (LUAD) patients and to analyze their clinicopathological correlations. METHODS We determined the presence of peritumoral mononuclear cells (mononuclear ring) and the density of intratumoral stromal mononuclear cells on brain metastasis tissue sections of 208 LUAD patients. PD-L1/PD-1 expressions were analyzed by immunohistochemistry. RESULTS Mononuclear rings were significantly associated with better survival after brain metastasis surgery. Cases with massive stromal IC infiltration also showed a tendency for better overall survival. Lower expression of PD-1 and PD-L1 was associated with better survival in patients who underwent surgery for the primary tumor and had multiple brain metastases. Steroid administration and chemotherapy appear not to influence the density of IC in brain metastasis. CONCLUSION This is the first study demonstrating the independent prognostic value of mononuclear rings in LUAD cases with brain metastasis. Our results also suggest that the density of tumor-associated ICs in addition to PD-L1 expression of tumor cells and ICs as well as PD-1 expression of ICs may hold relevant information for the appropriate selection of patients who might benefit from anti-PD-L1 or anti-PD-1 therapy.
Collapse
Affiliation(s)
- Vanda Téglási
- First Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary; MTA-SE NAP, Brain Metastasis Research Group, Hungarian Academy of Sciences, Second Department of Pathology, Semmelweis University, Budapest, Hungary; Department of Pulmonology, Semmelweis University, Budapest, Hungary; Department of Physics of Complex Systems, Eötvös Loránd University, Budapest, Hungary; Institute of Behavioural Sciences, Semmelweis University, Budapest, Hungary; Department of Neurooncology, National Institute of Clinical Neurosciences, Budapest, Hungary; Department of Radiology, National Institute of Clinical Neurosciences, Budapest, Hungary; Sixth Department of Pulmonology, National Korányi Institute of Pulmonology, Budapest, Hungary; Hungarian Academy of Sciences-Semmelweis University, Molecular Oncology Research Unit, Budapest, Hungary; Department of Tumor Biology, National Korányi Institute of Pulmonology-Semmelweis University, Budapest, Hungary; Department of Thoracic Surgery, National Institute of Oncology-Semmelweis University, Budapest, Hungary; Division of Thoracic Surgery, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria; Children's Hospital Informatics Program at the Harvard-Massachusetts Institute of Technology Division of Health Sciences and Technology, Harvard Medical School, Boston, Massachusetts, USA; Center for Biological Sequence Analysis, Department of Systems Biology, Technical University of Denmark, Lyngby, Denmark; CRUK Lung Cancer Centre of Excellence, UCL Cancer Institute, London, UK; Francis Crick Institute, London, UK
| | - Lilla Reiniger
- First Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary; MTA-SE NAP, Brain Metastasis Research Group, Hungarian Academy of Sciences, Second Department of Pathology, Semmelweis University, Budapest, Hungary; Department of Pulmonology, Semmelweis University, Budapest, Hungary; Department of Physics of Complex Systems, Eötvös Loránd University, Budapest, Hungary; Institute of Behavioural Sciences, Semmelweis University, Budapest, Hungary; Department of Neurooncology, National Institute of Clinical Neurosciences, Budapest, Hungary; Department of Radiology, National Institute of Clinical Neurosciences, Budapest, Hungary; Sixth Department of Pulmonology, National Korányi Institute of Pulmonology, Budapest, Hungary; Hungarian Academy of Sciences-Semmelweis University, Molecular Oncology Research Unit, Budapest, Hungary; Department of Tumor Biology, National Korányi Institute of Pulmonology-Semmelweis University, Budapest, Hungary; Department of Thoracic Surgery, National Institute of Oncology-Semmelweis University, Budapest, Hungary; Division of Thoracic Surgery, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria; Children's Hospital Informatics Program at the Harvard-Massachusetts Institute of Technology Division of Health Sciences and Technology, Harvard Medical School, Boston, Massachusetts, USA; Center for Biological Sequence Analysis, Department of Systems Biology, Technical University of Denmark, Lyngby, Denmark; CRUK Lung Cancer Centre of Excellence, UCL Cancer Institute, London, UK; Francis Crick Institute, London, UK
| | - Katalin Fábián
- First Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary; MTA-SE NAP, Brain Metastasis Research Group, Hungarian Academy of Sciences, Second Department of Pathology, Semmelweis University, Budapest, Hungary; Department of Pulmonology, Semmelweis University, Budapest, Hungary; Department of Physics of Complex Systems, Eötvös Loránd University, Budapest, Hungary; Institute of Behavioural Sciences, Semmelweis University, Budapest, Hungary; Department of Neurooncology, National Institute of Clinical Neurosciences, Budapest, Hungary; Department of Radiology, National Institute of Clinical Neurosciences, Budapest, Hungary; Sixth Department of Pulmonology, National Korányi Institute of Pulmonology, Budapest, Hungary; Hungarian Academy of Sciences-Semmelweis University, Molecular Oncology Research Unit, Budapest, Hungary; Department of Tumor Biology, National Korányi Institute of Pulmonology-Semmelweis University, Budapest, Hungary; Department of Thoracic Surgery, National Institute of Oncology-Semmelweis University, Budapest, Hungary; Division of Thoracic Surgery, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria; Children's Hospital Informatics Program at the Harvard-Massachusetts Institute of Technology Division of Health Sciences and Technology, Harvard Medical School, Boston, Massachusetts, USA; Center for Biological Sequence Analysis, Department of Systems Biology, Technical University of Denmark, Lyngby, Denmark; CRUK Lung Cancer Centre of Excellence, UCL Cancer Institute, London, UK; Francis Crick Institute, London, UK
| | - Orsolya Pipek
- First Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary; MTA-SE NAP, Brain Metastasis Research Group, Hungarian Academy of Sciences, Second Department of Pathology, Semmelweis University, Budapest, Hungary; Department of Pulmonology, Semmelweis University, Budapest, Hungary; Department of Physics of Complex Systems, Eötvös Loránd University, Budapest, Hungary; Institute of Behavioural Sciences, Semmelweis University, Budapest, Hungary; Department of Neurooncology, National Institute of Clinical Neurosciences, Budapest, Hungary; Department of Radiology, National Institute of Clinical Neurosciences, Budapest, Hungary; Sixth Department of Pulmonology, National Korányi Institute of Pulmonology, Budapest, Hungary; Hungarian Academy of Sciences-Semmelweis University, Molecular Oncology Research Unit, Budapest, Hungary; Department of Tumor Biology, National Korányi Institute of Pulmonology-Semmelweis University, Budapest, Hungary; Department of Thoracic Surgery, National Institute of Oncology-Semmelweis University, Budapest, Hungary; Division of Thoracic Surgery, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria; Children's Hospital Informatics Program at the Harvard-Massachusetts Institute of Technology Division of Health Sciences and Technology, Harvard Medical School, Boston, Massachusetts, USA; Center for Biological Sequence Analysis, Department of Systems Biology, Technical University of Denmark, Lyngby, Denmark; CRUK Lung Cancer Centre of Excellence, UCL Cancer Institute, London, UK; Francis Crick Institute, London, UK
| | - Irén Csala
- First Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary; MTA-SE NAP, Brain Metastasis Research Group, Hungarian Academy of Sciences, Second Department of Pathology, Semmelweis University, Budapest, Hungary; Department of Pulmonology, Semmelweis University, Budapest, Hungary; Department of Physics of Complex Systems, Eötvös Loránd University, Budapest, Hungary; Institute of Behavioural Sciences, Semmelweis University, Budapest, Hungary; Department of Neurooncology, National Institute of Clinical Neurosciences, Budapest, Hungary; Department of Radiology, National Institute of Clinical Neurosciences, Budapest, Hungary; Sixth Department of Pulmonology, National Korányi Institute of Pulmonology, Budapest, Hungary; Hungarian Academy of Sciences-Semmelweis University, Molecular Oncology Research Unit, Budapest, Hungary; Department of Tumor Biology, National Korányi Institute of Pulmonology-Semmelweis University, Budapest, Hungary; Department of Thoracic Surgery, National Institute of Oncology-Semmelweis University, Budapest, Hungary; Division of Thoracic Surgery, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria; Children's Hospital Informatics Program at the Harvard-Massachusetts Institute of Technology Division of Health Sciences and Technology, Harvard Medical School, Boston, Massachusetts, USA; Center for Biological Sequence Analysis, Department of Systems Biology, Technical University of Denmark, Lyngby, Denmark; CRUK Lung Cancer Centre of Excellence, UCL Cancer Institute, London, UK; Francis Crick Institute, London, UK
| | - Attila G Bagó
- First Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary; MTA-SE NAP, Brain Metastasis Research Group, Hungarian Academy of Sciences, Second Department of Pathology, Semmelweis University, Budapest, Hungary; Department of Pulmonology, Semmelweis University, Budapest, Hungary; Department of Physics of Complex Systems, Eötvös Loránd University, Budapest, Hungary; Institute of Behavioural Sciences, Semmelweis University, Budapest, Hungary; Department of Neurooncology, National Institute of Clinical Neurosciences, Budapest, Hungary; Department of Radiology, National Institute of Clinical Neurosciences, Budapest, Hungary; Sixth Department of Pulmonology, National Korányi Institute of Pulmonology, Budapest, Hungary; Hungarian Academy of Sciences-Semmelweis University, Molecular Oncology Research Unit, Budapest, Hungary; Department of Tumor Biology, National Korányi Institute of Pulmonology-Semmelweis University, Budapest, Hungary; Department of Thoracic Surgery, National Institute of Oncology-Semmelweis University, Budapest, Hungary; Division of Thoracic Surgery, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria; Children's Hospital Informatics Program at the Harvard-Massachusetts Institute of Technology Division of Health Sciences and Technology, Harvard Medical School, Boston, Massachusetts, USA; Center for Biological Sequence Analysis, Department of Systems Biology, Technical University of Denmark, Lyngby, Denmark; CRUK Lung Cancer Centre of Excellence, UCL Cancer Institute, London, UK; Francis Crick Institute, London, UK
| | - Péter Várallyai
- First Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary; MTA-SE NAP, Brain Metastasis Research Group, Hungarian Academy of Sciences, Second Department of Pathology, Semmelweis University, Budapest, Hungary; Department of Pulmonology, Semmelweis University, Budapest, Hungary; Department of Physics of Complex Systems, Eötvös Loránd University, Budapest, Hungary; Institute of Behavioural Sciences, Semmelweis University, Budapest, Hungary; Department of Neurooncology, National Institute of Clinical Neurosciences, Budapest, Hungary; Department of Radiology, National Institute of Clinical Neurosciences, Budapest, Hungary; Sixth Department of Pulmonology, National Korányi Institute of Pulmonology, Budapest, Hungary; Hungarian Academy of Sciences-Semmelweis University, Molecular Oncology Research Unit, Budapest, Hungary; Department of Tumor Biology, National Korányi Institute of Pulmonology-Semmelweis University, Budapest, Hungary; Department of Thoracic Surgery, National Institute of Oncology-Semmelweis University, Budapest, Hungary; Division of Thoracic Surgery, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria; Children's Hospital Informatics Program at the Harvard-Massachusetts Institute of Technology Division of Health Sciences and Technology, Harvard Medical School, Boston, Massachusetts, USA; Center for Biological Sequence Analysis, Department of Systems Biology, Technical University of Denmark, Lyngby, Denmark; CRUK Lung Cancer Centre of Excellence, UCL Cancer Institute, London, UK; Francis Crick Institute, London, UK
| | - Laura Vízkeleti
- First Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary; MTA-SE NAP, Brain Metastasis Research Group, Hungarian Academy of Sciences, Second Department of Pathology, Semmelweis University, Budapest, Hungary; Department of Pulmonology, Semmelweis University, Budapest, Hungary; Department of Physics of Complex Systems, Eötvös Loránd University, Budapest, Hungary; Institute of Behavioural Sciences, Semmelweis University, Budapest, Hungary; Department of Neurooncology, National Institute of Clinical Neurosciences, Budapest, Hungary; Department of Radiology, National Institute of Clinical Neurosciences, Budapest, Hungary; Sixth Department of Pulmonology, National Korányi Institute of Pulmonology, Budapest, Hungary; Hungarian Academy of Sciences-Semmelweis University, Molecular Oncology Research Unit, Budapest, Hungary; Department of Tumor Biology, National Korányi Institute of Pulmonology-Semmelweis University, Budapest, Hungary; Department of Thoracic Surgery, National Institute of Oncology-Semmelweis University, Budapest, Hungary; Division of Thoracic Surgery, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria; Children's Hospital Informatics Program at the Harvard-Massachusetts Institute of Technology Division of Health Sciences and Technology, Harvard Medical School, Boston, Massachusetts, USA; Center for Biological Sequence Analysis, Department of Systems Biology, Technical University of Denmark, Lyngby, Denmark; CRUK Lung Cancer Centre of Excellence, UCL Cancer Institute, London, UK; Francis Crick Institute, London, UK
| | - Lívia Rojkó
- First Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary; MTA-SE NAP, Brain Metastasis Research Group, Hungarian Academy of Sciences, Second Department of Pathology, Semmelweis University, Budapest, Hungary; Department of Pulmonology, Semmelweis University, Budapest, Hungary; Department of Physics of Complex Systems, Eötvös Loránd University, Budapest, Hungary; Institute of Behavioural Sciences, Semmelweis University, Budapest, Hungary; Department of Neurooncology, National Institute of Clinical Neurosciences, Budapest, Hungary; Department of Radiology, National Institute of Clinical Neurosciences, Budapest, Hungary; Sixth Department of Pulmonology, National Korányi Institute of Pulmonology, Budapest, Hungary; Hungarian Academy of Sciences-Semmelweis University, Molecular Oncology Research Unit, Budapest, Hungary; Department of Tumor Biology, National Korányi Institute of Pulmonology-Semmelweis University, Budapest, Hungary; Department of Thoracic Surgery, National Institute of Oncology-Semmelweis University, Budapest, Hungary; Division of Thoracic Surgery, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria; Children's Hospital Informatics Program at the Harvard-Massachusetts Institute of Technology Division of Health Sciences and Technology, Harvard Medical School, Boston, Massachusetts, USA; Center for Biological Sequence Analysis, Department of Systems Biology, Technical University of Denmark, Lyngby, Denmark; CRUK Lung Cancer Centre of Excellence, UCL Cancer Institute, London, UK; Francis Crick Institute, London, UK
| | - József Tímár
- First Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary; MTA-SE NAP, Brain Metastasis Research Group, Hungarian Academy of Sciences, Second Department of Pathology, Semmelweis University, Budapest, Hungary; Department of Pulmonology, Semmelweis University, Budapest, Hungary; Department of Physics of Complex Systems, Eötvös Loránd University, Budapest, Hungary; Institute of Behavioural Sciences, Semmelweis University, Budapest, Hungary; Department of Neurooncology, National Institute of Clinical Neurosciences, Budapest, Hungary; Department of Radiology, National Institute of Clinical Neurosciences, Budapest, Hungary; Sixth Department of Pulmonology, National Korányi Institute of Pulmonology, Budapest, Hungary; Hungarian Academy of Sciences-Semmelweis University, Molecular Oncology Research Unit, Budapest, Hungary; Department of Tumor Biology, National Korányi Institute of Pulmonology-Semmelweis University, Budapest, Hungary; Department of Thoracic Surgery, National Institute of Oncology-Semmelweis University, Budapest, Hungary; Division of Thoracic Surgery, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria; Children's Hospital Informatics Program at the Harvard-Massachusetts Institute of Technology Division of Health Sciences and Technology, Harvard Medical School, Boston, Massachusetts, USA; Center for Biological Sequence Analysis, Department of Systems Biology, Technical University of Denmark, Lyngby, Denmark; CRUK Lung Cancer Centre of Excellence, UCL Cancer Institute, London, UK; Francis Crick Institute, London, UK
| | - Balázs Döme
- First Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary; MTA-SE NAP, Brain Metastasis Research Group, Hungarian Academy of Sciences, Second Department of Pathology, Semmelweis University, Budapest, Hungary; Department of Pulmonology, Semmelweis University, Budapest, Hungary; Department of Physics of Complex Systems, Eötvös Loránd University, Budapest, Hungary; Institute of Behavioural Sciences, Semmelweis University, Budapest, Hungary; Department of Neurooncology, National Institute of Clinical Neurosciences, Budapest, Hungary; Department of Radiology, National Institute of Clinical Neurosciences, Budapest, Hungary; Sixth Department of Pulmonology, National Korányi Institute of Pulmonology, Budapest, Hungary; Hungarian Academy of Sciences-Semmelweis University, Molecular Oncology Research Unit, Budapest, Hungary; Department of Tumor Biology, National Korányi Institute of Pulmonology-Semmelweis University, Budapest, Hungary; Department of Thoracic Surgery, National Institute of Oncology-Semmelweis University, Budapest, Hungary; Division of Thoracic Surgery, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria; Children's Hospital Informatics Program at the Harvard-Massachusetts Institute of Technology Division of Health Sciences and Technology, Harvard Medical School, Boston, Massachusetts, USA; Center for Biological Sequence Analysis, Department of Systems Biology, Technical University of Denmark, Lyngby, Denmark; CRUK Lung Cancer Centre of Excellence, UCL Cancer Institute, London, UK; Francis Crick Institute, London, UK
| | - Zoltán Szállási
- First Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary; MTA-SE NAP, Brain Metastasis Research Group, Hungarian Academy of Sciences, Second Department of Pathology, Semmelweis University, Budapest, Hungary; Department of Pulmonology, Semmelweis University, Budapest, Hungary; Department of Physics of Complex Systems, Eötvös Loránd University, Budapest, Hungary; Institute of Behavioural Sciences, Semmelweis University, Budapest, Hungary; Department of Neurooncology, National Institute of Clinical Neurosciences, Budapest, Hungary; Department of Radiology, National Institute of Clinical Neurosciences, Budapest, Hungary; Sixth Department of Pulmonology, National Korányi Institute of Pulmonology, Budapest, Hungary; Hungarian Academy of Sciences-Semmelweis University, Molecular Oncology Research Unit, Budapest, Hungary; Department of Tumor Biology, National Korányi Institute of Pulmonology-Semmelweis University, Budapest, Hungary; Department of Thoracic Surgery, National Institute of Oncology-Semmelweis University, Budapest, Hungary; Division of Thoracic Surgery, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria; Children's Hospital Informatics Program at the Harvard-Massachusetts Institute of Technology Division of Health Sciences and Technology, Harvard Medical School, Boston, Massachusetts, USA; Center for Biological Sequence Analysis, Department of Systems Biology, Technical University of Denmark, Lyngby, Denmark; CRUK Lung Cancer Centre of Excellence, UCL Cancer Institute, London, UK; Francis Crick Institute, London, UK
| | - Charles Swanton
- First Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary; MTA-SE NAP, Brain Metastasis Research Group, Hungarian Academy of Sciences, Second Department of Pathology, Semmelweis University, Budapest, Hungary; Department of Pulmonology, Semmelweis University, Budapest, Hungary; Department of Physics of Complex Systems, Eötvös Loránd University, Budapest, Hungary; Institute of Behavioural Sciences, Semmelweis University, Budapest, Hungary; Department of Neurooncology, National Institute of Clinical Neurosciences, Budapest, Hungary; Department of Radiology, National Institute of Clinical Neurosciences, Budapest, Hungary; Sixth Department of Pulmonology, National Korányi Institute of Pulmonology, Budapest, Hungary; Hungarian Academy of Sciences-Semmelweis University, Molecular Oncology Research Unit, Budapest, Hungary; Department of Tumor Biology, National Korányi Institute of Pulmonology-Semmelweis University, Budapest, Hungary; Department of Thoracic Surgery, National Institute of Oncology-Semmelweis University, Budapest, Hungary; Division of Thoracic Surgery, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria; Children's Hospital Informatics Program at the Harvard-Massachusetts Institute of Technology Division of Health Sciences and Technology, Harvard Medical School, Boston, Massachusetts, USA; Center for Biological Sequence Analysis, Department of Systems Biology, Technical University of Denmark, Lyngby, Denmark; CRUK Lung Cancer Centre of Excellence, UCL Cancer Institute, London, UK; Francis Crick Institute, London, UK
| | - Judit Moldvay
- First Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary; MTA-SE NAP, Brain Metastasis Research Group, Hungarian Academy of Sciences, Second Department of Pathology, Semmelweis University, Budapest, Hungary; Department of Pulmonology, Semmelweis University, Budapest, Hungary; Department of Physics of Complex Systems, Eötvös Loránd University, Budapest, Hungary; Institute of Behavioural Sciences, Semmelweis University, Budapest, Hungary; Department of Neurooncology, National Institute of Clinical Neurosciences, Budapest, Hungary; Department of Radiology, National Institute of Clinical Neurosciences, Budapest, Hungary; Sixth Department of Pulmonology, National Korányi Institute of Pulmonology, Budapest, Hungary; Hungarian Academy of Sciences-Semmelweis University, Molecular Oncology Research Unit, Budapest, Hungary; Department of Tumor Biology, National Korányi Institute of Pulmonology-Semmelweis University, Budapest, Hungary; Department of Thoracic Surgery, National Institute of Oncology-Semmelweis University, Budapest, Hungary; Division of Thoracic Surgery, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria; Children's Hospital Informatics Program at the Harvard-Massachusetts Institute of Technology Division of Health Sciences and Technology, Harvard Medical School, Boston, Massachusetts, USA; Center for Biological Sequence Analysis, Department of Systems Biology, Technical University of Denmark, Lyngby, Denmark; CRUK Lung Cancer Centre of Excellence, UCL Cancer Institute, London, UK; Francis Crick Institute, London, UK
| |
Collapse
|
30
|
Aguiar PN, De Mello RA, Barreto CMN, Perry LA, Penny-Dimri J, Tadokoro H, Lopes GDL. Immune checkpoint inhibitors for advanced non-small cell lung cancer: emerging sequencing for new treatment targets. ESMO Open 2017; 2:e000200. [PMID: 29209522 PMCID: PMC5703392 DOI: 10.1136/esmoopen-2017-000200] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2017] [Revised: 06/08/2017] [Accepted: 06/09/2017] [Indexed: 12/20/2022] Open
Abstract
Lung cancer is the leading cause of cancer-related deaths in the world. Immune checkpoint inhibitors (ICI) stimulate cytotoxic lymphocyte activity against tumour cells. These agents are available for the treatment of non-small cell lung cancer (NSCLC) after failure of platinum-based therapy. One recent study has demonstrated that ICI monotherapy was superior to platinum-based chemotherapy for first-line treatment. Nevertheless, this benefit was only for a minority of the population (30%) whose tumour programmed death receptor ligand-1 (PD-L1) expression was above 50%. Therefore, several strategies are under investigation. One option for patients with PD-L1 expression lower than 50% may be the combination of ICI with platinum-based chemotherapy or with ICIs against different targets. However, all of these combinations are at an early stage of investigation and may be very expensive or toxic, producing several harmful adverse events.
Collapse
Affiliation(s)
| | - Ramon Andrade De Mello
- Department of Biomedical Sciences and Medicine, Division of Medical Oncology, University of Algarve, Faro, Portugal
| | | | | | | | - Hakaru Tadokoro
- Division of Medical Oncology, Federal University of São Paulo, São Paulo, Brazil
| | | |
Collapse
|
31
|
Abstract
Malignant mesothelioma is a universally lethal cancer that is increasing in incidence worldwide. There is a dearth of effective therapies, with only one treatment (pemetrexed and cisplatin combination chemotherapy) approved in the past 13 years. However, the past 5 years have witnessed an exponential growth in our understanding of mesothelioma pathobiology, which is set to revolutionize therapeutic strategies. From a genomic standpoint, mesothelioma is characterized by a preponderance of tumour suppressor alterations, for which novel therapies are currently in development. Other promising antitumour agents include inhibitors against angiogenesis, mesothelin and immune checkpoints, which are at various phases of clinical trial testing.
Collapse
Affiliation(s)
- Timothy A Yap
- The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| | - Joachim G Aerts
- Erasmus MC Cancer Institute, 3015 CE Rotterdam, The Netherlands
| | - Sanjay Popat
- Royal Marsden Hospital, London SW3 6JJ, UK
- National Heart and Lung Institute, Imperial College London SW3 6NP, UK
| | | |
Collapse
|
32
|
Miller RA, Cagle PT, Bernicker EH. First-Line Immune Therapy-Implications for Pathologists. Arch Pathol Lab Med 2017; 140:739-40. [PMID: 27472228 DOI: 10.5858/arpa.2016-0904-ed] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|
33
|
Wang S, Li J. [Progress in Immunotherapy for Squamous Non-small Cell Lung Cancer]. ZHONGGUO FEI AI ZA ZHI = CHINESE JOURNAL OF LUNG CANCER 2016; 19:682-686. [PMID: 27760599 PMCID: PMC5973414 DOI: 10.3779/j.issn.1009-3419.2016.10.09] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
近几年来,肺鳞癌在化疗及靶向治疗上的进展不够显著,但免疫治疗却在肺鳞癌的治疗上取得了突破性的进展。免疫治疗通过免疫系统来清除肿瘤细胞,主要分为免疫检查点抑制剂及治疗性疫苗。免疫检查点抑制剂,包括抗细胞毒性T淋巴细胞抗原4(cytotoxic T-lymphocyte associated antigen 4, CTLA-4)抗体与抗程序性死亡受体-1(programmed death receptor 1, PD-1)抗体等多种药物已进行了肺鳞癌的Ⅱ期、Ⅲ期临床试验,并取得了一定成果。免疫治疗将成为肺鳞癌治疗的一种重要手段。
Collapse
Affiliation(s)
- Shouzheng Wang
- National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College,
Beijing 100021, China
| | - Junling Li
- National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College,
Beijing 100021, China
| |
Collapse
|
34
|
Mazza V, Cappuzzo F. Immunotherapy and lung cancer: from therapeutic cancer vaccination to novel approaches. J Thorac Dis 2016; 8:E1348-E1350. [PMID: 27867624 DOI: 10.21037/jtd.2016.10.99] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Valentina Mazza
- Department of Medical Oncology, AUSL Romagna, Ravenna, Italy
| | | |
Collapse
|