1
|
Wen J, Cui W, Yin X, Chen Y, Liu A, Wang Q, Meng X. Application and future prospects of bispecific antibodies in the treatment of non-small cell lung cancer. Cancer Biol Med 2025; 22:j.issn.2095-3941.2024.0470. [PMID: 40192238 PMCID: PMC12032835 DOI: 10.20892/j.issn.2095-3941.2024.0470] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Accepted: 03/05/2025] [Indexed: 04/29/2025] Open
Abstract
As the leading cause of cancer-related deaths, lung cancer remains a noteworthy threat to human health. Although immunotherapies, such as immune checkpoint inhibitors (ICIs), have significantly increased the efficacy of lung cancer treatment, a significant percentage of patients are not sensitive to immunotherapies and patients who initially respond to treatment can quickly develop acquired drug resistance. Bispecific antibodies (bsAbs) bind two different antigens or epitopes simultaneously and have been shown to enhance antitumor efficacy with suitable safety profiles, thus attracting increasing attention as novel antitumor therapies. At present, in addition to the approved bsAb, amivantamab, three novel bsAbs (KN046, AK112, and SHR-1701) are being evaluated in phase 3 clinical trials and many bsAbs are being evaluated in phase 1/2 clinical trials for patients with non-small cell lung cancer (NSCLC). Herein we present the structure, classification, and mechanism of action underlying bsAbs in NSCLC and introduce related clinical trials. Finally, we discuss challenges, potential solutions, and future prospects in the context of cancer treatment with bsAbs.
Collapse
Affiliation(s)
- Junxu Wen
- Department of Radiation Oncology, Shandong Cancer Hospital Affiliated to Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan 250117, China
| | - Wenxing Cui
- Department of Radiation Oncology, Shandong Cancer Hospital Affiliated to Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan 250117, China
| | - Xiaoyan Yin
- Department of Radiation Oncology, Shandong Cancer Hospital Affiliated to Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan 250117, China
| | - Yu Chen
- Department of Radiation Oncology, Shandong Cancer Hospital Affiliated to Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan 250117, China
| | - Ailing Liu
- Department of Radiation Oncology, Shandong Cancer Hospital Affiliated to Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan 250117, China
| | - Qian Wang
- Department of Radiation Oncology, Shandong Cancer Hospital Affiliated to Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan 250117, China
| | - Xiangjiao Meng
- Department of Radiation Oncology, Shandong Cancer Hospital Affiliated to Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan 250117, China
| |
Collapse
|
2
|
Sardarabadi P, Lee KY, Sun WL, Kojabad AA, Liu CH. Investigating T Cell Immune Dynamics and IL-6's Duality in a Microfluidic Lung Tumor Model. ACS APPLIED MATERIALS & INTERFACES 2025; 17:4354-4367. [PMID: 39471283 PMCID: PMC11758792 DOI: 10.1021/acsami.4c09065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/02/2024] [Revised: 10/16/2024] [Accepted: 10/21/2024] [Indexed: 11/01/2024]
Abstract
Interleukin 6 (IL-6), produced by immune cells, is crucial in promoting T cell trafficking to infection and inflammation sites, influencing various physiological and pathological processes. Concentrations of IL-6 and other cytokines and chemokines can influence T cell differentiation and activation. Understanding the dual faces of IL-6 within the tumor microenvironment is crucial to understanding its role. A flow-based microsystem was designed to investigate CD4+ T cell activation in response to different IL-6 gradients in an under-control 3D culture. The study found that cancer cells' response to varying IL-6 concentrations was dynamic and dose-sensitive, with immune cell migration rates showing sensitivity to the IL-6 gradient. A549 cell expansion increases gradually and time-dependently with 50 ng of IL-6, while Jurkat cell migration follows a time-dependent pattern. However, when a total of 100 ng IL-6 concentration is applied, A549 cells expand rapidly, potentially influencing Jurkat cell migration. Jurkat cell mobility is lower, possibly due to increased A549 cell presence and heightened cell-cell interactions. Different IL-6 concentration gradients can modulate the expression of some CD markers like CD69 and programed cell death protein 1 in CD4+ T cells, suggesting that IL-6 concentration gradients affect immune cell phenotypes. This suggests that IL-6 plays a crucial role in activating T helper cells and may be involved in the later phases of inflammation. Also, the increased levels of IFN-γ and TNF-α highlight IL-6's impact on T cell inflammatory response. This study emphasizes the intricate effects of IL-6 on T cell activation, phenotype, cytokine production, and phenotypic heterogeneity, providing valuable insights into immune response modulation in an experimental setting.
Collapse
Affiliation(s)
- Parvaneh Sardarabadi
- Institute
of Nanoengineering and Microsystems, National
Tsing Hua University, Hsinchu 30044, Taiwan,
R.O.C
| | - Kang-Yun Lee
- Division
of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho
Hospital, Taipei Medical University, New Taipei City 235, Taiwan, R.O.C
- Division
of Pulmonary Medicine, Department of Internal Medicine, School of
Medicine, College of Medicine, Taipei Medical
University, Taipei 110, Taiwan, R.O.C
- TMU
Research Center for Thoracic Medicine, Taipei
Medical University, Taipei 110, Taiwan, R.O.C
| | - Wei-Lun Sun
- Pythia
Biotech LTD., New Taipei City 23561, Taiwan,
R.O.C
| | - Amir Asri Kojabad
- Department
of Hematology, School of Allied Medical Sciences, Iran University of Medical Sciences, Tehran 14535, Iran
| | - Cheng-Hsien Liu
- Institute
of Nanoengineering and Microsystems, National
Tsing Hua University, Hsinchu 30044, Taiwan,
R.O.C
- Department
of Power Mechanical Engineering, National
Tsing Hua University, Hsinchu 30044, Taiwan,
R.O.C
- College
of Semiconductor Research, National Tsing
Hua University, Hsinchu 30044, Taiwan, R.O.C
| |
Collapse
|
3
|
Kan SA, Zhang LW, Wang YC, Chiang CY, Chen MH, Huang SH, Chen MH, Liu TY. Bacterial Outer Membrane Vesicle (OMV)-Encapsulated TiO 2 Nanoparticles: A Dual-Action Strategy for Enhanced Radiotherapy and Immunomodulation in Oral Cancer Treatment. NANOMATERIALS (BASEL, SWITZERLAND) 2024; 14:2045. [PMID: 39728581 DOI: 10.3390/nano14242045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/16/2024] [Revised: 12/08/2024] [Accepted: 12/18/2024] [Indexed: 12/28/2024]
Abstract
Oral squamous-cell carcinoma (OSCC) poses significant treatment challenges due to its high recurrence rates and the limitations of current therapies. Titanium dioxide (TiO2) nanoparticles are promising radiosensitizers, while bacterial outer membrane vesicles (OMVs) are known for their immunomodulatory properties. This study investigates the potential of OMV-encapsulated TiO2 nanoparticles (TiO2@OMV) to combine these effects for improved OSCC treatment. TiO2 nanoparticles were synthesized using a hydrothermal method and encapsulated within OMVs derived from Escherichia coli. The TiO2@OMV carriers were evaluated for their ability to enhance radiosensitivity and stimulate immune responses in OSCC cell lines. Reactive oxygen species (ROS) production, macrophage recruitment, and selective cytotoxicity toward cancer cells were assessed. TiO2@OMV demonstrated significant radiosensitization and immune activation compared to unencapsulated TiO2 nanoparticles. The system selectively induced cytotoxicity in OSCC cells, sparing normal cells, and enhanced ROS generation and macrophage-mediated antitumor responses. This study highlights TiO2@OMV as a dual-action therapeutic platform that synergizes radiotherapy and immunomodulation, offering a targeted and effective strategy for OSCC treatment. The approach could improve therapeutic outcomes and reduce the adverse effects associated with conventional therapies.
Collapse
Affiliation(s)
- Shun-An Kan
- Department of Medical Education, Taipei Veterans General Hospital, Taipei 112, Taiwan
| | - Li-Wen Zhang
- Department of Biomedical Engineering, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
| | - Yu-Chi Wang
- Department of Biomedical Engineering, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
| | - Cheng-Yu Chiang
- Department of Biomedical Engineering, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
| | - Mei-Hsiu Chen
- Department of Internal Medicine, Far Eastern Memorial Hospital, New Taipei 220, Taiwan
| | - Shih-Hao Huang
- Division of Neurosurgery, Department of Surgery, Far Eastern Memorial Hospital, New Taipei 220, Taiwan
- Department of Healthcare Administration, Asia Eastern University of Science and Technology, New Taipei 220, Taiwan
| | - Ming-Hong Chen
- Department of Biomedical Engineering, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
- Division of Neurosurgery, Department of Surgery, Far Eastern Memorial Hospital, New Taipei 220, Taiwan
- Department of Electrical Engineering, Yuan Ze University, Taoyuan 320, Taiwan
| | - Tse-Ying Liu
- Department of Biomedical Engineering, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
| |
Collapse
|
4
|
Hasan S, Awasthi P, Malik S, Dwivedi M. Immunotherapeutic strategies to induce inflection in the immune response: therapy for cancer and COVID-19. Biotechnol Genet Eng Rev 2024; 40:3571-3610. [PMID: 36411974 DOI: 10.1080/02648725.2022.2147661] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Accepted: 10/11/2022] [Indexed: 11/23/2022]
Abstract
Cancer has agonized the human race for millions of years. The present decade witnesses biological therapeutics to combat cancer effectively. Cancer Immunotherapy involves the use of therapeutics for manipulation of the immune system by immune agents like cytokines, vaccines, and transfection agents. Recently, this therapeutic approach has got vast attention due to the current pandemic COVID-19 and has been very effective. Concerning cancer, immunotherapy is based on the activation of the host's antitumor response by enhancing effector cell number and the production of soluble mediators, thereby reducing the host's suppressor mechanisms by induction of a tumour killing environment and by modulating immune checkpoints. In the present era, immunotherapies have gained traction and momentum as a pedestal of cancer treatment, improving the prognosis of many patients with a wide variety of haematological and solid malignancies. Food supplements, natural immunomodulatory drugs, and phytochemicals, with recent developments, have shown positive trends in cancer treatment by improving the immune system. The current review presents the systematic studies on major immunotherapeutics and their development for the effective treatment of cancers as well as in COVID-19. The focus of the review is to highlight comparative analytics of existing and novel immunotherapies in cancers, concerning immunomodulatory drugs and natural immunosuppressants, including immunotherapy in COVID-19 patients.
Collapse
Affiliation(s)
- Saba Hasan
- Amity Institute of Biotechnology, Amity University Uttar Pradesh, Lucknow, India
| | - Prankur Awasthi
- Amity Institute of Biotechnology, Amity University Uttar Pradesh, Lucknow, India
| | - Sumira Malik
- Amity Institute of Biotechnology, Amity University, Ranchi, Jharkhand, India
| | - Manish Dwivedi
- Amity Institute of Biotechnology, Amity University Uttar Pradesh, Lucknow, India
| |
Collapse
|
5
|
Lu Y, Zheng J, Lin P, Lin Y, Zheng Y, Mai Z, Chen X, Xia T, Zhao X, Cui L. Tumor Microenvironment-Derived Exosomes: A Double-Edged Sword for Advanced T Cell-Based Immunotherapy. ACS NANO 2024; 18:27230-27260. [PMID: 39319751 DOI: 10.1021/acsnano.4c09190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/26/2024]
Abstract
The tumor microenvironment (TME) plays a crucial role in cancer progression and immune evasion, partially mediated by the activity of the TME-derived exosomes. These extracellular vesicles are pivotal in shaping immune responses through the transfer of proteins, lipids, and nucleic acids between cells, facilitating a complex interplay that promotes tumor growth and metastasis. This review delves into the dual roles of exosomes in the TME, highlighting both their immunosuppressive functions and their emerging therapeutic potential. Exosomes can inhibit T cell function and promote tumor immune escape by carrying immune-modulatory molecules, such as PD-L1, yet they also hold promise for cancer therapy as vehicles for delivering tumor antigens and costimulatory signals. Additionally, the review discusses the intricate crosstalk mediated by exosomes among various cell types within the TME, influencing both cancer progression and responses to immunotherapies. Moreover, this highlights current challenges and future directions. Collectively, elucidating the detailed mechanisms by which TME-derived exosomes mediate T cell function offers a promising avenue for revolutionizing cancer treatment. Understanding these interactions allows for the development of targeted therapies that manipulate exosomal pathways to enhance the immune system's response to tumors.
Collapse
Affiliation(s)
- Ye Lu
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong 510280, China
| | - Jiarong Zheng
- Department of Dentistry, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, China
| | - Pei Lin
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong 510280, China
| | - Yunfan Lin
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong 510280, China
| | - Yucheng Zheng
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong 510280, China
| | - Zizhao Mai
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong 510280, China
| | - Xu Chen
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong 510280, China
| | - Tian Xia
- Division of NanoMedicine, Department of Medicine, University of California Los Angeles, Los Angeles, California 90095, United States
| | - Xinyuan Zhao
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong 510280, China
| | - Li Cui
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong 510280, China
- School of Dentistry, University of California Los Angeles, Los Angeles, California 90095, United States
| |
Collapse
|
6
|
Serrano S, Barrio R, Martínez-Rubio Á, Belmonte-Beitia J, Pérez-García VM. Understanding the role of B cells in CAR T-cell therapy in leukemia through a mathematical model. CHAOS (WOODBURY, N.Y.) 2024; 34:083142. [PMID: 39191245 DOI: 10.1063/5.0206341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Accepted: 08/09/2024] [Indexed: 08/29/2024]
Abstract
Chimeric antigen receptor T (CAR T) cell therapy has been proven to be successful against a variety of leukemias and lymphomas. This paper undertakes an analytical and numerical study of a mathematical model describing the competition of CAR T, leukemia, tumor, and B cells. Considering its significance in sustaining anti-CD19 CAR T-cell stimulation, a B-cell source term is integrated into the model. Through stability and bifurcation analyses, the potential for tumor eradication, contingent on the continuous influx of B cells, has been revealed, showing a transcritical bifurcation at a critical B-cell input. Additionally, an almost heteroclinic cycle between equilibrium points is identified, providing a theoretical basis for understanding disease relapse. Analyzing the oscillatory behavior of the system, the time-dependent dynamics of CAR T cells and leukemic cells can be approximated, shedding light on the impact of initial tumor burden on therapeutic outcomes. In conclusion, the study provides insights into CAR T-cell therapy dynamics for acute lymphoblastic leukemias, offering a theoretical foundation for clinical observations and suggesting avenues for future immunotherapy modeling research.
Collapse
Affiliation(s)
- Sergio Serrano
- IUMA, CoDy and Department of Applied Mathematics, Universidad de Zaragoza, Zaragoza 50009, Spain
| | - Roberto Barrio
- IUMA, CoDy and Department of Applied Mathematics, Universidad de Zaragoza, Zaragoza 50009, Spain
| | - Álvaro Martínez-Rubio
- Department of Mathematics, Universidad de Cádiz, Puerto Real, Cádiz 11510, Spain
- Biomedical Research and Innovation Institute of Cádiz (INiBICA), Hospital Universitario Puerta del Mar, Cádiz 11002, Spain
| | - Juan Belmonte-Beitia
- Mathematical Oncology Laboratory (MOLAB), Departament of Mathematics, Instituto de Matemática Aplicada a la Ciencia y la Ingeniería, Universidad de Castilla-La Mancha, Ciudad Real 13071, Spain
| | - Víctor M Pérez-García
- Mathematical Oncology Laboratory (MOLAB), Departament of Mathematics, Instituto de Matemática Aplicada a la Ciencia y la Ingeniería, Universidad de Castilla-La Mancha, Ciudad Real 13071, Spain
| |
Collapse
|
7
|
Wang Z, Sun W, Hua R, Wang Y, Li Y, Zhang H. Promising dawn in tumor microenvironment therapy: engineering oral bacteria. Int J Oral Sci 2024; 16:24. [PMID: 38472176 PMCID: PMC10933493 DOI: 10.1038/s41368-024-00282-3] [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: 10/27/2023] [Revised: 01/06/2024] [Accepted: 01/07/2024] [Indexed: 03/14/2024] Open
Abstract
Despite decades of research, cancer continues to be a major global health concern. The human mouth appears to be a multiplicity of local environments communicating with other organs and causing diseases via microbes. Nowadays, the role of oral microbes in the development and progression of cancer has received increasing scrutiny. At the same time, bioengineering technology and nanotechnology is growing rapidly, in which the physiological activities of natural bacteria are modified to improve the therapeutic efficiency of cancers. These engineered bacteria were transformed to achieve directed genetic reprogramming, selective functional reorganization and precise control. In contrast to endotoxins produced by typical genetically modified bacteria, oral flora exhibits favorable biosafety characteristics. To outline the current cognitions upon oral microbes, engineered microbes and human cancers, related literatures were searched and reviewed based on the PubMed database. We focused on a number of oral microbes and related mechanisms associated with the tumor microenvironment, which involve in cancer occurrence and development. Whether engineering oral bacteria can be a possible application of cancer therapy is worth consideration. A deeper understanding of the relationship between engineered oral bacteria and cancer therapy may enhance our knowledge of tumor pathogenesis thus providing new insights and strategies for cancer prevention and treatment.
Collapse
Affiliation(s)
- Zifei Wang
- Key Laboratory of Oral Diseases Research of Anhui Province, College & Hospital of Stomatology, Anhui Medical University, Hefei, China
| | - Wansu Sun
- Department of Stomatology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Ruixue Hua
- Key Laboratory of Oral Diseases Research of Anhui Province, College & Hospital of Stomatology, Anhui Medical University, Hefei, China
| | - Yuanyin Wang
- Key Laboratory of Oral Diseases Research of Anhui Province, College & Hospital of Stomatology, Anhui Medical University, Hefei, China
| | - Yang Li
- Department of Genetics, School of Life Science, Anhui Medical University, Hefei, China.
| | - Hengguo Zhang
- Key Laboratory of Oral Diseases Research of Anhui Province, College & Hospital of Stomatology, Anhui Medical University, Hefei, China.
| |
Collapse
|
8
|
Gao Y, Feng C, Ma J, Yan Q. Protein arginine methyltransferases (PRMTs): Orchestrators of cancer pathogenesis, immunotherapy dynamics, and drug resistance. Biochem Pharmacol 2024; 221:116048. [PMID: 38346542 DOI: 10.1016/j.bcp.2024.116048] [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: 11/27/2023] [Revised: 01/15/2024] [Accepted: 02/06/2024] [Indexed: 02/16/2024]
Abstract
Protein Arginine Methyltransferases (PRMTs) are a family of enzymes regulating protein arginine methylation, which is a post-translational modification crucial for various cellular processes. Recent studies have highlighted the mechanistic role of PRMTs in cancer pathogenesis, immunotherapy, and drug resistance. PRMTs are involved in diverse oncogenic processes, including cell proliferation, apoptosis, and metastasis. They exert their effects by methylation of histones, transcription factors, and other regulatory proteins, resulting in altered gene expression patterns. PRMT-mediated histone methylation can lead to aberrant chromatin remodeling and epigenetic changes that drive oncogenesis. Additionally, PRMTs can directly interact with key signaling pathways involved in cancer progression, such as the PI3K/Akt and MAPK pathways, thereby modulating cell survival and proliferation. In the context of cancer immunotherapy, PRMTs have emerged as critical regulators of immune responses. They modulate immune checkpoint molecules, including programmed cell death protein 1 (PD-1), through arginine methylation. Drug resistance is a significant challenge in cancer treatment, and PRMTs have been implicated in this phenomenon. PRMTs can contribute to drug resistance through multiple mechanisms, including the epigenetic regulation of drug efflux pumps, altered DNA damage repair, and modulation of cell survival pathways. In conclusion, PRMTs play critical roles in cancer pathogenesis, immunotherapy, and drug resistance. In this overview, we have endeavored to illuminate the mechanistic intricacies of PRMT-mediated processes. Shedding light on these aspects will offer valuable insights into the fundamental biology of cancer and establish PRMTs as promising therapeutic targets.
Collapse
Affiliation(s)
- Yihang Gao
- Department of Laboratory Medicine, the Second Hospital of Jilin University, Changchun 130000, China
| | - Chongchong Feng
- Department of Laboratory Medicine, the Second Hospital of Jilin University, Changchun 130000, China.
| | - Jingru Ma
- Department of Laboratory Medicine, the Second Hospital of Jilin University, Changchun 130000, China
| | - Qingzhu Yan
- Department of Ultrasound Medicine, the Second Hospital of Jilin University, Changchun 130000, China
| |
Collapse
|
9
|
Smith A, Boby JM, Benny SJ, Ghazali N, Vermeulen E, George M. Immunotherapy in Older Patients with Cancer: A Narrative Review. Int J Gen Med 2024; 17:305-313. [PMID: 38298248 PMCID: PMC10830099 DOI: 10.2147/ijgm.s435001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Accepted: 01/09/2024] [Indexed: 02/02/2024] Open
Abstract
Purpose Immunotherapies have revolutionized cancer treatment; however, relatively little is known about their efficacy and toxicity in the elderly, a cohort accounting for more than half of total cancer cases. In this review, we aim to provide insight into the current knowledge base regarding the clinical utility and side effects of immunotherapies in the geriatric population as well as identify key gaps in the literature where further research is essential. Methods We conducted a rapid critical review of available literature, focusing on studies reporting on use of immunotherapy in cancer patients aged ≥65 years. The review assessed studies that included different types of cancer, were of multiple study types (although predominantly retrospective), had different study duration, and reported different outcomes of interest. Owing to this heterogeneity, meta-analysis and a direct comparison between studies were not feasible. Results Overall, the review findings indicate that certain malignancies have shown comparable survival rates in younger and older age groups when managed with immunotherapeutic drugs, the incidence of immunotherapy-related side effects varies only slightly by age groups, and in general there is a lack of studies on the determinants of the clinical outcomes of immunotherapy in or including geriatric patients. Conclusion Enhanced clinical benefits along with better tolerability associated with immunotherapies make it an attractive alternative to conventional chemotherapeutic drugs, especially in elderly patients. There is currently a limited number of studies assessing the clinical outcomes of immunotherapies, particularly in the elderly. Overall, our findings reflect a need for further prospective studies focussing on geriatric patients representative of the real-life population, in order to derive a more precise understanding of the clinical utility, toxicity profile, and cost-effectiveness of immune checkpoint inhibitors in older patients with cancer.
Collapse
Affiliation(s)
- Alexandra Smith
- Tamworth Hospital, Hunter New England Local Health District (NSW Health), Tamworth, NSW, Australia
| | | | | | | | - Elke Vermeulen
- Tamworth Hospital, Hunter New England Local Health District (NSW Health), Tamworth, NSW, Australia
| | - Mathew George
- Tamworth Hospital, Hunter New England Local Health District (NSW Health), Tamworth, NSW, Australia
| |
Collapse
|
10
|
Zhang S, Guo Y, Hu Y, Gao X, Bai F, Ding Q, Hou K, Wang Z, Sun X, Zhao H, Qu Z, Xu Q. The role of APOBEC3C in modulating the tumor microenvironment and stemness properties of glioma: evidence from pancancer analysis. Front Immunol 2023; 14:1242972. [PMID: 37809064 PMCID: PMC10551170 DOI: 10.3389/fimmu.2023.1242972] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Accepted: 09/04/2023] [Indexed: 10/10/2023] Open
Abstract
Background It is now understood that APOBEC3 family proteins (A3s) are essential in tumor progression, yet their involvement in tumor immunity and stemness across diverse cancer types remains poorly understood. Methods In the present study, comprehensive genome-wide statistical and bioinformatic analyses were conducted to elucidate A3 family expression patterns, establishing clinically relevant correlations with prognosis, the tumor microenvironment(TME), immune infiltration, checkpoint blockade, and stemness across cancers. Different experimental techniques were applied, including RT-qPCR, immunohistochemistry, sphere formation assays, Transwell migration assays, and wound-healing assays, to investigate the impact of A3C on low-grade glioma (LGG) and glioblastoma multiforme (GBM), as well as its function in glioma stem cells(GSCs). Results Dysregulated expression of A3s was observed in various human cancer tissues. The prognostic value of A3 expression differed across cancer types, with a link to particularly unfavorable outcomes in gliomas. A3s are associated with the the TME and stemness in multiple cancers. Additionally, we developed an independent prognostic model based on A3s expression, which may be an independent prognostic factor for OS in patients with glioma. Subsequent validation underscored a strong association between elevated A3C expression and adverse prognostic outcomes, higher tumor grades, and unfavorable histology in glioma. A potential connection between A3C and glioma progression was established. Notably, gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses implicated A3C in immune system-related diseases, with heightened A3C levels contributing to an immunosuppressive tumor microenvironment (TME) in glioma. Furthermore, in vitro experiments substantiated the role of A3C in sustaining and renewing glioma stem cells, as A3C deletion led to diminished proliferation, invasion, and migration of glioma cells. Conclusion The A3 family exhibits heterogeneous expression across various cancer types, with its expression profile serving as a predictive marker for overall survival in glioma patients. A3C emerges as a regulator of glioma progression, exerting its influence through modulation of the tumor microenvironment and regulation of stemness.
Collapse
Affiliation(s)
- Shoudu Zhang
- Henan Provincial Engineering Laboratory of Insects Bio-reactor, Nanyang Normal University, Nanyang, Henan, China
| | - Yugang Guo
- Henan Provincial Engineering Laboratory of Insects Bio-reactor, Nanyang Normal University, Nanyang, Henan, China
| | - Yuanzheng Hu
- Henan Provincial Engineering Laboratory of Insects Bio-reactor, Nanyang Normal University, Nanyang, Henan, China
| | - Xiaofang Gao
- The Department of Science and Technology, Zhengzhou Revogene Ltd, Zhengzhou, Henan, China
| | - Fanghui Bai
- Department of Oncology, Nanyang central Hospital, Nanyang, Henan, China
| | - Qian Ding
- Henan Provincial Engineering Laboratory of Insects Bio-reactor, Nanyang Normal University, Nanyang, Henan, China
| | - Kaiqi Hou
- Henan Provincial Engineering Laboratory of Insects Bio-reactor, Nanyang Normal University, Nanyang, Henan, China
| | - Zongqing Wang
- Henan Provincial Engineering Laboratory of Insects Bio-reactor, Nanyang Normal University, Nanyang, Henan, China
| | - Xing Sun
- Department of Oncology, Nanyang central Hospital, Nanyang, Henan, China
| | - Hui Zhao
- The Department of Science and Technology, Zhengzhou Revogene Ltd, Zhengzhou, Henan, China
| | - Zhongyu Qu
- Department of Oncology, Nanyang central Hospital, Nanyang, Henan, China
| | - Qian Xu
- Henan Provincial Engineering Laboratory of Insects Bio-reactor, Nanyang Normal University, Nanyang, Henan, China
| |
Collapse
|
11
|
Sbrana A, Mazzini G, Comolli G, Antonuzzo A, Danova M. The contribution of automated cytometry in immuno-oncology. Methods Cell Biol 2023; 195:23-37. [PMID: 40180453 DOI: 10.1016/bs.mcb.2023.03.005] [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] [Indexed: 04/05/2025]
Abstract
Cancer immunotherapy has been a real revolution and has given many survival chances to several patients. However, the understanding of resistance to immunotherapy is still an unmet need in clinical practice. Monitoring of immune mechanisms could be a tool to better understand this phenomenon. FCM and CyTOF could be used in this field, since they allow the simultaneous analysis of several protein expressions pattern, thus possibly understanding the functions of several immune cell populations, such as T cells, and their interactions with tumor cells and tumor microenvironment. Furthermore, automated cytometry could be used to understand the interaction of drugs with their target through the analysis of receptor occupancy. Spectral overlap, however, could be a limit for multiple simultaneous analyses. Other possible limitations of these techniques are a low number of cells in samples and the need for viable cells (with the possible interference of cell debris). The lack of standardized protocols, and thus the difficult reproducibility, have been the major limit to their application in clinical practice, so international efforts have been made to get to shared guidelines. Ongoing trials are to answer to the possibility of clinical application of these techniques.
Collapse
Affiliation(s)
- Andrea Sbrana
- Department of Surgical, Medical and Molecular Pathology and Critical Care Area, University of Pisa, Pisa, Italy; Service of Pneumo-Oncology, Unit of Pneumology, Pisa, Italy
| | | | - Giuditta Comolli
- Department of Microbiology and Virology and Laboratory of Biochemistry-Biotechnology and Advanced Diagnostics, IRCCS San Matteo Foundation, Pavia, Italy
| | | | - Marco Danova
- Unit of Internal Medicine and Medical Oncology, Vigevano Civic Hospital, Pavia, Italy; LIUC University, Castellanza, Varese, Italy.
| |
Collapse
|
12
|
Zeng L, Xu H, Li SH, Xu SY, Chen K, Qin LJ, Miao L, Wang F, Deng L, Wang FH, Li L, Fu S, Liu N, Wang R, Li YQ, Wang HY. Cross-cohort analysis identified an immune checkpoint-based signature to predict the clinical outcomes of neuroblastoma. J Immunother Cancer 2023; 11:jitc-2022-005980. [PMID: 37130627 PMCID: PMC10163522 DOI: 10.1136/jitc-2022-005980] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/16/2023] [Indexed: 05/04/2023] Open
Abstract
BACKGROUND Neuroblastoma (NB) places a substantial health burden on families worldwide. This study aimed to develop an immune checkpoint-based signature (ICS) based on the expression of immune checkpoints to better assess patient survival risk and potentially guide patient selection for immunotherapy of NB. METHODS Immunohistochemistry integrated with digital pathology was used to determine the expression levels of 9 immune checkpoints in 212 tumor tissues used as the discovery set. The GSE85047 dataset (n=272) was used as a validation set in this study. In the discovery set, the ICS was constructed using a random forest algorithm and confirmed in the validation set to predict overall survival (OS) and event-free survival (EFS). Kaplan-Meier curves with a log-rank test were drawn to compare the survival differences. A receiver operating characteristic (ROC) curve was applied to calculate the area under the curve (AUC). RESULTS Seven immune checkpoints, including PD-L1, B7-H3, IDO1, VISTA, T-cell immunoglobulin and mucin domain containing-3 (TIM-3), inducible costimulatory molecule (ICOS) and costimulatory molecule 40 (OX40), were identified as abnormally expressed in NB in the discovery set. OX40, B7-H3, ICOS and TIM-3 were eventually selected for the ICS model in the discovery set, and 89 patients with high risk had an inferior OS (HR 15.91, 95% CI 8.87 to 28.55, p<0.001) and EFS (HR 4.30, 95% CI 2.80 to 6.62, p<0.001). Furthermore, the prognostic value of the ICS was confirmed in the validation set (p<0.001). Multivariate Cox regression analysis demonstrated that age and the ICS were independent risk factors for OS in the discovery set (HR 6.17, 95% CI 1.78 to 21.29 and HR 1.18, 95% CI 1.12 to 1.25, respectively). Furthermore, nomogram A combining the ICS and age demonstrated significantly better prognostic value than age alone in predicting the patients' 1-year, 3-year and 5-year OS in the discovery set (1 year: AUC, 0.891 (95% CI 0.797 to 0.985) vs 0.675 (95% CI 0.592 to 0.758); 3 years: 0.875 (95% CI 0.817 to 0.933) vs 0.701 (95% CI 0.645 to 0.758); 5 years: 0.898 (95% CI 0.851 to 0.940) vs 0.724 (95% CI 0.673 to 0.775), respectively), which was confirmed in the validation set. CONCLUSIONS We propose an ICS that significantly differentiates between low-risk and high-risk patients, which might add prognostic value to age and provide clues for immunotherapy in NB.
Collapse
Affiliation(s)
- Liang Zeng
- Department of Pathology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangdong Provincial Clinical Research Center for Child Health, National Children's Medical Center for South Central Region, Guangzhou, China
| | - Hui Xu
- Department of Pathology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangdong Provincial Clinical Research Center for Child Health, National Children's Medical Center for South Central Region, Guangzhou, China
| | - Shu-Hua Li
- Molecular Diagnosis and Gene Testing Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Shuo-Yu Xu
- Department of General Surgery, Southern Medical University Nanfang Hospital, Guangzhou, China
- Bio-totem Pte. Ltd, Foshan, China
| | - Kai Chen
- Department of Pathology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangdong Provincial Clinical Research Center for Child Health, National Children's Medical Center for South Central Region, Guangzhou, China
| | - Liang-Jun Qin
- Department of Pathology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangdong Provincial Clinical Research Center for Child Health, National Children's Medical Center for South Central Region, Guangzhou, China
| | - Lei Miao
- Guangzhou Institute of Paediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangdong Provincial Clinical Research Center for Child Health,Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, National Children's Medical Center for South Central Region, Guangzhou, China
| | - Fang Wang
- Department of Molecular Diagnostics, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Ling Deng
- Department of Molecular Diagnostics, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Feng-Hua Wang
- Department of Thoracic Surgery, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Le Li
- Department of Thoracic Surgery, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Sha Fu
- Guangdong Provincial Key Laboratory of Malignant Tumour Epigenetics and Gene Regulation, Cellular & Molecular Diagnostics Center, Sun Yat-Sen Memorial Hospital, Guangzhou, China
| | - Na Liu
- Department of Experimental Research, State Key Laboratory of Oncology in Southern China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Ran Wang
- Department of Pathology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Ying-Qing Li
- State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center; Collaborative Innovation Center of Cancer Medicine, Guangzhou, China
| | - Hai-Yun Wang
- Department of Pathology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangdong Provincial Clinical Research Center for Child Health, National Children's Medical Center for South Central Region, Guangzhou, China
- Guangzhou Institute of Paediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangdong Provincial Clinical Research Center for Child Health,Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, National Children's Medical Center for South Central Region, Guangzhou, China
| |
Collapse
|
13
|
A Review of RRx-001: A Late-Stage Multi-Indication Inhibitor of NLRP3 Activation and Chronic Inflammation. Drugs 2023; 83:389-402. [PMID: 36920652 PMCID: PMC10015535 DOI: 10.1007/s40265-023-01838-z] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/22/2023] [Indexed: 03/16/2023]
Abstract
Chronic unresolving inflammation is emerging as a key underlying pathological feature of many if not most diseases ranging from autoimmune conditions to cardiometabolic and neurological disorders. Dysregulated immune and inflammasome activation is thought to be the central driver of unresolving inflammation, which in some ways provides a unified theory of disease pathology and progression. Inflammasomes are a group of large cytosolic protein complexes that, in response to infection- or stress-associated stimuli, oligomerize and assemble to generate a platform for driving inflammation. This occurs through proteolytic activation of caspase-1-mediated inflammatory responses, including cleavage and secretion of the proinflammatory cytokines interleukin (IL)-1β and IL-18, and initiation of pyroptosis, an inflammatory form of cell death. Several inflammasomes have been characterized. The most well-studied is the nucleotide-binding domain (NOD)-like receptor protein 3 (NLRP3) inflammasome, so named because the NLRP3 protein in the complex, which is primarily present in immune and inflammatory cells following activation by inflammatory stimuli, belongs to the family of nucleotide-binding and oligomerization domain (Nod) receptor proteins. Several NLRP3 inflammasome inhibitors are in development, all with multi-indication activity. This review discusses the current status, known mechanisms of action, and disease-modifying therapeutic potential of RRx-001, a direct NLRP3 inflammasome inhibitor under investigation in several late-stage anticancer clinical trials, including a phase 3 trial for the treatment of third-line and beyond small cell lung cancer (SCLC), an indication with no treatment, in which RRx-001 is combined with reintroduced chemotherapy from the first line, carboplatin/cisplatin and etoposide (ClinicalTrials.gov Identifier: NCT03699956). Studies from multiple independent groups have now confirmed that RRx-001 is safe and well tolerated in humans. Additionally, emerging evidence in preclinical animal models suggests that RRx-001 could be effective in a wide range of diseases where immune and inflammasome activation drives disease pathology.
Collapse
|
14
|
Zaman A, Bivona TG. Quantitative Framework for Bench-to-Bedside Cancer Research. Cancers (Basel) 2022; 14:5254. [PMID: 36358671 PMCID: PMC9658824 DOI: 10.3390/cancers14215254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 10/20/2022] [Accepted: 10/24/2022] [Indexed: 11/29/2022] Open
Abstract
Bioscience is an interdisciplinary venture. Driven by a quantum shift in the volume of high throughput data and in ready availability of data-intensive technologies, mathematical and quantitative approaches have become increasingly common in bioscience. For instance, a recent shift towards a quantitative description of cells and phenotypes, which is supplanting conventional qualitative descriptions, has generated immense promise and opportunities in the field of bench-to-bedside cancer OMICS, chemical biology and pharmacology. Nevertheless, like any burgeoning field, there remains a lack of shared and standardized framework for quantitative cancer research. Here, in the context of cancer, we present a basic framework and guidelines for bench-to-bedside quantitative research and therapy. We outline some of the basic concepts and their parallel use cases for chemical-protein interactions. Along with several recommendations for assay setup and conditions, we also catalog applications of these quantitative techniques in some of the most widespread discovery pipeline and analytical methods in the field. We believe adherence to these guidelines will improve experimental design, reduce variabilities and standardize quantitative datasets.
Collapse
Affiliation(s)
- Aubhishek Zaman
- Department of Medicine, University of California, San Francisco, CA 94158, USA
- UCSF Helen Diller Comprehensive Cancer Center, University of California, San Francisco, CA 94158, USA
| | - Trever G. Bivona
- Department of Medicine, University of California, San Francisco, CA 94158, USA
- UCSF Helen Diller Comprehensive Cancer Center, University of California, San Francisco, CA 94158, USA
- Chan-Zuckerberg Biohub, San Francisco, CA 94158, USA
| |
Collapse
|
15
|
Bausart M, Préat V, Malfanti A. Immunotherapy for glioblastoma: the promise of combination strategies. J Exp Clin Cancer Res 2022; 41:35. [PMID: 35078492 PMCID: PMC8787896 DOI: 10.1186/s13046-022-02251-2] [Citation(s) in RCA: 119] [Impact Index Per Article: 39.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Accepted: 01/11/2022] [Indexed: 12/11/2022] Open
Abstract
Glioblastoma (GBM) treatment has remained almost unchanged for more than 20 years. The current standard of care involves surgical resection (if possible) followed by concomitant radiotherapy and chemotherapy. In recent years, immunotherapy strategies have revolutionized the treatment of many cancers, increasing the hope for GBM therapy. However, mostly due to the high, multifactorial immunosuppression occurring in the microenvironment, the poor knowledge of the neuroimmune system and the presence of the blood-brain barrier, the efficacy of immunotherapy in GBM is still low. Recently, new strategies for GBM treatments have employed immunotherapy combinations and have provided encouraging results in both preclinical and clinical studies. The lessons learned from clinical trials highlight the importance of tackling different arms of immunity. In this review, we aim to summarize the preclinical evidence regarding combination immunotherapy in terms of immune and survival benefits for GBM management. The outcomes of recent studies assessing the combination of different classes of immunotherapeutic agents (e.g., immune checkpoint blockade and vaccines) will be discussed. Finally, future strategies to ameliorate the efficacy of immunotherapy and facilitate clinical translation will be provided to address the unmet medical needs of GBM.
Collapse
Affiliation(s)
- Mathilde Bausart
- UCLouvain, Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, Avenue Mounier 73 B1.73.12, 1200, Brussels, Belgium
| | - Véronique Préat
- UCLouvain, Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, Avenue Mounier 73 B1.73.12, 1200, Brussels, Belgium.
| | - Alessio Malfanti
- UCLouvain, Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, Avenue Mounier 73 B1.73.12, 1200, Brussels, Belgium
| |
Collapse
|
16
|
Hußtegge M, Hoang NA, Rebstock J, Monecke A, Gockel I, Weimann A, Schumacher G, Bechmann I, Lordick F, Kallendrusch S, Körfer J. PD-1 inhibition in patient derived tissue cultures of human gastric and gastroesophageal adenocarcinoma. Oncoimmunology 2021; 10:1960729. [PMID: 34434611 PMCID: PMC8381835 DOI: 10.1080/2162402x.2021.1960729] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Emerging immunotherapies quest for better patient stratification in cancer treatment decisions. Moderate response rates of PD-1 inhibition in gastric and esophagogastric junction cancers urge for meaningful human model systems that allow for investigating immune responses ex vivo. Here, the standardized patient-derived tissue culture (PDTC) model was applied to investigate tumor response to the PD-1 inhibitor Nivolumab and the CD3/CD28 t-lymphocyte activator ImmunoCultTM. Resident t-lymphocytes, tumor proliferation and apoptosis, as well as bulk gene expression data were analyzed after 72 h of PD-1 inhibition either as monotherapy or combined with Oxaliplatin or ImmunoCultTM. Individual responses to PD-1 inhibition were found ex vivo and combination with chemotherapy or t-lymphocyte activation led to enhanced antitumoral effects in PDTCs. T-lymphocyte activation as well as the addition of pre-cultured peripheral blood mononuclear cells improved PDTC for studying t-lymphocyte and tumor cell communication. These data support the potential of PDTC to investigate immunotherapy ex vivo in gastric and esophagogastric junction cancer.
Collapse
Affiliation(s)
- Marlon Hußtegge
- Institute of Anatomy, University of Leipzig, Leipzig.,Department of Oncology, Gastroenterology, Hepatology, Pulmonology, and Infectious Diseases, University Cancer Center Leipzig, University Hospital Leipzig, Leipzig, Germany
| | - Ngoc Anh Hoang
- Department of Oncology, Gastroenterology, Hepatology, Pulmonology, and Infectious Diseases, University Cancer Center Leipzig, University Hospital Leipzig, Leipzig, Germany
| | - Jakob Rebstock
- Institute of Anatomy, University of Leipzig, Leipzig.,Department of Oncology, Gastroenterology, Hepatology, Pulmonology, and Infectious Diseases, University Cancer Center Leipzig, University Hospital Leipzig, Leipzig, Germany
| | - Astrid Monecke
- Institute of Pathology, University Hospital Leipzig, Leipzig, Germany
| | - Ines Gockel
- Department of Visceral, Transplantation, Thoracic and Vascular Surgery, University Hospital Leipzig, Leipzig, Germany
| | - Arved Weimann
- Department for General and Visceral Surgery, Hospital St. Georg Leipzig, Leipzig, Germany
| | - Guido Schumacher
- Department for General and Visceral Surgery, Hospital Braunschweig, Braunschweig, Germany
| | - Ingo Bechmann
- Institute of Anatomy, University of Leipzig, Leipzig
| | - Florian Lordick
- Department of Oncology, Gastroenterology, Hepatology, Pulmonology, and Infectious Diseases, University Cancer Center Leipzig, University Hospital Leipzig, Leipzig, Germany
| | | | - Justus Körfer
- Department of Oncology, Gastroenterology, Hepatology, Pulmonology, and Infectious Diseases, University Cancer Center Leipzig, University Hospital Leipzig, Leipzig, Germany
| |
Collapse
|
17
|
Park C, Jeong K, Park JH, Jung S, Bae JM, Kim K, Ock CY, Kim M, Keam B, Kim TM, Jeon YK, Lee SH, Lee JS, Kim DW, Kang GH, Chung DH, Heo DS. Pan-cancer methylation analysis reveals an inverse correlation of tumor immunogenicity with methylation aberrancy. Cancer Immunol Immunother 2021; 70:1605-1617. [PMID: 33230567 DOI: 10.1007/s00262-020-02796-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Accepted: 11/07/2020] [Indexed: 12/21/2022]
Abstract
Tumor immunogenicity is driven by various genomic and transcriptomic factors but the association with the overall status of methylation aberrancy is not well established. We analyzed The Cancer Genome Atlas pan-cancer database to investigate whether the overall methylation aberrancy links to the immune evasion of tumor. We created the definitions of hypermethylation burden, hypomethylation burden and methylation burden to establish the values that represent the degree of methylation aberrancy from human methylation 450 K array data. Both hypermethylation burden and hypomethylation burden significantly correlated with global methylation level as well as methylation subtypes defined in previous literatures. Then we evaluated whether methylation burden correlates with tumor immunogenicity and found that methylation burden showed a significant negative correlation with cytolytic activity score, which represent cytotoxic T cell activity, in pan-cancer (Spearman rho = - 0.37, p < 0.001) and 30 of 33 individual cancer types. Furthermore, this correlation was independent of mutation burden and chromosomal instability in multivariate regression analysis. We validated the findings in the external cohorts and outcomes of patients who were treated with immune checkpoint inhibitors, which showed that high methylation burden group had significantly poor progression-free survival (Hazard ratio 1.74, p = 0.038). Overall, the degree of methylation aberrancy negatively correlated with tumor immunogenicity. These findings emphasize the importance of methylation aberrancy for tumors to evade immune surveillance and warrant further development of methylation biomarker.
Collapse
Affiliation(s)
- Changhee Park
- Department of Internal Medicine, Seoul National University Hospital, 101 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea
| | - Kyeonghun Jeong
- Division of Clinical Bioinformatics, Biomedical Research Institute, Seoul National University Hospital, 101 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea
| | - Joon-Hyeong Park
- Division of Clinical Bioinformatics, Biomedical Research Institute, Seoul National University Hospital, 101 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea
| | - Sohee Jung
- Division of Clinical Bioinformatics, Biomedical Research Institute, Seoul National University Hospital, 101 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea
| | - Jeong Mo Bae
- Department of Pathology, Seoul National University Hospital, Seoul, Republic of Korea
| | - Kwangsoo Kim
- Transdisciplinary Department of Medicine & Advanced Technology, Seoul National University Hospital, Seoul, Republic of Korea.
| | - Chan-Young Ock
- Department of Internal Medicine, Seoul National University Hospital, 101 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea.
| | - Miso Kim
- Department of Internal Medicine, Seoul National University Hospital, 101 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea
| | - Bhumsuk Keam
- Department of Internal Medicine, Seoul National University Hospital, 101 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea
- Cancer Research Institute, Seoul National University, Seoul, Republic of Korea
| | - Tae Min Kim
- Department of Internal Medicine, Seoul National University Hospital, 101 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea
- Cancer Research Institute, Seoul National University, Seoul, Republic of Korea
| | - Yoon Kyung Jeon
- Department of Pathology, Seoul National University Hospital, Seoul, Republic of Korea
| | - Se-Hoon Lee
- Division of Hematology/Oncology, Department of Internal Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Ju-Seog Lee
- Department of Systems Biology, Division of Basic Sciences, MD Anderson Cancer Center, Houston, TX, USA
| | - Dong-Wan Kim
- Department of Internal Medicine, Seoul National University Hospital, 101 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea
- Cancer Research Institute, Seoul National University, Seoul, Republic of Korea
| | - Gyeong Hoon Kang
- Department of Pathology, Seoul National University Hospital, Seoul, Republic of Korea
| | - Doo Hyun Chung
- Department of Pathology, Seoul National University Hospital, Seoul, Republic of Korea
| | - Dae Seog Heo
- Department of Internal Medicine, Seoul National University Hospital, 101 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea
- Cancer Research Institute, Seoul National University, Seoul, Republic of Korea
| |
Collapse
|
18
|
Assessment of Immune Status in Dynamics for Patients with Cancer Undergoing Immunotherapy. JOURNAL OF ONCOLOGY 2021; 2021:6698969. [PMID: 34054956 PMCID: PMC8112935 DOI: 10.1155/2021/6698969] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Revised: 04/14/2021] [Accepted: 04/20/2021] [Indexed: 11/30/2022]
Abstract
Immunotherapy using immune checkpoint inhibitors has revolutionized the treatment, and many types of cancer show a response rate of 20–40% and a significant increase in five-year survival. However, immunotherapy is expensive and may cause serious adverse events. Therefore, a predictive method allowing identification of responding patients before starting the treatment would be very useful. In this study, we aimed to identify and implement other individual prognosis factors, factors that could lead to an improved clinical decision made in regard to the patient to establish an individualized treatment. Materials and Methods. All patients recruited from October 2018 to July 2019 were treated in OncoFort Hospital, Bucharest, with nivolumab or pembrolizumab. We investigated T lymphocyte CD3+, CD4+, CD8+, and CD4/CD8 cells by flow cytometry in patients before and after receiving treatment with anti-PD-1 agents. Results. We found that the responder group showed higher expression on CD4+ cells than the nonresponder group after the first cycle of immunotherapy. The prediction of the immunotherapeutic effect revealed that the elevation of T lymphocytes CD8+ and CD4+ after the first cycle of immunotherapy was followed by a decrease in their expression after the second cycle and was followed by a return almost to that one after the first administration. Conclusion. Our work indicates that the evaluation of the cells of the immune system in relation to the tumor and immunotherapy may lead to a better understanding of the pathogenic mechanisms and the identification of prognostic and predictive factors that will more effectively model the therapeutic approach.
Collapse
|
19
|
Bagherifar R, Kiaie SH, Hatami Z, Ahmadi A, Sadeghnejad A, Baradaran B, Jafari R, Javadzadeh Y. Nanoparticle-mediated synergistic chemoimmunotherapy for tailoring cancer therapy: recent advances and perspectives. J Nanobiotechnology 2021; 19:110. [PMID: 33865432 PMCID: PMC8052859 DOI: 10.1186/s12951-021-00861-0] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Accepted: 04/09/2021] [Indexed: 12/15/2022] Open
Abstract
Nowadays, a potent challenge in cancer treatment is considered the lack of efficacious strategy, which has not been able to significantly reduce mortality. Chemoimmunotherapy (CIT) as a promising approach in both for the first-line and relapsed therapy demonstrated particular benefit from two key gating strategies, including chemotherapy and immunotherapy to cancer therapy; therefore, the discernment of their participation and role of potential synergies in CIT approach is determinant. In this study, in addition to balancing the pros and cons of CIT with the challenges of each of two main strategies, the recent advances in the cancer CIT have been discussed. Additionally, immunotherapeutic strategies and the immunomodulation effect induced by chemotherapy, which boosts CIT have been brought up. Finally, harnessing and development of the nanoparticles, which mediated CIT have expatiated in detail.
Collapse
Affiliation(s)
- Rafieh Bagherifar
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Pharmaceutics, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Seyed Hossein Kiaie
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
- Nano Drug Delivery Research Center, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Zahra Hatami
- Department of Immunology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Armin Ahmadi
- Department of Chemical & Materials Engineering, The University of Alabama in Huntsville, Huntsville, AL, 35899, USA
| | | | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Reza Jafari
- Solid Tumor Research Center, Cellular and Molecular Medicine Institute, Urmia University of Medical Sciences, Shafa St, Ershad Blvd., P.O. BoX: 1138, 57147, Urmia, Iran.
- Department of Immunology and Genetics, School of Medicine, Urmia University of Medical Sciences, Urmia, Iran.
| | - Yousef Javadzadeh
- Biotechnology Research Center, and Faculty of Pharmacy, Tabriz University of Medical Science, 5166-15731, Tabriz, Iran.
| |
Collapse
|
20
|
Jindal A, Sarkar S, Alam A. Nanomaterials-Mediated Immunomodulation for Cancer Therapeutics. Front Chem 2021; 9:629635. [PMID: 33708759 PMCID: PMC7940769 DOI: 10.3389/fchem.2021.629635] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2020] [Accepted: 01/18/2021] [Indexed: 12/16/2022] Open
Abstract
Immunotherapy holds great promise in overcoming the limitations of conventional regimens for cancer therapeutics. There is growing interest among researchers and clinicians to develop novel immune-strategies for cancer diagnosis and treatment with better specificity and lesser adversity. Immunomodulation-based cancer therapies are rapidly emerging as an alternative approach that employs the host's own defense mechanisms to recognize and selectively eliminate cancerous cells. Recent advances in nanotechnology have pioneered a revolution in the field of cancer therapy. Several nanomaterials (NMs) have been utilized to surmount the challenges of conventional anti-cancer treatments like cytotoxic chemotherapy, radiation, and surgery. NMs offer a plethora of exceptional features such as a large surface area to volume ratio, effective loading, and controlled release of active drugs, tunable dimensions, and high stability. Moreover, they also possess the inherent property of interacting with living cells and altering the immune responses. However, the interaction between NMs and the immune system can give rise to unanticipated adverse reactions such as inflammation, necrosis, and hypersensitivity. Therefore, to ensure a successful and safe clinical application of immunomodulatory nanomaterials, it is imperative to acquire in-depth knowledge and a clear understanding of the complex nature of the interactions between NMs and the immune system. This review is aimed at providing an overview of the recent developments, achievements, and challenges in the application of immunomodulatory nanomaterials (iNMs) for cancer therapeutics with a focus on elucidating the mechanisms involved in the interplay between NMs and the host's immune system.
Collapse
Affiliation(s)
- Ajita Jindal
- School of Biotechnology, Jawaharlal Nehru University, New Delhi, India
| | - Sounik Sarkar
- Flowcytometry Facility, Modern Biology Department, University of Calcutta, Kolkata, India
| | - Aftab Alam
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, United Kingdom
- Clare Hall, University of Cambridge, Cambridge, United Kingdom
- Charles River Laboratories, Cambridge Biomedical Campus, Cambridge, United Kingdom
| |
Collapse
|
21
|
Frontiers in combining immune checkpoint inhibitors for advanced urothelial cancer management. Curr Opin Urol 2020; 30:457-466. [PMID: 32235284 DOI: 10.1097/mou.0000000000000765] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
PURPOSE OF REVIEW This review provides an overview of currently ongoing clinical trials evaluating the combination of immune checkpoint inhibitors (CPI) with other therapies in locally advanced or metastatic urothelial cancer and the rationale for this combination approach. We discuss the preliminary results from early data presented at recent meetings regarding the efficacy and safety of novel combination therapies including a CPI for metastatic urothelial cancer. RECENT FINDINGS CPI emerged as novel first-line or second-line treatment options in advanced and metastatic urothelial cancer (mUC). Although the response rates and their sustainability are promising, it is far from a home run. Combination therapies have already shown improved efficacy in several other tumor entities. SUMMARY Numerous clinical trials currently investigate combinations of CPI with other CPI, previously established systemic chemotherapy, targeted therapies, vaccines, or accompanied with radiotherapy. Preliminary data shows promising results. These results suggest that targeting pathways of immune response combined with established or novel oncological therapies may lead to a synergistic antitumor effect.
Collapse
|
22
|
Shofolawe-Bakare OT, Stokes LD, Hossain M, Smith AE, Werfel TA. Immunostimulatory biomaterials to boost tumor immunogenicity. Biomater Sci 2020; 8:5516-5537. [PMID: 33049007 PMCID: PMC7837217 DOI: 10.1039/d0bm01183e] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Cancer immunotherapy is exhibiting great promise as a new therapeutic modality for cancer treatment. However, immunotherapies are limited by the inability of some tumors to provoke an immune response. These tumors with a 'cold' immunological phenotype are characterized by low numbers of tumor-infiltrating lymphocytes, high numbers of immunosuppressive leukocytes (e.g. regulatory T cells, tumor-associated macrophages), and high production of immune-dampening signals (e.g. IL-10, TGF-β, IDO-1). Strategies to boost the aptitude of tumors to initiate an immune response (i.e. boost tumor immunogenicity) will turn 'cold' tumors 'hot' and augment the anti-tumor efficacy of current immunotherapies. Approaches to boost tumor immunogenicity already show promise; however, multifaceted delivery and immunobiology challenges exist. For instance, systemic delivery of many immune-stimulating agents causes off-target toxicity and/or the development of autoimmunity, limiting the administrable dose below the threshold needed to achieve efficacy. Moreover, once administered in vivo, molecules such as the nucleic acid-based agonists for many pattern recognition receptors are either rapidly cleared or degraded, and don't efficiently traffic to the intracellular compartments where the receptors are located. Thus, these nucleic acid-based drugs are ineffective without a delivery system. Biomaterials-based approaches aim to enhance current strategies to boost tumor immunogenicity, enable novel strategies, and spare dose-limiting toxicities. Here, we review recent progress to improve cancer immunotherapies by boosting immunogenicity within tumors using immunostimulatory biomaterials.
Collapse
|
23
|
Jyoti K, Katare OP, Kamboj A, Madan J. Protamine sulphate coated poly (lactide-co-glycolide) nanoparticles of MUC-1 peptide improved cellular uptake and cytokine release in mouse antigen presenting cells. J Microencapsul 2020; 37:566-576. [PMID: 32928025 DOI: 10.1080/02652048.2020.1823500] [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: 10/23/2022]
Abstract
AIM MUC-1-peptide (M-1-pep) loaded poly (lactide-co-glycolide) nanoparticles were coated with protamine sulphate (PS), M-1-pep-PS-P-NPs for targeting antigen presenting cells (APCs) to evoke cytokine release. METHODS AND RESULTS M-1-pep-PS-P-NPs were tailored by emulsion-diffusion evaporation method and characterised in vitro under a set of rigorous parameters. The average particle size and zeta potential of optimised M-1-pep-PS-P-B-NPs was measured to be 132.21 ± 30.71 nm and 6.29 ± 0.71 mV, significantly (p < 0.01) higher than 71.24 ± 17.76-nm and -43.41 ± 3.37 mV of M-1-pep-P-NPs. Further, 50-μg/ml concentration of M-1-pep-PS-P-B-NPs displayed 82.4% cellular uptake in RAW 264.7 cells calculated in setting of fluorescence intensity significantly (p < 0.05) elevated than 63.1% of M-1-pep-P-NPs. Consistent to quantitative results, M-1-pep-PS-P-B-NPs also confirmed advanced cellular uptake (CU) in RAW 264.7 cells in contrast to M-1-pep-P-NPs suppose to be through multiple mechanisms including phagocytosis and clathrin mediated endocytosis. CONCLUSION M-1-pep-PS-P-B-NPs must be evaluated in vivo through inhalation route of administration for antitumor prospective in lung cancer xenograft model.
Collapse
Affiliation(s)
- Kiran Jyoti
- Research Division, IKG Punjab Technical University, Jalandhar, India.,Department of Pharmaceutics, Chandigarh College of Pharmacy, Mohali, India
| | - Om Prakash Katare
- University Institute of Pharmaceutical Sciences, Punjab University, Chandigarh, India
| | - Anjoo Kamboj
- Department of Pharmaceutics, Chandigarh College of Pharmacy, Mohali, India
| | - Jitender Madan
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Hyderabad, India
| |
Collapse
|
24
|
Cocks M, Chaux A, Jenson EG, Miller JA, Rodriguez Pena MDC, Tregnago AC, Taheri D, Eich ML, Sharma R, Vang R, Netto GJ. Immune checkpoint status and tumor microenvironment in vulvar squamous cell carcinoma. Virchows Arch 2020; 477:93-102. [PMID: 31993774 DOI: 10.1007/s00428-020-02759-y] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Revised: 12/31/2019] [Accepted: 01/21/2020] [Indexed: 12/31/2022]
Abstract
Vulvar squamous cell carcinoma accounts for 5% of cancers of the female genital tract. Current guidelines recommend wide local excision with negative surgical margins as the standard treatment. However, the extent of the tumor-free resection margin after wide local excision is still controversial in many cases. Drugs targeting immune checkpoints such as PD-1 or its ligand PD-L1 have potential clinical utility in these patients. We examined the expression of PD-L1 in tumor cells and immune cells, as well as the proportion of PD-1, CD8, and FOXP3 positive lymphocytes. Twenty-one cases of invasive vulvar squamous cell carcinomas were reviewed. Whole slides of representative formalin-fixed, paraffin-embedded archival material were used for analysis. Odds ratios (OR) and hazard ratios (HR) were used to estimate risk for disease recurrence, overall mortality, and cancer mortality. PD-L1 expression was found in 43% of tumor cells, with higher proportions in intratumoral (67%) and peritumoral (81%) immune cells. OR and HR for disease recurrence and cancer mortality were higher in tumors with higher CD8 expression. OR and HR for overall mortality were also higher in tumors with higher PD-L1 and CD8 expression. In conclusion, nearly half of cases were PD-L1 positive in tumor cells with over two-third of cases demonstrating PD-L1 positivity in immune cells. Immunohistochemical expression of PD-L1 and CD8 could be used to suggest higher risk of disease recurrence, overall mortality, and cancer mortality. Furthermore, our data contributes to the growing evidence that targeting the PD-1/PD-L1 pathway may be beneficial in vulvar squamous cell carcinomas.
Collapse
Affiliation(s)
- Margaret Cocks
- Department of Pathology, The Johns Hopkins Hospital, Baltimore, MD, USA
| | - Alcides Chaux
- Department of Scientific Research, School of Postgraduate Studies, Norte University, Asunción, Paraguay
| | - Erik G Jenson
- Department of Pathology, The Johns Hopkins Hospital, Baltimore, MD, USA
| | - James A Miller
- Department of Pathology, The Johns Hopkins Hospital, Baltimore, MD, USA
| | - Maria Del Carmen Rodriguez Pena
- Department of Pathology, The University of Alabama at Birmingham, West Pavilion P210, 619 19th Street, South Birmingham, AL, 35249-7331, USA
| | - Aline C Tregnago
- Department of Pathology, The Johns Hopkins Hospital, Baltimore, MD, USA
| | - Diana Taheri
- Department of Pathology, The Johns Hopkins Hospital, Baltimore, MD, USA
- Isfahan Kidney Diseases Research Center, Pathology, Isfahan University of Medical Sciences, Isfahan, Islamic Republic of Iran
| | - Marie-Lisa Eich
- Department of Pathology, The University of Alabama at Birmingham, West Pavilion P210, 619 19th Street, South Birmingham, AL, 35249-7331, USA
- Department of Pathology, University Hospital Cologne, Cologne, Germany
| | - Rajni Sharma
- Department of Pathology, The Johns Hopkins Hospital, Baltimore, MD, USA
| | - Russell Vang
- Department of Pathology, The Johns Hopkins Hospital, Baltimore, MD, USA
| | - George J Netto
- Department of Pathology, The University of Alabama at Birmingham, West Pavilion P210, 619 19th Street, South Birmingham, AL, 35249-7331, USA.
| |
Collapse
|
25
|
Roles for receptor tyrosine kinases in tumor progression and implications for cancer treatment. Adv Cancer Res 2020; 147:1-57. [PMID: 32593398 DOI: 10.1016/bs.acr.2020.04.002] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Growth factors and their receptor tyrosine kinases (RTKs), a group of transmembrane molecules harboring cytoplasm-facing tyrosine-specific kinase functions, play essential roles in migration of multipotent cell populations and rapid proliferation of stem cells' descendants, transit amplifying cells, during embryogenesis and tissue repair. These intrinsic functions are aberrantly harnessed when cancer cells undergo intertwined phases of cell migration and proliferation during cancer progression. For example, by means of clonal expansion growth factors fixate the rarely occurring driver mutations, which initiate tumors. Likewise, autocrine and stromal growth factors propel angiogenesis and penetration into the newly sprouted vessels, which enable seeding micro-metastases at distant organs. We review genetic and other mechanisms that preempt ligand-mediated activation of RTKs, thereby supporting sustained cancer progression. The widespread occurrence of aberrant RTKs and downstream signaling pathways in cancer, identifies molecular targets suitable for pharmacological intervention. We list all clinically approved cancer drugs that specifically intercept oncogenic RTKs. These are mainly tyrosine kinase inhibitors and monoclonal antibodies, which can inhibit cancer but inevitably become progressively less effective due to adaptive rewiring processes or emergence of new mutations, processes we overview. Similarly important are patient treatments making use of radiation, chemotherapeutic agents and immune checkpoint inhibitors. The many interfaces linking RTK-targeted therapies and these systemic or local regimens are described in details because of the great promise offered by combining pharmacological modalities.
Collapse
|
26
|
Immunotherapy Approaches for Pediatric CNS Tumors and Associated Neurotoxicity. Pediatr Neurol 2020; 107:7-15. [PMID: 32113728 DOI: 10.1016/j.pediatrneurol.2020.01.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 12/07/2019] [Accepted: 01/15/2020] [Indexed: 12/24/2022]
Abstract
Treatment for brain tumors has recently shifted to using the power of the immune system to destroy cancer cells with promising results. Many immunotherapeutic approaches that have been used in adults, including checkpoint inhibitors, vaccine therapy, adoptive immunotherapy, such as chimeric antigen receptor T cell therapy, and viral therapy, are now being evaluated in children. Although these treatments work through different mechanisms, they all activate the immune system and can result in inflammation at the site of disease. This can be especially problematic in the confined area of the brain causing potentially severe neurological side effects, which are of special concern in children with central nervous system malignancies. Steroids can be helpful in the management of neurological complications but carry the risk of making immunotherapeutic approaches less effective. Alternative therapeutic interventions to mitigate side effects are being evaluated. This review describes the most common immunotherapeutic modalities that are now under study for the treatment of pediatric brain tumors, their rationale, associated neurotoxicities, and current management.
Collapse
|
27
|
Jyoti K, Jain S, Katare OP, Katyal A, Chandra R, Madan J. Non-small cell lung cancer tumour antigen, MUC-1 peptide-loaded non-aggregated poly (lactide-co-glycolide) nanoparticles augmented cellular uptake in mouse professional antigen-presenting cells: optimisation and characterisation. J Microencapsul 2019; 37:14-28. [DOI: 10.1080/02652048.2019.1692943] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Affiliation(s)
- Kiran Jyoti
- Department of Pharmaceutics, Chandigarh College of Pharmacy, Mohali, Punjab, India
- IK Gujral Punjab Technical University, Jalandhar, Punjab, India
| | - Sanyog Jain
- Centre for Pharmaceutical Nanotechnology, Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, SAS Nagar, Punjab, India
| | - Om Prakash Katare
- University Institute of Pharmaceutical Sciences, Punjab University, Chandigarh, India
| | - Anju Katyal
- Dr. B. R Ambedkar Centre for Biomedical Research, University of Delhi, Delhi, India
| | - Ramesh Chandra
- Dr. B. R Ambedkar Centre for Biomedical Research, University of Delhi, Delhi, India
- Department of Chemistry, University of Delhi, Delhi, India
| | - Jitender Madan
- Department of Pharmaceutics, Chandigarh College of Pharmacy, Mohali, Punjab, India
| |
Collapse
|
28
|
Vianna P, Mendes MF, Bragatte MA, Ferreira PS, Salzano FM, Bonamino MH, Vieira GF. pMHC Structural Comparisons as a Pivotal Element to Detect and Validate T-Cell Targets for Vaccine Development and Immunotherapy-A New Methodological Proposal. Cells 2019; 8:E1488. [PMID: 31766602 PMCID: PMC6952977 DOI: 10.3390/cells8121488] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Revised: 11/15/2019] [Accepted: 11/16/2019] [Indexed: 12/02/2022] Open
Abstract
The search for epitopes that will effectively trigger an immune response remains the "El Dorado" for immunologists. The development of promising immunotherapeutic approaches requires the appropriate targets to elicit a proper immune response. Considering the high degree of HLA/TCR diversity, as well as the heterogeneity of viral and tumor proteins, this number will invariably be higher than ideal to test. It is known that the recognition of a peptide-MHC (pMHC) by the T-cell receptor is performed entirely in a structural fashion, where the atomic interactions of both structures, pMHC and TCR, dictate the fate of the process. However, epitopes with a similar composition of amino acids can produce dissimilar surfaces. Conversely, sequences with no conspicuous similarities can exhibit similar TCR interaction surfaces. In the last decade, our group developed a database and in silico structural methods to extract molecular fingerprints that trigger T-cell immune responses, mainly referring to physicochemical similarities, which could explain the immunogenic differences presented by different pMHC-I complexes. Here, we propose an immunoinformatic approach that considers a structural level of information, combined with an experimental technology that simulates the presentation of epitopes for a T cell, to improve vaccine production and immunotherapy efficacy.
Collapse
Affiliation(s)
- Priscila Vianna
- Laboratory of Human Teratogenesis and Population Medical Genetics, Department of Genetics, Institute of Biosciences, Federal University of Rio Grande do Sul, Porto Alegre 91.501-970, Brazil;
| | - Marcus F.A. Mendes
- Laboratory of Bioinformatics (NBLI), Department of Genetics, Institute of Biosciences, Federal University of Rio Grande do Sul, Porto Alegre 91.501-970, Brazil (M.A.B.)
| | - Marcelo A. Bragatte
- Laboratory of Bioinformatics (NBLI), Department of Genetics, Institute of Biosciences, Federal University of Rio Grande do Sul, Porto Alegre 91.501-970, Brazil (M.A.B.)
| | - Priscila S. Ferreira
- Program of Immunology and Tumor Biology, Division of Experimental and Translational Research, Brazilian National Cancer Institute, Rio de Janeiro 20231-050, Brazil; (P.S.F.); (M.H.B.)
| | - Francisco M. Salzano
- Laboratory of Molecular Evolution, Department of Genetics, Institute of Biosciences, Federal University of Rio Grande do Sul, Porto Alegre 91.501-970, Brazil;
| | - Martin H. Bonamino
- Program of Immunology and Tumor Biology, Division of Experimental and Translational Research, Brazilian National Cancer Institute, Rio de Janeiro 20231-050, Brazil; (P.S.F.); (M.H.B.)
- Vice Presidency of Research and Biological Collections, Fundação Oswaldo Cruz, Rio de Janeiro 21040-900, Brazil
| | - Gustavo F. Vieira
- Laboratory of Bioinformatics (NBLI), Department of Genetics, Institute of Biosciences, Federal University of Rio Grande do Sul, Porto Alegre 91.501-970, Brazil (M.A.B.)
- Laboratory of Health Bioinformatics, Post Graduate Program in Health and Human Development, La Salle University, Canoas 91.501-970, Brazil
| |
Collapse
|
29
|
Targeting Negative and Positive Immune Checkpoints with Monoclonal Antibodies in Therapy of Cancer. Cancers (Basel) 2019; 11:cancers11111756. [PMID: 31717326 PMCID: PMC6895894 DOI: 10.3390/cancers11111756] [Citation(s) in RCA: 91] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Revised: 11/05/2019] [Accepted: 11/06/2019] [Indexed: 02/06/2023] Open
Abstract
The immune checkpoints are regulatory molecules that maintain immune homeostasis in physiological conditions. By sending T cells a series of co-stimulatory or co-inhibitory signals via receptors, immune checkpoints can both protect healthy tissues from adaptive immune response and activate lymphocytes to remove pathogens effectively. However, due to their mode of action, suppressive immune checkpoints may serve as unwanted protection for cancer cells. To restore the functioning of the immune system and make the patient’s immune cells able to recognize and destroy tumors, monoclonal antibodies are broadly used in cancer immunotherapy to block the suppressive or to stimulate the positive immune checkpoints. In this review, we aim to present the current state of application of monoclonal antibodies in clinics, used either as single agents or in a combined treatment. We discuss the limitations of these therapies and possible problem-solving with combined treatment approaches involving both non-biological and biological agents. We also highlight the most promising strategies based on the use of monoclonal or bispecific antibodies targeted on immune checkpoints other than currently implemented in clinics.
Collapse
|
30
|
Menz BD, Johnson JL, Gillard DF, Chong W, Ward MB. The Role of the Pharmacist in Optimizing Cancer Immunotherapy: A Retrospective Study of Nivolumab Adverse Events. J Pharm Pract 2019; 34:386-396. [PMID: 33969772 DOI: 10.1177/0897190019872937] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
BACKGROUND Immune checkpoint inhibitors (ICIs) are an emerging treatment in cancer therapy for prolonging life, minimizing symptoms, and selectively targeting cancer. Program death 1 (PD-1) inhibitors, such as nivolumab, fall within this class, enabling the patient's immune system to detect and destroy cancer. The introduction of ICIs is changing cancer therapy, with new drugs and new toxicities-an evolving area encountered by pharmacists. OBJECTIVE This study aims to compare the pattern of nivolumab-induced adverse events observed in practice, when compared with clinical trial and literature data. The secondary aim of the study is to identify the presentation and treatment modalities initiated in practice. METHODS We performed a retrospective case note review across 2 South Australian hospitals to identify the common toxicities and symptomatic treatments experienced by patients receiving nivolumab. Results were compared with clinical trial data from product innovator Bristol-Myer Squib and other published literature. RESULTS Seventy patients were included in the study; of these, 60 (86%) experienced any grade adverse event(s). A total of 59 (84%) of 70 experienced mild to moderate grade 1 to grade 2 adverse events and 10 (14%) of 70 patients experienced severe grade 3 to grade 4 adverse events, displaying some consistencies with clinical trial and published literature data. Together, the prevalence of adverse events with details on presentation and treatments illustrates possible pharmacy practice strategies and areas for intervention. CONCLUSIONS The listed prevalence of adverse events and practice strategies identified throughout this study highlights how pharmacists may assist in the identification of predictable ICI toxicities associated with gastrointestinal, endocrine, dermatological toxicities, and fatigue.
Collapse
Affiliation(s)
- Bradley D Menz
- School of Pharmacy and Medical Sciences, 1067University of South Australia, North Terrace, Adelaide, South Australia, Australia.,375072Central Adelaide Local Health Network, Royal Adelaide Hospital, SA Pharmacy, North Terrace, Adelaide, South Australia, Australia.,6677Southern Adelaide Local Health Network, Flinders Medical Centre, SA Pharmacy, Bedford Park, South Australia, Australia
| | - Jacinta L Johnson
- School of Pharmacy and Medical Sciences, 1067University of South Australia, North Terrace, Adelaide, South Australia, Australia.,6677Southern Adelaide Local Health Network, Flinders Medical Centre, SA Pharmacy, Bedford Park, South Australia, Australia
| | - Davina F Gillard
- 375072Central Adelaide Local Health Network, Royal Adelaide Hospital, SA Pharmacy, North Terrace, Adelaide, South Australia, Australia
| | - William Chong
- 6677Southern Adelaide Local Health Network, Flinders Medical Centre, SA Pharmacy, Bedford Park, South Australia, Australia
| | - Michael B Ward
- School of Pharmacy and Medical Sciences, 1067University of South Australia, North Terrace, Adelaide, South Australia, Australia.,375072Central Adelaide Local Health Network, Royal Adelaide Hospital, SA Pharmacy, North Terrace, Adelaide, South Australia, Australia
| |
Collapse
|
31
|
Marchini A, Daeffler L, Pozdeev VI, Angelova A, Rommelaere J. Immune Conversion of Tumor Microenvironment by Oncolytic Viruses: The Protoparvovirus H-1PV Case Study. Front Immunol 2019; 10:1848. [PMID: 31440242 PMCID: PMC6692828 DOI: 10.3389/fimmu.2019.01848] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Accepted: 07/23/2019] [Indexed: 12/21/2022] Open
Abstract
Cancer cells utilize multiple mechanisms to evade and suppress anticancer immune responses creating a “cold” immunosuppressive tumor microenvironment. Oncolytic virotherapy is emerging as a promising approach to revert tumor immunosuppression and enhance the efficacy of other forms of immunotherapy. Growing evidence indicates that oncolytic viruses (OVs) act in a multimodal fashion, inducing immunogenic cell death and thereby eliciting robust anticancer immune responses. In this review, we summarize information about OV-mediated immune conversion of the tumor microenvironment. As a case study we focus on the rodent protoparvovirus H-1PV and its dual role as an oncolytic and immune modulatory agent. Potential strategies to improve H-1PV anticancer efficacy are also discussed.
Collapse
Affiliation(s)
- Antonio Marchini
- Laboratory of Oncolytic Virus Immuno-Therapeutics, Luxembourg Institute of Health, Luxembourg, Luxembourg.,Laboratory of Oncolytic Virus Immuno-Therapeutics, German Cancer Research Center, Heidelberg, Germany
| | - Laurent Daeffler
- Université de Strasbourg, IPHC, Strasbourg, France.,CNRS, UMR7178, Strasbourg, France
| | - Vitaly I Pozdeev
- Laboratory of Oncolytic Virus Immuno-Therapeutics, Luxembourg Institute of Health, Luxembourg, Luxembourg
| | - Assia Angelova
- Infection, Inflammation and Cancer Program, German Cancer Research Center, Heidelberg, Germany
| | - Jean Rommelaere
- Infection, Inflammation and Cancer Program, German Cancer Research Center, Heidelberg, Germany
| |
Collapse
|
32
|
Teufel A, Zhan T, Härtel N, Bornschein J, Ebert MP, Schulte N. Management of immune related adverse events induced by immune checkpoint inhibition. Cancer Lett 2019; 456:80-87. [DOI: 10.1016/j.canlet.2019.04.018] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Revised: 04/02/2019] [Accepted: 04/09/2019] [Indexed: 12/17/2022]
|
33
|
Shan T, Chen S, Wu T, Yang Y, Li S, Chen X. PD-L1 expression in colon cancer and its relationship with clinical prognosis. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2019; 12:1764-1769. [PMID: 31933995 PMCID: PMC6947132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Accepted: 01/23/2018] [Indexed: 06/10/2023]
Abstract
OBJECTIVES PD-L1 is closely associated with tumorigenesis and development. However, expression of PD-L1 protein in colon cancer and its significance in clinical prognosis are yet to be fully clarified. This study examined the relationship between PD-L1 expression with the clinicopathological features and prognosis of colon cancer. METHODS This study collected cases of primary colon cancer that had not undergone preoperative chemotherapy and had complete clinical data. Eighty specimens each were obtained from cancer tissues, paracancer tissues, and normal tissues. Immunohistochemical assays were performed to detect PD-L1 expression. The relationship between PD-L1 expression and clinicopathologic features was compared. This was combined with follow-up data, to analyze the relationship between positive or negative PD-L1 expression and prognosis. RESULTS Among 80 tumor specimens, 22 cases (27.5%) showed high PD-L1 expression, 24 cases (30.0%) showed moderate expression, and 34 cases (42.5%) showed weak or no PD-L1 staining. High expression of PD-L1 in paracancer and normal tissues were 9 (11.3%) and 5 (6.3%) cases, respectively. PD-L1 expression was also positively correlated with TNM stage (P=0.009), lymph node metastasis (P=0.000), distant metastasis (P=0.014). There were no significant differences in different age, gender, histologic grade, and tumor size groups (P>0.05). Regression analysis revealed that poorer tumor differentiation, later TNM stages, presence of lymph node metastasis, and positive PD-L1 expression were factors that influenced prognosis. Multivariate analysis indicated that late TNM stage and positive PD-L1 expression were independent risk factors that influenced prognosis. CONCLUSION PD-L1 expression is significantly elevated in colon cancer tissues, and is closely associated with lymph node metastasis, prognosis, and survival.
Collapse
Affiliation(s)
- Tao Shan
- Department of General Surgery, The Second Affiliated Hospital of Medical College, Xi'an Jiaotong University Xi'an 710004, Shaanxi, China
| | - Shuo Chen
- Department of General Surgery, The Second Affiliated Hospital of Medical College, Xi'an Jiaotong University Xi'an 710004, Shaanxi, China
| | - Tao Wu
- Department of General Surgery, The Second Affiliated Hospital of Medical College, Xi'an Jiaotong University Xi'an 710004, Shaanxi, China
| | - Yi Yang
- Department of General Surgery, The Second Affiliated Hospital of Medical College, Xi'an Jiaotong University Xi'an 710004, Shaanxi, China
| | - Shunle Li
- Department of General Surgery, The Second Affiliated Hospital of Medical College, Xi'an Jiaotong University Xi'an 710004, Shaanxi, China
| | - Xi Chen
- Department of General Surgery, The Second Affiliated Hospital of Medical College, Xi'an Jiaotong University Xi'an 710004, Shaanxi, China
| |
Collapse
|
34
|
Abstract
Recent reports describe how genome-wide transcriptional analysis of cancer tissues can be exploited to identify molecular signatures of immune infiltration in cancer. We hypothesize that immune infiltration in cancer may also be defined by changes in certain epigenetic signatures. In this context, a primary objective is to identify site-specific CpG markers whose levels of methylation may be highly indicative of known transcriptional markers of immune infiltration such as GZMA, PRF1, T cell receptor genes, PDCD1, and CTLA4. This has been accomplished by integrating genome-wide transcriptional expression and methylation data for different types of cancer (melanoma, kidney cancers, lung cancers, gliomas, head and neck cancer). Our findings establish that cancers of related histology also have a high degree of similarity in immune-infiltration CpG markers. For example, the epigenetic immune infiltration signatures in lung adenocarcinoma (LUAD), mesothelioma (MESO), lung squamous cell carcinoma (LUSC), and head and neck squamous cell carcinoma (HNSC) are distinctly similar. So are glioblastoma multiforme (GBM) and brain lower grade glioma (LGG); and kidney renal papillary cell carcinoma (KIRP) and kidney renal clear cell carcinoma (KIRC). Kidney chromophobe (KICH), on the other hand has markers that are unique to this cohort. The strong relationships between immune infiltration and CpG methylation (for certain sites) in cancer tissues were not observed upon integrated analysis of publicly available cancer cell line datasets. Results from comparative pathways analyses offer further justification to methylation at certain CpG sites as being indicators of cancer immune infiltration, and possibly of predicting patient response to immunotherapeutic drugs. Achieving this target objective would significantly enhance therapeutic outcomes employing immunotherapy through focused patient-centric personalized medicine.
Collapse
|
35
|
Abstract
Immune checkpoint inhibitors, mainly drugs targeting the programmed cell death 1
(PD-1)/programmed cell death ligand 1 (PD-L1) and cytotoxic T-lymphocyte antigen
4 (CTLA4) pathways, represent a remarkable advance in lung cancer treatment.
Immune checkpoint inhibitors targeting PD-1 and PD-L1 are approved for the
treatment of patients with non-small-cell lung cancer, with impressive clinical
activity and durable responses in some patients. This review will summarize the
mechanism of action of these drugs, the clinical development of these agents and
the current role of these agents in the management of patients with lung cancer.
In addition, the review will discuss the role of predictive biomarkers for
optimal patient selection for immunotherapy and management of autoimmune side
effects of these agents.
Collapse
|
36
|
Bacolod MD, Barany F, Pilones K, Fisher PB, de Castro RJ. Pathways- and epigenetic-based assessment of relative immune infiltration in various types of solid tumors. Adv Cancer Res 2019; 142:107-143. [PMID: 30885360 DOI: 10.1016/bs.acr.2019.01.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Recent clinical studies document the power of immunotherapy in treating subsets of patients with advanced cancers. In this context and with multiple cancer immunotherapeutics already evaluated in the clinic and a large number in various stages of clinical trials, it is imperative to comprehensively examine genomics data to better comprehend the role of immunity in different cancers in predicting response to therapy and in directing appropriate therapies. The approach we chose is to scrutinize the pathways and epigenetic factors predicted to drive immune infiltration in different cancer types using publicly available TCGA transcriptional and methylation datasets, along with accompanying clinico-pathological data. We observed that the relative activation of T cells and other immune signaling pathways differs across cancer types. For example, pathways related to activation and proliferation of helper and cytotoxic T cells appear to be more highly enriched in kidney, skin, head and neck, and esophageal cancers compared to those of lung, colorectal, and liver or bile duct cancers. The activation of these immune-related pathways positively associated with prognosis in certain cancer types, most notably melanoma, head and neck, and cervical cancers. Integrated methylation and expression data (along with publicly available, ENCODE-generated histone ChIP Seq and DNAse hypersensitivity data) predict that epigenetic regulation is a primary factor driving transcriptional activation of a number of genes crucial to immunity in cancer, including T cell receptor genes (e.g., CD3D, CD3E), CTLA4, and GZMA. However, the extent to which epigenetic factors (primarily methylation at promoter regions) affect transcription of immune-related genes may vary across cancer types. For example, there is a high negative correlation between promoter CpG methylation and CD3D expression in renal and thyroid cancers, but not in brain tumors. The types of analyses we have undertaken provide insights into the relationships between immune modulation and cancer etiology and progression, offering clues into ways of therapeutically manipulating the immune system to promote immune recognition and immunotherapy.
Collapse
Affiliation(s)
- Manny D Bacolod
- Department of Microbiology and Immunology, Weill Cornell Medicine, New York, NY, United States.
| | - Francis Barany
- Department of Microbiology and Immunology, Weill Cornell Medicine, New York, NY, United States
| | - Karsten Pilones
- Department of Radiation Oncology, Weill Cornell Medicine, New York, NY, United States
| | - Paul B Fisher
- Department of Human and Molecular Genetics, VCU Institute of Molecular Medicine, VCU Massey Cancer Center, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States
| | | |
Collapse
|
37
|
Mandarano M, Bellezza G, Belladonna ML, Van den Eynde BJ, Chiari R, Vannucci J, Mondanelli G, Ludovini V, Ferri I, Bianconi F, Del Sordo R, Cagini L, Albini E, Metro G, Puma F, Sidoni A. Assessment of TILs, IDO-1, and PD-L1 in resected non-small cell lung cancer: an immunohistochemical study with clinicopathological and prognostic implications. Virchows Arch 2018; 474:159-168. [PMID: 30448912 DOI: 10.1007/s00428-018-2483-1] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Revised: 10/31/2018] [Accepted: 11/05/2018] [Indexed: 12/18/2022]
Abstract
Several cancers, especially non-small cell lung cancer (NSCLC), are able to escape the immunosurveillance of tumor-infiltrating lymphocytes (TILs); among the molecules involved, the indoleamine 2,3-dioxygenase 1 (IDO-1) and the programmed cell death ligand-1 (PD-L1) play a crucial role. These aspects are of great interest in the current immunotherapeutic era, therefore the current study analyses the TILs, IDO-1, and PD-L1 interactions and their correlations with clinicopathological parameters and prognosis in NSCLC. One hundred ninety-three NSCLC surgical specimens, formalin-fixed, and paraffin-embedded were assessed for TILs density, TILs localization, IDO-1 (clone 4.16H1), and PD-L1 (clone E1L3N) immunohistochemical expressions. This data was correlated with clinicopathological parameters, disease free, and overall survivals. IDO-1 and PD-L1 high expressions were related to the solid pattern of adenocarcinomas (respectively p = 0.036 and p = 0.026); high PD-L1 expression was correlated with squamous histotype (p = 0.048). IDO-1 overexpression correlated with former smokers (p = 0.041), higher adenocarcinoma stages (p = 0.039), and with both higher TILs density and PD-L1 expression (respectively p = 0.025 and p = 0.0003). A better prognosis was associated with TILs intratumoral or mixed localizations (p = 0.029). TILs localization affects NSCLC prognosis; the higher expression of IDO-1 and PD-L1 in poorly differentiated and more aggressive lung adenocarcinomas, as well as the correlation between high PD-L1 expression and squamous cell histotype, confirm the more efficient immunoescaping of these NSCLC subgroups.
Collapse
Affiliation(s)
- Martina Mandarano
- Department of Experimental Medicine, Section of Anatomic Pathology and Histology, Medical School, University of Perugia, Perugia, Italy.
| | - Guido Bellezza
- Department of Experimental Medicine, Section of Anatomic Pathology and Histology, Medical School, University of Perugia, Perugia, Italy
| | | | - Benoit J Van den Eynde
- Ludwig Institute for Cancer Research, de Duve Institute, Université catholique de Louvain, Louvain-la-Neuve, Belgium
| | - Rita Chiari
- Department of Medical Oncology, Santa Maria della Misericordia Hospital, Perugia, Italy
| | - Jacopo Vannucci
- Department of Thoracic Surgery, Medical School, University of Perugia, Perugia, Italy
| | - Giada Mondanelli
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | - Vienna Ludovini
- Department of Medical Oncology, Santa Maria della Misericordia Hospital, Perugia, Italy
| | - Ivana Ferri
- Department of Experimental Medicine, Section of Anatomic Pathology and Histology, Medical School, University of Perugia, Perugia, Italy
| | | | - Rachele Del Sordo
- Department of Experimental Medicine, Section of Anatomic Pathology and Histology, Medical School, University of Perugia, Perugia, Italy
| | - Lucio Cagini
- Department of Thoracic Surgery, Medical School, University of Perugia, Perugia, Italy
| | - Elisa Albini
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | - Giulio Metro
- Department of Medical Oncology, Santa Maria della Misericordia Hospital, Perugia, Italy
| | - Francesco Puma
- Department of Thoracic Surgery, Medical School, University of Perugia, Perugia, Italy
| | - Angelo Sidoni
- Department of Experimental Medicine, Section of Anatomic Pathology and Histology, Medical School, University of Perugia, Perugia, Italy
| |
Collapse
|
38
|
Abstract
Immune therapy to ease the burden of sepsis has thus far failed to consistently improve patient outcomes. Advances in cancer immune therapy and awareness that prolonged immune-suppression in sepsis can leave patients vulnerable to secondary infection and death have driven resurgence in the field of sepsis immune-therapy investigation. As we develop and evaluate these novel therapies, we must learn from past experiences where single-mediator targeted immune therapies were blindly delivered to heterogeneous patient cohorts with complex and evolving immune responses. Advances in genomics, proteomics, metabolomics, and point-of-care technology, coupled with a better understanding of sepsis pathogenesis, have meant that personalised immune-therapy is on the horizon. Here, we review the complex immune pathogenesis in sepsis and the contemporary immune therapies that are being investigated to manipulate this response. An outline of the immune biomarkers that may be used to support this approach is also provided.
Collapse
Affiliation(s)
- Roger Davies
- Department of Anaesthetics, Pain and Intensive Care Medicine, Imperial College London, UK
- Chelsea and Westminster Hospital NHS Foundation Trust, London, UK
| | - Kieran O’Dea
- Department of Anaesthetics, Pain and Intensive Care Medicine, Imperial College London, UK
| | - Anthony Gordon
- Department of Anaesthetics, Pain and Intensive Care Medicine, Imperial College London, UK
- Imperial College Healthcare NHS Trust, London, UK
| |
Collapse
|
39
|
Deniz B, Altincatal A, Ambavane A, Rao S, Doan J, Malcolm B, Michaelson MD, Yang S. Application of dynamic modeling for survival estimation in advanced renal cell carcinoma. PLoS One 2018; 13:e0203406. [PMID: 30161244 PMCID: PMC6117067 DOI: 10.1371/journal.pone.0203406] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Accepted: 08/20/2018] [Indexed: 01/05/2023] Open
Abstract
Objective In oncology, extrapolation of clinical outcomes beyond trial duration is traditionally achieved by parametric survival analysis using population-level outcomes. This approach may not fully capture the benefit/risk profile of immunotherapies due to their unique mechanisms of action. We evaluated an alternative approach—dynamic modeling—to predict outcomes in patients with advanced renal cell carcinoma. We compared standard parametric fitting and dynamic modeling for survival estimation of nivolumab and everolimus using data from the phase III CheckMate 025 study. Methods We developed two statistical approaches to predict longer-term outcomes (progression, treatment discontinuation, and survival) for nivolumab and everolimus, then compared these predictions against follow-up clinical trial data to assess their proximity to observed outcomes. For the parametric survival analyses, we selected a probability distribution based on its fit to observed population-level outcomes at 14-month minimum follow-up and used it to predict longer-term outcomes. For dynamic modeling, we used a multivariate Cox regression based on patient-level data, which included risk scores, and probability and duration of response as predictors of longer-term outcomes. Both sets of predictions were compared against trial data with 26- and 38-month minimum follow-up. Results Both statistical approaches led to comparable fits to observed trial data for median progression, discontinuation, and survival. However, beyond the trial duration, mean survival predictions differed substantially between methods for nivolumab (30.8 and 51.5 months), but not everolimus (27.2 and 29.8 months). Longer-term follow-up data from CheckMate 025 and phase I/II studies resembled dynamic model predictions for nivolumab. Conclusions Dynamic modeling can be a good alternative to parametric survival fitting for immunotherapies because it may help better capture the longer-term benefit/risk profile and support health-economic evaluations of immunotherapies.
Collapse
Affiliation(s)
- Baris Deniz
- Evidera Inc., Bethesda, Maryland, United States of America
- * E-mail:
| | | | | | - Sumati Rao
- Bristol-Myers Squibb, Princeton, New Jersey, United States of America
| | - Justin Doan
- Bristol-Myers Squibb, Princeton, New Jersey, United States of America
| | - Bill Malcolm
- Bristol-Myers Squibb, Princeton, New Jersey, United States of America
| | - M. Dror Michaelson
- Massachusetts General Hospital, Boston, Massachusetts, United States of America
| | - Shuo Yang
- Bristol-Myers Squibb, Princeton, New Jersey, United States of America
| |
Collapse
|
40
|
Danova M, Torchio M, Comolli G, Sbrana A, Antonuzzo A, Mazzini G. The role of automated cytometry in the new era of cancer immunotherapy. Mol Clin Oncol 2018; 9:355-361. [PMID: 30233791 DOI: 10.3892/mco.2018.1701] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Accepted: 08/09/2018] [Indexed: 12/11/2022] Open
Abstract
The introduction in the clinical practice of several new approaches to cancer immunotherapy has greatly increased the interest in analytical methodologies that can define the immunological profile of patients in the clinical setting. This requires huge effort to obtain reliable monitoring tools that could be used to improve the patient's clinical outcome. The clinical applications of flow cytometry (FCM) in oncology started with the measurement of DNA content for the evaluation of both ploidy and cell cycle profile as potential prognostic parameters in the majority of human solid cancer types. The availability of monoclonal antibodies widely broadened the spectrum of clinical applications of this technique, which rapidly became a fundamental tool for the diagnosis and prognosis of malignant hematological diseases. Among the emerging clinical applications of FCM, the study of minimal residual disease in hematological malignancies, the quantification of blood dendritic cells in various types of tumors, the study of metastatic spread in solid tumors throughout both the analysis of circulating endothelial progenitor cells and the identification and characterization of circulating tumor cells, all appear very promising. More recently, an advanced single cell analysis technique has been developed that combines the precision of mass spectrometry with the unique advantages of FCM. This approach, termed mass cytometry, utilizes antibodies conjugated to heavy metal ions for the analysis of cellular proteins by a mass spectrometer. It provides measurement of over 100 simultaneous cellular parameters in a single sample and has evolved from a promising technology to a high recognized platform for multi-dimensional single-cell analysis. Should a careful standardization of the analytical procedures be reached, both FCM and mass cytometry could effectively become ideal tools for the optimization of new immunotherapeutic approaches in cancer patients.
Collapse
Affiliation(s)
- Marco Danova
- Department of Internal Medicine and Medical Oncology, Vigevano Civic Hospital, ASST of Pavia, I-27029 Vigevano, Italy
| | - Martina Torchio
- Department of Internal Medicine and Medical Oncology, Vigevano Civic Hospital, ASST of Pavia, I-27029 Vigevano, Italy
| | - Giuditta Comolli
- Department of Microbiology and Virology and Biotechnology Laboratories, IRCCS San Matteo Foundation, I-27100 Pavia, Italy
| | - Andrea Sbrana
- Department of Medical Oncology 2, University Hospital of Pisa, I-56126 Pisa, Italy
| | - Andrea Antonuzzo
- Department of Medical Oncology 2, University Hospital of Pisa, I-56126 Pisa, Italy
| | | |
Collapse
|
41
|
CORR Insights®: Intratibial Injection Causes Direct Pulmonary Seeding of Osteosarcoma Cells and Is Not a Spontaneous Model Metastasis: A Mouse Osteosarcoma Model. Clin Orthop Relat Res 2018; 476:1523-1525. [PMID: 29794865 PMCID: PMC6437585 DOI: 10.1097/corr.0000000000000356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
|
42
|
Ji N, Zhang Y, Liu Y, Xie J, Wang Y, Hao S, Gao Z. Heat shock protein peptide complex-96 vaccination for newly diagnosed glioblastoma: a phase I, single-arm trial. JCI Insight 2018; 3:99145. [PMID: 29769450 DOI: 10.1172/jci.insight.99145] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Accepted: 04/12/2018] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Heat shock protein peptide complex-96 (HSPPC-96) triggers adaptive and innate antitumor immune responses. The safety and efficacy of HSPPC-96 vaccination was examined in patients with newly diagnosed glioblastoma multiforme (GBM). METHODS In this open-label, single-arm, phase I study, adult patients were vaccinated with HSPPC-96 in combination with the standard treatment for newly diagnosed GBM after surgical resection. Primary endpoints were frequency of adverse events and progression-free survival (PFS) at 6 months. Secondary endpoints included overall survival (OS), PFS, and tumor-specific immune response (TSIR). RESULTS A total of 20 patients with newly diagnosed GBM were enrolled from September 2013 to February 2015. No grade 3 or 4 vaccine-related adverse events were noted. After a median follow-up of 42.3 months, PFS was 89.5% (95% CI, 66.9%-98.7%) at 6 months, median PFS was 11.0 months (95% CI, 8.2-13.8), and median OS was 31.4 months (95% CI, 14.9-47.9). TSIR was significantly increased by 2.3-fold (95% CI, 1.7-3.2) after vaccination. Median OS for patients with high TSIR after vaccination was >40.5 months (95% CI, incalculable) as compared with 14.6 months (95% CI, 7.0-22.2) for patients with low TSIR after vaccination (hazard ratio, 0.25; 95% CI, 0.071-0.90; P = 0.034). A multivariate Cox regression model revealed TSIR after vaccination as a primary independent predicator for survival. CONCLUSION The HSPPC-96 vaccination, combined with the standard therapy, is a safe and effective strategy for treatment of newly diagnosed GBM patients. TSIR after vaccination would be a good indicator predicting the vaccine efficacy. TRIAL REGISTRATION ClinicalTrials.gov NCT02122822. FUNDING National Key Technology Research and Development Program of the Ministry of Science and Technology of China (2014BAI04B01, 2014BAI04B02), Beijing Natural Science Foundation (7164253), Beijing Talents Fund (2014000021469G257), and Shenzhen Science and Technology Innovation Committee (JSGG20170413151359491).
Collapse
|
43
|
Wang M, Bu J, Zhou M, Sido J, Lin Y, Liu G, Lin Q, Xu X, Leavenworth JW, Shen E. CD8 +T cells expressing both PD-1 and TIGIT but not CD226 are dysfunctional in acute myeloid leukemia (AML) patients. Clin Immunol 2018; 190:64-73. [PMID: 28893624 DOI: 10.1016/j.clim.2017.08.021] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2017] [Revised: 08/16/2017] [Accepted: 08/24/2017] [Indexed: 01/07/2023]
Abstract
Acute myeloid leukemia (AML) is one of the most common types of leukemia among adults with an overall poor prognosis and very limited treatment management. Immune checkpoint blockade of PD-1 alone or combined with other immune checkpoint blockade has gained impressive results in murine AML models by improving anti-leukemia CD8+T cell function, which has greatly promoted the strategy to utilize combined immune checkpoint inhibitors to treat AML patients. However, the expression profiles of these immune checkpoint receptors, such as co-inhibitory receptors PD-1 and TIGIT and co-stimulatory receptor CD226, in T cells from AML patients have not been clearly defined. Here we have defined subsets of CD8+ and CD4+ T cells in the peripheral blood (PB) from newly diagnosed AML patients and healthy controls (HCs). We have observed increased frequencies of PD-1- and TIGIT- expressing CD8+ T cells but decreased occurrence of CD226-expressing CD8+T cells in AML patients. Further analysis of these CD8+ T cells revealed a unique CD8+ T cell subset that expressed PD-1 and TIGIT but displayed lower levels of CD226 was associated with failure to achieve remission after induction chemotherapy and FLT3-ITD mutations which predict poor clinical prognosis in AML patients. Importantly, these PD-1+TIGIT+CD226-CD8+T cells are dysfunctional with lower expression of intracellular IFN-γ and TNF-α than their counterparts in HCs. Therefore, our studies revealed that an increased frequency of a unique CD8+ T cell subset, PD-1+TIGIT+CD226-CD8+T cells, is associated with CD8+T cell dysfunction and poor clinical prognosis of AML patients, which may reveal critical diagnostic or prognostic biomarkers and direct more efficient therapeutic strategies.
Collapse
Affiliation(s)
- Mengjie Wang
- Department of Pathogenic Biology and Immunology, Guangzhou Hoffmann Institute of Immunology, School of Basic Sciences, Guangzhou Medical University, Guangzhou 510182, China
| | - Jin Bu
- Editorial Department of Journals of Nanjing Medical University, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Maohua Zhou
- Department of Laboratory Medicine, Guangdong General Hospital, Academy of Medical Sciences, Guangzhou 510080, China
| | - Jessica Sido
- Department of Cancer Immunology and Virology, Dana Farber Cancer Institute, Boston, MA 02115, USA; Department of Microbiology & Immunobiology, Division of Immunology, Harvard Medical School, Boston, MA 02115, USA
| | - Yu Lin
- Shenzhen Withsum Technology Limited, Shenzhen 518031, China
| | - Guanfang Liu
- Department of Pathogenic Biology and Immunology, Guangzhou Hoffmann Institute of Immunology, School of Basic Sciences, Guangzhou Medical University, Guangzhou 510182, China
| | - Qiwen Lin
- Guangzhou Blood Center, Guangzhou 510095, China
| | - Xiuzhang Xu
- Guangzhou Blood Center, Guangzhou 510095, China
| | - Jianmei W Leavenworth
- Department of Neurosurgery, University of Alabama at Birmingham, Birmingham, AL 35233, USA; Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL 35233, USA.
| | - Erxia Shen
- Department of Pathogenic Biology and Immunology, Guangzhou Hoffmann Institute of Immunology, School of Basic Sciences, Guangzhou Medical University, Guangzhou 510182, China; Department of Cancer Immunology and Virology, Dana Farber Cancer Institute, Boston, MA 02115, USA.
| |
Collapse
|
44
|
Guerriero JL. Macrophages: The Road Less Traveled, Changing Anticancer Therapy. Trends Mol Med 2018; 24:472-489. [PMID: 29655673 PMCID: PMC5927840 DOI: 10.1016/j.molmed.2018.03.006] [Citation(s) in RCA: 213] [Impact Index Per Article: 30.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2018] [Revised: 03/04/2018] [Accepted: 03/12/2018] [Indexed: 12/13/2022]
Abstract
Macrophages are present in all vertebrate tissues and have emerged as multifarious cells with complex roles in development, tissue homeostasis, and disease. Macrophages are a major constituent of the tumor microenvironment, where they either promote or inhibit tumorigenesis and metastasis depending on their state. Successful preclinical strategies to target macrophages for anticancer therapy are now being evaluated in the clinic and provide proof of concept that targeting macrophages may enhance current therapies; however, clinical success has been limited. This review discusses the promise of targeting macrophages for anticancer therapy, yet highlights how much is unknown regarding their ontogeny, regulation, and tissue-specific diversity. Further work might identify subsets of macrophages within different tissues, which could reveal novel therapeutic opportunities for anticancer therapy.
Collapse
Affiliation(s)
- Jennifer L Guerriero
- Department of Medical Oncology, Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, MA 02215, USA.
| |
Collapse
|
45
|
Immunotherapies: Exploiting the Immune System for Cancer Treatment. J Immunol Res 2018; 2018:9585614. [PMID: 29725606 PMCID: PMC5872614 DOI: 10.1155/2018/9585614] [Citation(s) in RCA: 88] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2017] [Revised: 12/21/2017] [Accepted: 01/11/2018] [Indexed: 12/31/2022] Open
Abstract
Cancer is a condition that has plagued humanity for thousands of years, with the first depictions dating back to ancient Egyptian times. However, not until recent decades have biological therapeutics been developed and refined enough to safely and effectively combat cancer. Three unique immunotherapies have gained traction in recent decades: adoptive T cell transfer, checkpoint inhibitors, and bivalent antibodies. Each has led to clinically approved therapies, as well as to therapies in preclinical and ongoing clinical trials. In this review, we outline the method by which these 3 immunotherapies function as well as any major immunotherapeutic drugs developed for treating a variety of cancers.
Collapse
|
46
|
Alguacil-Núñez C, Ferrer-Ortiz I, García-Verdú E, López-Pirez P, Llorente-Cortijo IM, Sainz B. Current perspectives on the crosstalk between lung cancer stem cells and cancer-associated fibroblasts. Crit Rev Oncol Hematol 2018; 125:102-110. [PMID: 29650269 DOI: 10.1016/j.critrevonc.2018.02.015] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2017] [Revised: 02/17/2018] [Accepted: 02/26/2018] [Indexed: 12/16/2022] Open
Abstract
Lung cancer, in particular non-small cell lung carcinoma (NSCLC), is the second most common cancer in both men and women and the leading cause of cancer-related deaths worldwide. Its prognosis and diagnosis are determined by several driver mutations and diverse risk factors (e.g. smoking). While immunotherapy has proven effective in some patients, treatment of NSCLC using conventional chemotherapy is largely ineffective. The latter is believed to be due to the existence of a subpopulation of stem-like, highly tumorigenic and chemoresistant cells within the tumor population known as cancer stem cells (CSC). To complicate the situation, CSCs interact with the tumor microenvironment, which include cancer-associated fibroblasts (CAFs), immune cells, endothelial cells, growth factors, cytokines and connective tissue components, which via a dynamic crosstalk, composed of proteins and exosomes, activates the CSC compartment. In this review, we analyze the crosstalk between CSCs and CAFs, the primary component of the NSCLC microenvironment, at the molecular and extracellular level and contemplate therapies to disrupt this communication.
Collapse
Affiliation(s)
- Cristina Alguacil-Núñez
- Department of Biochemistry, Cancer Stem Cell and Tumor Microenvironment Group, Universidad Autónoma de Madrid (UAM), Madrid, Spain
| | - Inés Ferrer-Ortiz
- Department of Biochemistry, Cancer Stem Cell and Tumor Microenvironment Group, Universidad Autónoma de Madrid (UAM), Madrid, Spain
| | - Elena García-Verdú
- Department of Biochemistry, Cancer Stem Cell and Tumor Microenvironment Group, Universidad Autónoma de Madrid (UAM), Madrid, Spain
| | - Pilar López-Pirez
- Department of Biochemistry, Cancer Stem Cell and Tumor Microenvironment Group, Universidad Autónoma de Madrid (UAM), Madrid, Spain
| | - Irene Maria Llorente-Cortijo
- Department of Biochemistry, Cancer Stem Cell and Tumor Microenvironment Group, Universidad Autónoma de Madrid (UAM), Madrid, Spain
| | - Bruno Sainz
- Department of Biochemistry, Cancer Stem Cell and Tumor Microenvironment Group, Universidad Autónoma de Madrid (UAM), Madrid, Spain; Department of Cancer Biology, Instituto de Investigaciones Biomédicas "Alberto Sols" (IIBM), CSIC-UAM, Madrid, Spain; Chronic Diseases and Cancer Area 3 - Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain.
| |
Collapse
|
47
|
Microorganisms in the Treatment of Cancer: Advantages and Limitations. J Immunol Res 2018; 2018:2397808. [PMID: 29682586 PMCID: PMC5848056 DOI: 10.1155/2018/2397808] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Accepted: 01/09/2018] [Indexed: 12/22/2022] Open
Abstract
Cancer remains one of the major challenges of the 21st century. The increasing numbers of cases are not accompanied by adequate progress in therapy. The standard methods of treatment often do not lead to the expected effects. Therefore, it is extremely important to find new, more effective treatments. One of the most promising research directions is immunotherapy, including the use of specific types of microorganisms. This type of treatment is expected to stimulate the immune system for the selective elimination of cancer cells. The research results seem to be promising and show the intensive activation of the immune response as a result of bacterial stimulation. In addition, it is possible to use microorganisms in many different ways, based on their specific properties, that is, toxin production, anaerobic lifestyle, or binding substances that can be delivered to a specific location (vectors). This paper provides an overview of selected microorganisms which are already in use or that are in the experimental phase. Just like any other therapy, the use of microbes for cancer treatment also has some disadvantages. Nevertheless, this kind of treatment can supplement conventional anticancer therapy, giving cancer patients a chance and hope of recovery.
Collapse
|
48
|
Golan T, Milella M, Ackerstein A, Berger R. The changing face of clinical trials in the personalized medicine and immuno-oncology era: report from the international congress on clinical trials in Oncology & Hemato-Oncology (ICTO 2017). JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2017; 36:192. [PMID: 29282151 PMCID: PMC5745625 DOI: 10.1186/s13046-017-0668-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 12/16/2017] [Accepted: 12/18/2017] [Indexed: 01/10/2023]
Abstract
In the past decade, the oncology community has witnessed major advances in the understanding of cancer biology and major breakthroughs in several different therapeutic areas, from solid tumors to hematological malignancies; moreover, the advent of effective immunotherapy approaches, such as immune-checkpoint blockade, is revolutionizing treatment algorithms in almost all oncology disease areas. As knowledge evolves and new weapons emerge in the “war against cancer”, clinical and translational research need to adapt to a rapidly changing environment to effectively translate novel concepts into sustainable and accessible therapeutic options for cancer patients. With this in mind, translational cancer researchers, oncology professionals, treatment experts, CRO and industry leaders, as well as patient representatives gathered in London, 16-17 March 2017, for The International Congress on Clinical Trials in Oncology and Hemato-Oncology (ICTO2017), to discuss the changing face of oncology clinical trials in the new era of personalized medicine and immuno-oncology. A wide range of topics, including clinical trial design in immuno-oncology, biomarker-oriented drug development paths, statistical design and endpoint selection, challenges in the design and conduct of personalized medicine clinical trials, risk-based monitoring, financing and reimbursement, as well as best operational practices, were discussed in an open, highly interactive format, favoring networking among all relevant stakeholders. The most relevant data, approaches and issues emerged and discussed during the conference are summarized in this report.
Collapse
Affiliation(s)
- Talia Golan
- Oncology Institute, Sheba Medical Center, Emek HaEla St 1, Tel Hashomer, Ramat Gan, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Michele Milella
- Division of Medical Oncology 1, Regina Elena National Cancer Institute, via Elio Chianesi 53, 00144, Rome, Italy.
| | - Aliza Ackerstein
- Oncology Institute, Sheba Medical Center, Emek HaEla St 1, Tel Hashomer, Ramat Gan, Israel
| | - Ranaan Berger
- Oncology Institute, Sheba Medical Center, Emek HaEla St 1, Tel Hashomer, Ramat Gan, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
49
|
Cruz-Adalia A, Ramirez-Santiago G, Osuna-Pérez J, Torres-Torresano M, Zorita V, Martínez-Riaño A, Boccasavia V, Borroto A, Martínez Del Hoyo G, González-Granado JM, Alarcón B, Sánchez-Madrid F, Veiga E. Conventional CD4 + T cells present bacterial antigens to induce cytotoxic and memory CD8 + T cell responses. Nat Commun 2017; 8:1591. [PMID: 29147022 PMCID: PMC5691066 DOI: 10.1038/s41467-017-01661-7] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Accepted: 10/06/2017] [Indexed: 01/15/2023] Open
Abstract
Bacterial phagocytosis and antigen cross-presentation to activate CD8+ T cells are principal functions of professional antigen presenting cells. However, conventional CD4+ T cells also capture and kill bacteria from infected dendritic cells in a process termed transphagocytosis (also known as transinfection). Here, we show that transphagocytic T cells present bacterial antigens to naive CD8+ T cells, which proliferate and become cytotoxic in response. CD4+ T-cell-mediated antigen presentation also occurs in vivo in the course of infection, and induces the generation of central memory CD8+ T cells with low PD-1 expression. Moreover, transphagocytic CD4+ T cells induce protective anti-tumour immune responses by priming CD8+ T cells, highlighting the potential of CD4+ T cells as a tool for cancer immunotherapy. Antigen presentation is generally considered the domain of innate immune cells, but CD4+ T cells can transphagocytose bacteria from infected dendritic cells. Here the authors show CD4+ T cells can transphagocytose bacterial and tumour antigens and present them to CD8+ T cells to activate memory and cytotoxic functions.
Collapse
Affiliation(s)
- Aránzazu Cruz-Adalia
- Department of Molecular & Cellular Biology, Centro Nacional de Biotecnología; Consejo Superior de Investigaciones Científicas (CNB-CSIC), Darwin 3, 28049, Madrid, Spain.
| | - Guillermo Ramirez-Santiago
- Department of Molecular & Cellular Biology, Centro Nacional de Biotecnología; Consejo Superior de Investigaciones Científicas (CNB-CSIC), Darwin 3, 28049, Madrid, Spain.,Hospital de Santa Cristina, Instituto de Investigación Sanitaria Princesa, 28009, Madrid, Spain
| | - Jesús Osuna-Pérez
- Department of Molecular & Cellular Biology, Centro Nacional de Biotecnología; Consejo Superior de Investigaciones Científicas (CNB-CSIC), Darwin 3, 28049, Madrid, Spain
| | - Mónica Torres-Torresano
- Department of Molecular & Cellular Biology, Centro Nacional de Biotecnología; Consejo Superior de Investigaciones Científicas (CNB-CSIC), Darwin 3, 28049, Madrid, Spain
| | - Virgina Zorita
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Melchor Fernández Almagro 3, 28029, Madrid, Spain
| | - Ana Martínez-Riaño
- Department of Cell Biology and Immunology, Centro de Biología Molecular Severo Ochoa (CBMSO); Nicolás Cabrera 1, Universidad Autónoma de Madrid, 28049, Madrid, Spain
| | - Viola Boccasavia
- Department of Cell Biology and Immunology, Centro de Biología Molecular Severo Ochoa (CBMSO); Nicolás Cabrera 1, Universidad Autónoma de Madrid, 28049, Madrid, Spain
| | - Aldo Borroto
- Department of Cell Biology and Immunology, Centro de Biología Molecular Severo Ochoa (CBMSO); Nicolás Cabrera 1, Universidad Autónoma de Madrid, 28049, Madrid, Spain
| | - Gloria Martínez Del Hoyo
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Melchor Fernández Almagro 3, 28029, Madrid, Spain
| | - José María González-Granado
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Melchor Fernández Almagro 3, 28029, Madrid, Spain.,Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), 28041, Madrid, Spain
| | - Balbino Alarcón
- Department of Cell Biology and Immunology, Centro de Biología Molecular Severo Ochoa (CBMSO); Nicolás Cabrera 1, Universidad Autónoma de Madrid, 28049, Madrid, Spain
| | | | - Esteban Veiga
- Department of Molecular & Cellular Biology, Centro Nacional de Biotecnología; Consejo Superior de Investigaciones Científicas (CNB-CSIC), Darwin 3, 28049, Madrid, Spain.
| |
Collapse
|
50
|
Wang K, Shan S, Yang Z, Gu X, Wang Y, Wang C, Ren T. IL-33 blockade suppresses tumor growth of human lung cancer through direct and indirect pathways in a preclinical model. Oncotarget 2017; 8:68571-68582. [PMID: 28978138 PMCID: PMC5620278 DOI: 10.18632/oncotarget.19786] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2017] [Accepted: 06/18/2017] [Indexed: 02/07/2023] Open
Abstract
Non-small-cell lung cancer (NSCLC) is the most common type in lung cancer, a leading cause of cancer-related death worldwide. Our previous study unraveled a pro-cancer function of IL-33 in fueling outgrowth and metastasis of human NSCLC cells. Herein, we determined that interfere with IL-33 activity was an effective strategy for limiting NSCLC tumor growth using a preclinical model with human NSCLC xenografts. IL-33 blockade efficiently inhibited tumor growth of NSCLC xenografts in immune-deficient mice. Mechanistically, IL-33 blockade suppressed outgrowth capacity of human NSCLC cells. Meanwhile, IL-33 blockade abrogated polarization of M2 tumor-associated macrophages (TAMs) and reduced accumulation of regulatory T cells (Tregs) in tumor microenvironments, shaping functional immune surveillance. In NSCLC patients, IL-33 expressions were positively correlated with Ki-67 proliferation index and expressions of M2 TAM- and Teg-related genes. These findings identify IL-33 as a dual-functional factor in NSCLC pathogenesis and suggest IL-33 blockade as a promising therapeutic for NSCLC patients.
Collapse
Affiliation(s)
- Kailing Wang
- Department of Respiratory Medicine, East Hospital, Tongji University School of Medicine, Shanghai 200120, China
| | - Shan Shan
- Department of Respiratory Medicine, East Hospital, Tongji University School of Medicine, Shanghai 200120, China.,Department of Respiratory Medicine, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China
| | - Zongjun Yang
- Department of Clinical Laboratory, Qingdao Women & Children Hospital, Qingdao 266034, China
| | - Xia Gu
- Department of Respiratory Medicine, East Hospital, Tongji University School of Medicine, Shanghai 200120, China
| | - Yuanyuan Wang
- Department of Respiratory Medicine, East Hospital, Tongji University School of Medicine, Shanghai 200120, China
| | - Chunhong Wang
- Department of Respiratory Medicine, East Hospital, Tongji University School of Medicine, Shanghai 200120, China
| | - Tao Ren
- Department of Respiratory Medicine, East Hospital, Tongji University School of Medicine, Shanghai 200120, China.,Department of Respiratory Medicine, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China
| |
Collapse
|