1
|
Poudel K, Vithiananthan T, Kim JO, Tsao H. Recent progress in cancer vaccines and nanovaccines. Biomaterials 2025; 314:122856. [PMID: 39366184 DOI: 10.1016/j.biomaterials.2024.122856] [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: 04/25/2024] [Revised: 09/03/2024] [Accepted: 09/26/2024] [Indexed: 10/06/2024]
Abstract
Vaccine science, nanotechnology, and immunotherapy are at the forefront of cancer treatment strategies, each offering significant potential for enhancing tumor-specific immunity and establishing long-lasting immune memory to prevent tumor recurrence. Despite the promise of these personalized and precision-based anti-cancer approaches, challenges such as immunosuppression, suboptimal immune activation, and T-cell exhaustion continue to hinder their effectiveness. The limited clinical success of cancer vaccines often stems from difficulties in identifying effective antigens, efficiently targeting immune cells, lymphoid organs, and the tumor microenvironment, overcoming immune evasion, enhancing immunogenicity, and avoiding lysosomal degradation. However, numerous studies have demonstrated that integrating nanotechnology with immunotherapeutic strategies in vaccine development can overcome these challenges, leading to potent antitumor immune responses and significant progress in the field. This review highlights the critical components of cancer vaccine and nanovaccine strategies for immunomodulatory antitumor therapy. It covers general vaccine strategies, types of vaccines, antigen forms, nanovaccine platforms, challenges faced, potential solutions, and key findings from preclinical and clinical studies, along with future perspectives. To fully unlock the potential of cancer vaccines and nanovaccines, precise immunological monitoring during early-phase trials is essential. This approach will help identify and address obstacles, ultimately expanding the available options for patients who are resistant to conventional cancer immunotherapies.
Collapse
Affiliation(s)
- Kishwor Poudel
- Wellman Center for Photomedicine and Department of Dermatology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Tulasi Vithiananthan
- Wellman Center for Photomedicine and Department of Dermatology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Jong Oh Kim
- College of Pharmacy, Yeungnam University, Gyeongsan, 38541, Republic of Korea
| | - Hensin Tsao
- Wellman Center for Photomedicine and Department of Dermatology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
2
|
Karimi-Sani I, Molavi Z, Naderi S, Mirmajidi SH, Zare I, Naeimzadeh Y, Mansouri A, Tajbakhsh A, Savardashtaki A, Sahebkar A. Personalized mRNA vaccines in glioblastoma therapy: from rational design to clinical trials. J Nanobiotechnology 2024; 22:601. [PMID: 39367418 PMCID: PMC11453023 DOI: 10.1186/s12951-024-02882-x] [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: 06/03/2024] [Accepted: 09/26/2024] [Indexed: 10/06/2024] Open
Abstract
Glioblastomas (GBMs) are the most common and aggressive malignant brain tumors, presenting significant challenges for treatment due to their invasive nature and localization in critical brain regions. Standard treatment includes surgical resection followed by radiation and adjuvant chemotherapy with temozolomide (TMZ). Recent advances in immunotherapy, including the use of mRNA vaccines, offer promising alternatives. This review focuses on the emerging use of mRNA vaccines for GBM treatment. We summarize recent advancements, evaluate current obstacles, and discuss notable successes in this field. Our analysis highlights that while mRNA vaccines have shown potential, their use in GBM treatment is still experimental. Ongoing research and clinical trials are essential to fully understand their therapeutic potential. Future developments in mRNA vaccine technology and insights into GBM-specific immune responses may lead to more targeted and effective treatments. Despite the promise, further research is crucial to validate and optimize the effectiveness of mRNA vaccines in combating GBM.
Collapse
Affiliation(s)
- Iman Karimi-Sani
- Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Zahra Molavi
- Proteomics Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Samaneh Naderi
- Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Seyedeh-Habibeh Mirmajidi
- Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Iman Zare
- Research and Development Department, Sina Medical Biochemistry Technologies Co. Ltd., Shiraz, 7178795844, Iran
| | - Yasaman Naeimzadeh
- Department of Molecular Medicine, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Atena Mansouri
- Cellular and Molecular Research Center, Birjand University of Medical Sciences, Birjand, Iran
| | - Amir Tajbakhsh
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Amir Savardashtaki
- Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran.
- Infertility Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
- Department of Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
3
|
Gurunathan S, Thangaraj P, Wang L, Cao Q, Kim JH. Nanovaccines: An effective therapeutic approach for cancer therapy. Biomed Pharmacother 2024; 170:115992. [PMID: 38070247 DOI: 10.1016/j.biopha.2023.115992] [Citation(s) in RCA: 23] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2023] [Revised: 11/23/2023] [Accepted: 12/06/2023] [Indexed: 01/10/2024] Open
Abstract
Cancer vaccines hold considerable promise for the immunotherapy of solid tumors. Nanomedicine offers several strategies for enhancing vaccine effectiveness. In particular, molecular or (sub) cellular vaccines can be delivered to the target lymphoid tissues and cells by nanocarriers and nanoplatforms to increase the potency and durability of antitumor immunity and minimize negative side effects. Nanovaccines use nanoparticles (NPs) as carriers and/or adjuvants, offering the advantages of optimal nanoscale size, high stability, ample antigen loading, high immunogenicity, tunable antigen presentation, increased retention in lymph nodes, and immunity promotion. To induce antitumor immunity, cancer vaccines rely on tumor antigens, which are administered in the form of entire cells, peptides, nucleic acids, extracellular vesicles (EVs), or cell membrane-encapsulated NPs. Ideal cancer vaccines stimulate both humoral and cellular immunity while overcoming tumor-induced immune suppression. Herein, we review the key properties of nanovaccines for cancer immunotherapy and highlight the recent advances in their development based on the structure and composition of various (including synthetic and semi (biogenic) nanocarriers. Moreover, we discuss tumor cell-derived vaccines (including those based on whole-tumor-cell components, EVs, cell membrane-encapsulated NPs, and hybrid membrane-coated NPs), nanovaccine action mechanisms, and the challenges of immunocancer therapy and their translation to clinical applications.
Collapse
Affiliation(s)
- Sangiliyandi Gurunathan
- Department of Biotechnology, Rathinam College of Arts and Science, Eachanari, Coimbatore 641 021, Tamil Nadu, India.
| | - Pratheep Thangaraj
- Department of Biotechnology, Rathinam College of Arts and Science, Eachanari, Coimbatore 641 021, Tamil Nadu, India
| | - Lin Wang
- Research and Development Department, Qingdao Haier Biotech Co., Ltd., Qingdao, China
| | - Qilong Cao
- Research and Development Department, Qingdao Haier Biotech Co., Ltd., Qingdao, China
| | - Jin-Hoi Kim
- Department of Stem Cell and Regenerative Biotechnology, Konkuk University, Seoul 05029, Republic of Korea.
| |
Collapse
|
4
|
Li DD, Tang YL, Wang X. Challenges and exploration for immunotherapies targeting cold colorectal cancer. World J Gastrointest Oncol 2023; 15:55-68. [PMID: 36684057 PMCID: PMC9850757 DOI: 10.4251/wjgo.v15.i1.55] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 10/28/2022] [Accepted: 12/07/2022] [Indexed: 01/10/2023] Open
Abstract
In recent years, immune checkpoint inhibitors (ICIs) have made significant breakthroughs in the treatment of various tumors, greatly improving clinical efficacy. As the fifth most common antitumor treatment strategy for patients with solid tumors after surgery, chemotherapy, radiotherapy and targeted therapy, the therapeutic response to ICIs largely depends on the number and spatial distribution of effector T cells that can effectively identify and kill tumor cells, features that are also important when distinguishing malignant tumors from “cold tumors” or “hot tumors”. At present, only a small proportion of colorectal cancer (CRC) patients with deficient mismatch repair (dMMR) or who are microsatellite instability-high (MSI-H) can benefit from ICI treatments because these patients have the characteristics of a “hot tumor”, with a high tumor mutational burden (TMB) and massive immune cell infiltration, making the tumor more easily recognized by the immune system. In contrast, a majority of CRC patients with proficient MMR (pMMR) or who are microsatellite stable (MSS) have a low TMB, lack immune cell infiltration, and have almost no response to immune monotherapy; thus, these tumors are “cold”. The greatest challenge today is how to improve the immunotherapy response of “cold tumor” patients. With the development of clinical research, immunotherapies combined with other treatment strategies (such as targeted therapy, chemotherapy, and radiotherapy) have now become potentially effective clinical strategies and research hotspots. Therefore, the question of how to promote the transformation of “cold tumors” to “hot tumors” and break through the bottleneck of immunotherapy for cold tumors in CRC patients urgently requires consideration. Only by developing an in-depth understanding of the immunotherapy mechanisms of cold CRCs can we screen out the immunotherapy-dominant groups and explore the most suitable treatment options for individuals to improve therapeutic efficacy.
Collapse
Affiliation(s)
- Dan-Dan Li
- Department of Abdominal Oncology/Radiation Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Yuan-Ling Tang
- Department of Abdominal Oncology/Radiation Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Xin Wang
- Department of Abdominal Oncology/Radiation Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, Sichuan Province, China
| |
Collapse
|
5
|
Blazeska N, Kosaloglu-Yalcin Z, Vita R, Peters B, Sette A. IEDB and CEDAR: Two Sibling Databases to Serve the Global Scientific Community. Methods Mol Biol 2023; 2673:133-149. [PMID: 37258911 PMCID: PMC11008223 DOI: 10.1007/978-1-0716-3239-0_9] [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] [Indexed: 06/02/2023]
Abstract
Various methodologies have been utilized to analyze epitope-specific responses in the context of non-self-antigens, such as those associated with infectious diseases and allergies, and in the context of self-antigens, such as those associated with transplantation, autoimmunity, and cancer. Further to this, epitope-specific data, and its associated immunological context, are crucial to training and developing predictive algorithms and pipelines for the development of specific vaccines and diagnostics. In this chapter, we describe the methodology utilized to derive two sibling resources, the Immune Epitope Database (IEDB) and Cancer Epitope Database and Analysis Resource (CEDAR), to specifically host this data, and make them freely available to the scientific community.
Collapse
Affiliation(s)
- Nina Blazeska
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Zeynep Kosaloglu-Yalcin
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Randi Vita
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Bjoern Peters
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA, USA
- Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Alessandro Sette
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA, USA.
- Department of Medicine, University of California San Diego, La Jolla, CA, USA.
| |
Collapse
|
6
|
Sharma G, Pothuraju R, Kanchan RK, Batra SK, Siddiqui JA. Chemokines network in bone metastasis: Vital regulators of seeding and soiling. Semin Cancer Biol 2022; 86:457-472. [PMID: 35124194 PMCID: PMC9744380 DOI: 10.1016/j.semcancer.2022.02.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 01/20/2022] [Accepted: 02/01/2022] [Indexed: 02/07/2023]
Abstract
Chemokines are well equipped with chemo-attractive signals that can regulate cancer cell trafficking to specific organ sites. Currently, updated concepts have revealed the diverse role of chemokines in the biology of cancer initiation and progression. Genomic instabilities and alterations drive tumor heterogeneity, providing more options for the selection and metastatic progression to cancer cells. Tumor heterogeneity and acquired drug resistance are the main obstacles in managing cancer therapy and the primary root cause of metastasis. Studies emphasize that multiple chemokine/receptor axis are involved in cancer cell-mediated organ-specific distant metastasis. One of the persuasive mechanisms for heterogeneity and subsequent events is sturdily interlinked with the crosstalk between chemokines and their receptors on cancer cells and tissue-specific microenvironment. Among different metastatic niches, skeletal metastasis is frequently observed in the late stages of prostate, breast, and lung cancer and significantly reduces the survival of cancer patients. Therefore, it is crucial to elucidate the role of chemokines and their receptors in metastasis and bone remodeling. Here, we review the potential chemokine/receptor axis in tumorigenesis, tumor heterogeneity, metastasis, and vicious cycle in bone microenvironment.
Collapse
Affiliation(s)
- Gunjan Sharma
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Ramesh Pothuraju
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Ranjana Kumari Kanchan
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Surinder Kumar Batra
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, 68198, USA; Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, 68198, USA; Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Jawed Akhtar Siddiqui
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, 68198, USA; Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, 68198, USA.
| |
Collapse
|
7
|
Ghasemii K, Darroudi M, Rahimmanesh I, Ghomi M, Hassanpour M, Sharifi E, Yousefiasl S, Ahmadi S, Zarrabi A, Borzacchiello A, Rabiee M, Paiva-Santos AC, Rabiee N. Advances in aptamer-based drug delivery vehicles for cancer therapy. BIOMATERIALS ADVANCES 2022; 140:213077. [PMID: 35952549 DOI: 10.1016/j.bioadv.2022.213077] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/28/2022] [Revised: 08/01/2022] [Accepted: 08/04/2022] [Indexed: 06/15/2023]
Abstract
Overall, aptamers are special classes of nucleic acid-based macromolecules that are beginning to investigate because of their capability of avidity binding to a specific target for clinical use. Taking advantage of target-specific medicine led to more effective therapeutic and limitation of side effects of drugs. Herein, we discuss several aptamers and their binding capability and capacity for selecting tumor biomarkers and usage of them as targeting ligands for the functionalization of nanomaterials. We review recent applications based on aptamers and several nanoparticles to rise efficacy and develop carrier systems such as graphene oxide, folic acid, gold, mesopores silica, and various polymers and copolymer, polyethylene glycol, cyclodextrin, chitosan. The nanocarriers have been characterized by particle size, zeta potential, aptamer conjugation, and drug encapsulation efficiency. Hydrodynamic diameter and Zeta potential can used in order to monitor aptamers' crosslinking, in-vitro drug release, intracellular delivery of nanocarriers, and cellular cytotoxicity assay. Also, they are studied for cellular uptake and internalization to types of cancer cell lines such as colorectal, breast, prostate, leukemia and etc. The results are investigated in in-vivo cytotoxicity assay and cell viability assay. Targeted cancer therapy seems a good and promising strategy to overcome the systemic toxicity of chemotherapy.
Collapse
Affiliation(s)
- Kousar Ghasemii
- Department of Organic Chemistry, Faculty of Chemistry, University of Mazandaran, Babolsar, Iran
| | - Mahdieh Darroudi
- Department of Medical Biotechnology and Nanotechnology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Ilnaz Rahimmanesh
- Applied Physiology Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan 8174673461, Iran
| | - Matineh Ghomi
- School of Chemistry, Damghan University, Damghan 36716-41167, Iran
| | - Mahnaz Hassanpour
- Department of Chemistry, Institute for Advanced Studies in Basic Sciences (IASBS), Zanjan 45137-66731, Iran
| | - Esmaeel Sharifi
- Institute for Polymers, Composites and Biomaterials, National Research Council (IPCB-CNR), Naples 80125, Italy; Department of Tissue Engineering and Biomaterials, School of Advanced Medical Sciences and Technologies, Hamadan University of Medical Sciences, 6517838736 Hamadan, Iran
| | - Satar Yousefiasl
- Department of Tissue Engineering and Biomaterials, School of Advanced Medical Sciences and Technologies, Hamadan University of Medical Sciences, 6517838736 Hamadan, Iran
| | - Sepideh Ahmadi
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran 19857-17443, Iran; Cellular and Molecular Biology Research Center, Shahid Beheshti University of Medical Sciences, Tehran 19857-17443, Iran
| | - Ali Zarrabi
- Department of Biomedical Engineering, Faculty of Engineering & Natural Science, Istinye University, Sariyer 34396, Istanbul, Turkey
| | - Assunta Borzacchiello
- Institute for Polymers, Composites and Biomaterials, National Research Council, IPCB-CNR, 80125 Naples, Italy
| | - Mohammad Rabiee
- Biomaterial group, Department of Biomedical Engineering, Amirkabir University of Technology, Tehran, Iran
| | - Ana Cláudia Paiva-Santos
- Department of Pharmaceutical Technology, Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal; REQUIMTE/LAQV, Group of Pharmaceutical Technology, Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal.
| | - Navid Rabiee
- Department of Materials Science and Engineering, Pohang University of Science and Technology (POSTECH), 77 Cheongam-ro, Nam-gu, Pohang, Gyeongbuk 37673, South Korea; School of Engineering, Macquarie University, Sydney, New South Wales 2109, Australia.
| |
Collapse
|
8
|
Fang X, Lan H, Jin K, Gong D, Qian J. Nanovaccines for Cancer Prevention and Immunotherapy: An Update Review. Cancers (Basel) 2022; 14:3842. [PMID: 36010836 PMCID: PMC9405528 DOI: 10.3390/cancers14163842] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2022] [Revised: 07/21/2022] [Accepted: 07/24/2022] [Indexed: 11/23/2022] Open
Abstract
Cancer immunotherapy has received more and more attention from cancer researchers over the past few decades. Various methods such as cell therapy, immune checkpoint blockers, and cancer vaccines alone or in combination therapies have achieved relatively satisfactory results in cancer therapy. Among these immunotherapy-based methods, cancer vaccines alone have not yet had the necessary efficacy in the clinic. Therefore, nanomaterials have increased the efficacy and ef-fectiveness of cancer vaccines by increasing their half-life and durability, promoting tumor mi-croenvironment (TME) reprogramming, and enhancing their anti-tumor immunity with minimal toxicity. In this review, according to the latest studies, the structure and different types of nanovaccines, the mechanisms of these vaccines in cancer treatment, as well as the advantages and disadvantages of these nanovaccines are discussed.
Collapse
Affiliation(s)
- Xingliang Fang
- Department of Hepatobiliary Surgery, Affiliated Hospital of Shaoxing University, Shaoxing 312000, China
| | - Huanrong Lan
- Department of Breast and Thyroid Surgery, Affiliated Jinhua Hosptial, Zhejiang University School of Medicine, Jinhua 321000, China
| | - Ketao Jin
- Department of Colorectal Surgery, Affiliated Jinhua Hosptial, Zhejiang University School of Medicine, Jinhua 321000, China
| | - Daojun Gong
- Department of Gastrointestinal Surgery, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua 321000, China
| | - Jun Qian
- Department of Colorectal Surgery, Xinchang People’s Hospital, Affiliated Xinchang Hosptial, Wenzhou Medical University, Xinchang 312500, China
| |
Collapse
|
9
|
Ben Khelil M, Aeberli L, Perchaud M, Genolet R, Abdeljaoued S, Borg C, Binda D, Harari A, Jandus C, Muller G, Loyon R. A new workflow combining magnetic cell separation and impedance-based cell dispensing for gentle, simple and reliable cloning of specific CD8+ T cells. SLAS Technol 2022; 27:130-134. [DOI: 10.1016/j.slast.2021.11.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
10
|
Davis L, Tarduno A, Lu YC. Neoantigen-Reactive T Cells: The Driving Force behind Successful Melanoma Immunotherapy. Cancers (Basel) 2021; 13:cancers13236061. [PMID: 34885172 PMCID: PMC8657037 DOI: 10.3390/cancers13236061] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Revised: 11/24/2021] [Accepted: 11/28/2021] [Indexed: 12/20/2022] Open
Abstract
Simple Summary Cancer immunotherapy is a revolutionary type of cancer therapy. It uses the patient’s own immune system to fight and potentially cure cancer. The first major breakthrough of immunotherapy came from successful clinical trials for melanoma treatments. Since then, researchers have focused on understanding the science behind immunotherapy, so that patients with other types of cancer may also benefit. One of the major findings is that the T cells in melanoma patients may recognize a specific type of tumor antigen, called neoantigens, and then kill tumor cells that present these neoantigens. The neoantigens mainly arise from the DNA mutations found in tumor cells. These mutations are translated into mutated proteins that are then distinguished by T cells. In this article, we discuss the critical role of T cells in immunotherapy, as well as the clinical trials that shaped the treatments for melanoma. Abstract Patients with metastatic cutaneous melanoma have experienced significant clinical responses after checkpoint blockade immunotherapy or adoptive cell therapy. Neoantigens are mutated proteins that arise from tumor-specific mutations. It is hypothesized that the neoantigen recognition by T cells is the critical step for T-cell-mediated anti-tumor responses and subsequent tumor regressions. In addition to describing neoantigens, we review the sentinel and ongoing clinical trials that are helping to shape the current treatments for patients with cutaneous melanoma. We also present the existing evidence that establishes the correlations between neoantigen-reactive T cells and clinical responses in melanoma immunotherapy.
Collapse
Affiliation(s)
- Lindy Davis
- Department of Surgery, Albany Medical Center, Albany, NY 12208, USA; (L.D.); (A.T.)
| | - Ashley Tarduno
- Department of Surgery, Albany Medical Center, Albany, NY 12208, USA; (L.D.); (A.T.)
| | - Yong-Chen Lu
- Department of Pathology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
- Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
- Correspondence:
| |
Collapse
|
11
|
Sun Y, Li F, Sonnemann H, Jackson KR, Talukder AH, Katailiha AS, Lizee G. Evolution of CD8 + T Cell Receptor (TCR) Engineered Therapies for the Treatment of Cancer. Cells 2021; 10:cells10092379. [PMID: 34572028 PMCID: PMC8469972 DOI: 10.3390/cells10092379] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2021] [Revised: 08/31/2021] [Accepted: 09/01/2021] [Indexed: 12/30/2022] Open
Abstract
Engineered T cell receptor T (TCR-T) cell therapy has facilitated the generation of increasingly reliable tumor antigen-specific adaptable cellular products for the treatment of human cancer. TCR-T cell therapies were initially focused on targeting shared tumor-associated peptide targets, including melanoma differentiation and cancer-testis antigens. With recent technological developments, it has become feasible to target neoantigens derived from tumor somatic mutations, which represents a highly personalized therapy, since most neoantigens are patient-specific and are rarely shared between patients. TCR-T therapies have been tested for clinical efficacy in treating solid tumors in many preclinical studies and clinical trials all over the world. However, the efficacy of TCR-T therapy for the treatment of solid tumors has been limited by a number of factors, including low TCR avidity, off-target toxicities, and target antigen loss leading to tumor escape. In this review, we discuss the process of deriving tumor antigen-specific TCRs, including the identification of appropriate tumor antigen targets, expansion of antigen-specific T cells, and TCR cloning and validation, including techniques and tools for TCR-T cell vector construction and expression. We highlight the achievements of recent clinical trials of engineered TCR-T cell therapies and discuss the current challenges and potential solutions for improving their safety and efficacy, insights that may help guide future TCR-T studies in cancer.
Collapse
Affiliation(s)
- Yimo Sun
- Department of Melanoma, University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA; (Y.S.); (F.L.); (H.S.); (K.R.J.); (A.H.T.); (A.S.K.)
| | - Fenge Li
- Department of Melanoma, University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA; (Y.S.); (F.L.); (H.S.); (K.R.J.); (A.H.T.); (A.S.K.)
| | - Heather Sonnemann
- Department of Melanoma, University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA; (Y.S.); (F.L.); (H.S.); (K.R.J.); (A.H.T.); (A.S.K.)
| | - Kyle R. Jackson
- Department of Melanoma, University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA; (Y.S.); (F.L.); (H.S.); (K.R.J.); (A.H.T.); (A.S.K.)
| | - Amjad H. Talukder
- Department of Melanoma, University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA; (Y.S.); (F.L.); (H.S.); (K.R.J.); (A.H.T.); (A.S.K.)
| | - Arjun S. Katailiha
- Department of Melanoma, University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA; (Y.S.); (F.L.); (H.S.); (K.R.J.); (A.H.T.); (A.S.K.)
| | - Gregory Lizee
- Department of Melanoma, University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA; (Y.S.); (F.L.); (H.S.); (K.R.J.); (A.H.T.); (A.S.K.)
- Department of Immunology, University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA
- Correspondence:
| |
Collapse
|
12
|
Han Y, Xia K, Su T. Exploration of the Important Role of Microfibril-Associated Protein 4 Gene in Oral Squamous Cell Carcinoma. Med Sci Monit 2021; 27:e931238. [PMID: 34210950 PMCID: PMC8259352 DOI: 10.12659/msm.931238] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
Background Oral squamous cell carcinoma (OSCC) is a common tumor of the head and neck. Its treatment usually requires multiple modalities. Currently, there are no molecular biomarkers to guide these treatment strategies. Studies have shown that microfibril-associated protein 4 (MFAP4) is potentially useful for non-invasive assessment of various diseases; however, its biological function in tumors is still unknown. In this study, we propose that MFAP4 is a new prognostic target for OSCC. Material/Methods First, we collected OSCC data (GSE25099 and GSE30784 datasets) from the Gene Expression Omnibus (GEO) database and compared the differential expression of MFAP4 gene between the patients (tumor) and normal (control) groups. The comparison was done with University of California Santa Cruz Xena (https://xenabrowser.net/Datapages/), and we calculated the difference in MFAP4 gene expression between normal and tumor tissues in a pan-cancer analysis. Then, we compared the 2 groups with high and low expression of MFAP4 gene in terms of tumor mutation burden (TMB), miRNA regulation, and immune cell infiltration. Results We found that the expression of MFAP4 gene was significantly decreased in tumors. Our research also showed that high expression of MFAP4 was related to better prognosis of patients and may be related to tumor gene mutation, miRNA regulation, and infiltration of different immune cells. Conclusions Our work provides evidence that expression of MFAP4 can be used as a prognostic biomarker for risk stratification of OSCC patients and elaborates on its relation with the regulation of TMB, miRNAs, and immune cell infiltration.
Collapse
Affiliation(s)
- Ying Han
- Center for Medical Genetics and Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, China (mainland)
| | - Kun Xia
- Center for Medical Genetics and Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, China (mainland)
| | - Tong Su
- Department of Oral and Maxillofacial Surgery, Center of Stomatology, Xiangya Hospital Central South University, Changsha, Hunan, China (mainland).,Institute of Oral Cancer and Precancerous Lesions, Central South University, Changsha, Hunan, China (mainland).,Research Center of Oral and Maxillofacial Tumors, Xiangya Hospital, Central South University, Changsha, Hunan, China (mainland)
| |
Collapse
|
13
|
Amiset L, Fend L, Gatard-Scheikl T, Rittner K, Duong V, Rooke R, Muller S, Bonnefoy JY, Préville X, Haegel H. TLR2 ligation protects effector T cells from regulatory T-cell mediated suppression and repolarizes T helper responses following MVA-based cancer immunotherapy. Oncoimmunology 2021; 1:1271-1280. [PMID: 23243590 PMCID: PMC3518499 DOI: 10.4161/onci.21479] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Cancer immunotherapy is hampered by the immunosuppression maintained by regulatory T cells (Tregs) in tumor-bearing hosts. Stimulation of the Toll-like receptor 2 (TLR2) by Pam3Cys is known to affect Treg-mediated suppression. We found that Pam3Cys increases the proliferation of both CD4+ effector T cells (Teffs) and Tregs co-cultured in vitro, but did not induce the proliferation of Tregs alone upon CD3 and CD28 stimulation. In a mouse model of RMA-MUC1 tumors, Pam3Cys was administered either alone or in combination with a modified vaccinia ankara (MVA)-based mucin 1 (MUC1) therapeutic vaccine. The combination of Pam3Cys with MVA-MUC1 (1) diminished splenic Treg/CD4+ T-cell ratios to those found in tumor-free mice, (2) stimulated a specific anti-MUC1 interferon γ (IFNγ) response and (3) had a significant therapeutic effect on tumor growth and mouse survival. When CD4+ Teffs and Tregs were isolated from Pam3Cys-treated mice, Teffs had become resistant to Treg-mediated suppression while upregulating the expression of BclL-xL. Tregs from Pam3Cys-treated mice were fully suppressive for Teffs from naïve mice. Bcl-xL was induced by Pam3Cys with different kinetics in Tregs and Teffs. Teff from Pam3Cys-treated mice produced increased levels of Th1 and Th2-type cytokines and an interleukin (IL)-6-dependent secretion of IL-17 was observed in Teff:Treg co-cultures, suggesting that TLR2 stimulation had skewed the immune response toward a Th17 profile. Our results show for the first time that in a tumor-bearing host, TLR2 stimulation with Pam3Cys affects both Tregs and Teffs, protects Teff from Treg-mediated suppression and has strong therapeutic effects when combined with an MVA-based antitumor vaccine.
Collapse
Affiliation(s)
- Laurent Amiset
- Département d'Immunopharmacologie; Transgene S.A.; Parc d'Innovation; Illkirch-Graffenstaden, France
| | | | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Manfredi F, Cianciotti BC, Potenza A, Tassi E, Noviello M, Biondi A, Ciceri F, Bonini C, Ruggiero E. TCR Redirected T Cells for Cancer Treatment: Achievements, Hurdles, and Goals. Front Immunol 2020; 11:1689. [PMID: 33013822 PMCID: PMC7494743 DOI: 10.3389/fimmu.2020.01689] [Citation(s) in RCA: 72] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Accepted: 06/24/2020] [Indexed: 12/11/2022] Open
Abstract
Adoptive T cell therapy (ACT) is a rapidly evolving therapeutic approach designed to harness T cell specificity and function to fight diseases. Based on the evidence that T lymphocytes can mediate a potent anti-tumor response, initially ACT solely relied on the isolation, in vitro expansion, and infusion of tumor-infiltrating or circulating tumor-specific T cells. Although effective in a subset of cases, in the first ACT clinical trials several patients experienced disease progression, in some cases after temporary disease control. This evidence prompted researchers to improve ACT products by taking advantage of the continuously evolving gene engineering field and by improving manufacturing protocols, to enable the generation of effective and long-term persisting tumor-specific T cell products. Despite recent advances, several challenges, including prioritization of antigen targets, identification, and optimization of tumor-specific T cell receptors, in the development of tools enabling T cells to counteract the immunosuppressive tumor microenvironment, still need to be faced. This review aims at summarizing the major achievements, hurdles and possible solutions designed to improve the ACT efficacy and safety profile in the context of liquid and solid tumors.
Collapse
Affiliation(s)
- Francesco Manfredi
- Vita-Salute San Raffaele University, Milan, Italy
- Experimental Hematology Unit, Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Beatrice Claudia Cianciotti
- Experimental Hematology Unit, Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Fondazione Centro San Raffaele, Milan, Italy
| | - Alessia Potenza
- Experimental Hematology Unit, Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
- School of Medicine and Surgery, University of Milano – Bicocca, Milan, Italy
| | - Elena Tassi
- Experimental Hematology Unit, Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Maddalena Noviello
- Experimental Hematology Unit, Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Andrea Biondi
- Clinica Pediatrica Università degli Studi di Milano Bicocca, Fondazione MBBM, Monza, Italy
| | - Fabio Ciceri
- Vita-Salute San Raffaele University, Milan, Italy
- Experimental Hematology Unit, Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Chiara Bonini
- Vita-Salute San Raffaele University, Milan, Italy
- Experimental Hematology Unit, Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Eliana Ruggiero
- Experimental Hematology Unit, Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
| |
Collapse
|
15
|
Bai R, Chen N, Liang T, Li L, Lv Z, Lv X, Cui J. Novel Frontiers of Treatment for Advanced Gastric or Gastroesophageal Junction Cancer (GC/GEJC): Will Immunotherapy Be a Future Direction? Front Oncol 2020; 10:912. [PMID: 32793461 PMCID: PMC7386300 DOI: 10.3389/fonc.2020.00912] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Accepted: 05/11/2020] [Indexed: 12/19/2022] Open
Abstract
Considering the limited progress of chemotherapy and targeted therapy in improving the generally disappointing outcomes of advanced gastric or gastroesophageal junction cancer (GC/GEJC), immunotherapies have been gradually developed and advanced into novel frontiers of treatment for advanced GC/GEJC. Nevertheless, the response to immunotherapy was not always satisfactory, and the emergence of resistance was unavoidable. These factors prompt the development of different combination therapies and predictive and prognostic biomarkers of efficacy to improve the outcomes of patients with advanced GC/GEJC and to overcome drug resistance. This article discusses the advances of immune monotherapy, multiple current and ongoing clinical trials of immune combination therapy, immune-related adverse events, and various biomarkers in GC/GEJC.
Collapse
Affiliation(s)
- Rilan Bai
- Cancer Center, The First Hospital of Jilin University, Changchun, China
| | - Naifei Chen
- Cancer Center, The First Hospital of Jilin University, Changchun, China
| | - Tingting Liang
- Cancer Center, The First Hospital of Jilin University, Changchun, China
| | - Lingyu Li
- Cancer Center, The First Hospital of Jilin University, Changchun, China
| | - Zheng Lv
- Cancer Center, The First Hospital of Jilin University, Changchun, China
| | - Xiaomin Lv
- Department of Neurology, The First Hospital of Jilin University, Changchun, China
| | - Jiuwei Cui
- Cancer Center, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
16
|
Gerber HP, Sibener LV, Lee LJ, Gee MH. Identification of Antigenic Targets. Trends Cancer 2020; 6:299-318. [PMID: 32209445 DOI: 10.1016/j.trecan.2020.01.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2019] [Accepted: 01/06/2020] [Indexed: 12/31/2022]
Abstract
The ideal cancer target antigen (Ag) is expressed at high copy numbers on neoplastic cells, absent on normal tissues, and contributes to the survival of cancer cells. Despite significant investments in the identification of cell surface Ags, there is a paucity of targets that meet such ideal cancer target criteria. Recent clinical trials in patients with cancer treated with immune checkpoint inhibitors (ICIs) indicate that cluster of differentiation (CD)8+ T cells, by means of their T cell receptors (TCRs) recognizing intracellular targets presented as peptides in the context of human leukocyte antigen (peptide-human leukocyte antigen complex; pHLA) molecules on tumor cells, can mediate deep and long-lasting antitumor responses in patients with solid tumors. Therefore, pHLA-target Ags may represent the long sought-after, ideal targets for solid tumor targeting by high-potency oncology compounds.
Collapse
Affiliation(s)
| | - Leah V Sibener
- 3T Biosciences, 1455 Adams Drive, Menlo Park, CA 94025, USA
| | - Luke J Lee
- 3T Biosciences, 1455 Adams Drive, Menlo Park, CA 94025, USA
| | - Marvin H Gee
- 3T Biosciences, 1455 Adams Drive, Menlo Park, CA 94025, USA
| |
Collapse
|
17
|
Genetically Modified T-Cell Therapy for Osteosarcoma: Into the Roaring 2020s. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1257:109-131. [PMID: 32483735 DOI: 10.1007/978-3-030-43032-0_10] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
T-cell immunotherapy may offer an approach to improve outcomes for patients with osteosarcoma who fail current therapies. In addition, it has the potential to reduce treatment-related complications for all patients. Generating tumor-specific T cells with conventional antigen-presenting cells ex vivo is time-consuming and often results in T-cell products with a low frequency of tumor-specific T cells. Furthermore, the generated T cells remain sensitive to the immunosuppressive tumor microenvironment. Genetic modification of T cells is one strategy to overcome these limitations. For example, T cells can be genetically modified to render them antigen specific, resistant to inhibitory factors, or increase their ability to home to tumor sites. Most genetic modification strategies have only been evaluated in preclinical models; however, early clinical phase trials are in progress. In this chapter, we will review the current status of gene-modified T-cell therapy with special focus on osteosarcoma, highlighting potential antigenic targets, preclinical and clinical studies, and strategies to improve current T-cell therapy approaches.
Collapse
|
18
|
Fain JS, Van Tongelen A, Loriot A, De Smet C. Epigenetic Coactivation of MAGEA6 and CT-GABRA3 Defines Orientation of a Segmental Duplication in the Human X Chromosome. Cytogenet Genome Res 2019; 159:12-18. [PMID: 31593956 DOI: 10.1159/000502933] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/08/2019] [Indexed: 12/17/2022] Open
Abstract
The human genome harbors many duplicated segments, which sometimes show very high sequence identity. This may complicate assignment during genome assembly. One such example is in Xq28, where the arrangement of 2 recently duplicated segments varies between genome assembly versions. The duplicated segments comprise highly similar genes, including MAGEA3 and MAGEA6, which display specific expression in testicular germline cells, and also become aberrantly activated in a variety of tumors. Recently, a new gene was identified, CT-GABRA3, the transcription of which initiates inside the segmental duplication but extends far outside. According to the latest genome annotation, CT- GABRA3 starts near MAGEA3, with which it shares a bidirectional promoter. In an earlier annotation, however, the duplicated segment was positioned in the opposite orientation, and CT-GABRA3 was instead coupled with MAGEA6. To resolve this discrepancy, and based on the contention that genes connected by a bidirectional promoter are almost always co-expressed, we decided to compare the expression profiles of CT-GABRA3, MAGEA3, and MAGEA6. We found that in tumor tissues and cell lines of different origins, the expression of CT-GABRA3 was better correlated with that of MAGEA6. Moreover, in a cellular model of experimental induction with a DNA demethylation agent, activation CT-GABRA3 was associated with that of MAGEA6, but not with that of MAGEA3. Together these results support a connection between CT-GABRA3 and MAGEA6 and illustrate how promoter-sharing genes can be exploited to resolve genome assembly uncertainties.
Collapse
MESH Headings
- Antigens, Neoplasm/genetics
- Antigens, Neoplasm/metabolism
- Chromosomes, Human, X/genetics
- Epigenesis, Genetic/genetics
- Gene Duplication/genetics
- Gene Expression Regulation, Neoplastic/genetics
- Genome, Human/genetics
- Humans
- Neoplasm Proteins/genetics
- Neoplasm Proteins/metabolism
- Neoplasms/genetics
- Neoplasms/pathology
- Promoter Regions, Genetic/genetics
- Receptors, GABA-A/genetics
- Segmental Duplications, Genomic/genetics
- Tumor Cells, Cultured
Collapse
|
19
|
Zhang Y, Lin S, Wang XY, Zhu G. Nanovaccines for cancer immunotherapy. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2019; 11:e1559. [PMID: 31172659 PMCID: PMC7040494 DOI: 10.1002/wnan.1559] [Citation(s) in RCA: 74] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Accepted: 03/27/2019] [Indexed: 12/17/2022]
Abstract
The past few decades have witnessed the booming field of cancer immunotherapy. Cancer therapeutic vaccines, either alone or in combination with other immunotherapies such as adoptive cell therapy or immune checkpoint blockade therapy, are an attractive class of cancer immunotherapeutics. However, cancer vaccines have thus far shown suboptimal efficacy in the clinic. Nanomedicines offer unique opportunities to improve the efficacy of these vaccines. A variety of nanoplatforms have been investigated to deliver molecular or cellular or subcellular vaccines to target lymphoid tissues and cells, thereby promoting the potency and durability of anti-tumor immunity while reducing adverse side effects. In this article, we reviewed the key parameters and features of nanovaccines for cancer immunotherapy; we highlighted recent advances in the development of cancer nanovaccines based on synthetic nanocarriers, biogenic nanocarriers, as well as semi-biogenic nanocarriers; and we summarized newly emerging types of nanovaccines, such as those based on stimulator of interferon genes agonists, cancer neoantigens, mRNA vaccines, as well as artificial antigen-presenting cells. This article is categorized under: Therapeutic Approaches and Drug Discovery > Nanomedicine for Oncologic Disease.
Collapse
Affiliation(s)
- Yu Zhang
- Department of Rehabilitation Medicine, Center for Translational Medicine, Precision Medicine Institute, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
- Center for Pharmaceutical Engineering and Sciences, Department of Pharmaceutics, School of Pharmacy, Virginia Commonwealth University, Richmond, Virginia
| | - Shuibin Lin
- Department of Rehabilitation Medicine, Center for Translational Medicine, Precision Medicine Institute, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Xiang-Yang Wang
- Department of Human and Molecular Genetics, Virginia Commonwealth University, Richmond, Virginia
- Institute of Molecular Medicine, Virginia Commonwealth University, Richmond, Virginia
- Massey Cancer Center, Virginia Commonwealth University, Richmond, Virginia
| | - Guizhi Zhu
- Center for Pharmaceutical Engineering and Sciences, Department of Pharmaceutics, School of Pharmacy, Virginia Commonwealth University, Richmond, Virginia
- The Developmental Therapeutics Program, Massey Cancer Center, Virginia Commonwealth University, Richmond, Virginia
| |
Collapse
|
20
|
Baldin AV, Zamyatnin AA, Bazhin AV, Xu WH, Savvateeva LV. Advances in the Development of Anticancer HSP-based Vaccines. Curr Med Chem 2019; 26:427-445. [PMID: 29376489 DOI: 10.2174/0929867325666180129100015] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2017] [Revised: 10/11/2017] [Accepted: 01/01/2018] [Indexed: 01/01/2023]
Abstract
Current advances in cancer treatment are based on the recent discoveries of molecular mechanisms of tumour maintenance. It was shown that heat shock proteins (HSPs) play a crucial role in the development of immune response against tumours. Thus, HSPs represent multifunctional agents not only with chaperone functions, but also possessing immunomodulatory properties. These properties are exploited for the development of HSP-based anticancer vaccines aimed to induce cytotoxic responses against tumours. To date, a number of strategies have been suggested to facilitate HSP-based vaccine production and to increase its effectiveness. The present review focuses on the current trend for the development of HSPbased vaccines aimed at inducing strong immunological tumour-specific responses against cancer cells of distinct etiology and localization.
Collapse
Affiliation(s)
- Alexey V Baldin
- Sechenov First Moscow State Medical University, Institute of Molecular Medicine, 119991, Moscow, Russian Federation
| | - Andrey A Zamyatnin
- Sechenov First Moscow State Medical University, Institute of Molecular Medicine, 119991, Moscow, Russian Federation.,Lomonosov Moscow State University, Department of Cell Signaling, Belozersky Institute of Physico- Chemical Biology, 119991, Moscow, Russian Federation
| | - Alexandr V Bazhin
- Department of General, Visceral, and Transplant Surgery, Ludwig-Maximilians-University Munich, Germany.,German Cancer Consortium (DKTK), Partner Site Munich, Germany
| | - Wan-Hai Xu
- Department of Urology, the Fourth Hospital of Harbin Medical University, Harbin, China
| | - Lyudmila V Savvateeva
- Sechenov First Moscow State Medical University, Institute of Molecular Medicine, 119991, Moscow, Russian Federation
| |
Collapse
|
21
|
Antohe M, Nedelcu RI, Nichita L, Popp CG, Cioplea M, Brinzea A, Hodorogea A, Calinescu A, Balaban M, Ion DA, Diaconu C, Bleotu C, Pirici D, Zurac SA, Turcu G. Tumor infiltrating lymphocytes: The regulator of melanoma evolution. Oncol Lett 2019; 17:4155-4161. [PMID: 30944610 PMCID: PMC6444298 DOI: 10.3892/ol.2019.9940,] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Accepted: 09/27/2018] [Indexed: 01/27/2025] Open
Abstract
Melanoma is the most severe type of skin cancer and its incidence has increased in the last decades. In the United States, it is the 6th most common cancer in both men and women. Prognosis for patients with melanoma depends on the stage of the disease at the time of diagnosis and it can be influenced by the immunologic response. Melanoma has been historically considered an immunogenic malignancy. It often contains great amount of immune cells (different subsets of T-cells, dendritic cells, macrophages, neutrophils, mast cells, B lymphocytes), which may reflect a continuous intercommunication between host and tumor. It is not established if tumor-infiltrating lymphocytes (TILs) are induced by tumor cells or by other components of the microenvironment or when they are a host direct immunologic reaction. It has been observed that in many cases, the presence of a dense TIL is associated with good prognosis. The pattern and activation state of the cells which constitute TIL is variable and modulates the clinical outcome. An important step in the understanding of tumor immunobiology is the analysis of the populations and subsets of immune cells that form TIL. Besides its prognostic significance, after approval of cytotoxic T lymphocyte antigen 4, programmed cell death-1 and programmed death-1 ligand antibodies for the treatment of melanoma, the assessment of immune infiltrate composition has become even more captivating, as it could provide new target molecules and new biomarkers for predicting the effect of the treatment and disease outcome in patients treated with immunotherapy. In this review we discuss current state of knowledge in the field of immune cells that infiltrate melanoma, resuming the potential of TIL components to become prognostic markers for natural evolution, for response to drugs or valuable targets for new medication.
Collapse
Affiliation(s)
- Mihaela Antohe
- Department of Pathophysiology, ‘Carol Davila’ University of Medicine and Pharmacy, 050474 Bucharest, Romania
- Department of Dermatology, Derma 360° Clinic, 011274 Bucharest, Romania
| | - Roxana Ioana Nedelcu
- Department of Pathophysiology, ‘Carol Davila’ University of Medicine and Pharmacy, 050474 Bucharest, Romania
- Department of Dermatology, Derma 360° Clinic, 011274 Bucharest, Romania
| | - Luciana Nichita
- Department of Pathology, Colentina Clinical Hospital, 021103 Bucharest, Romania
| | | | - Mirela Cioplea
- Department of Pathology, Colentina Clinical Hospital, 021103 Bucharest, Romania
| | - Alice Brinzea
- Department of Pathophysiology, ‘Carol Davila’ University of Medicine and Pharmacy, 050474 Bucharest, Romania
- National Institute for Infectious Diseases ‘Prof. Dr. Matei Balș’, Ambulatory Service, 021105 Bucharest, Romania
| | - Anastasia Hodorogea
- Department of Pathophysiology, ‘Carol Davila’ University of Medicine and Pharmacy, 050474 Bucharest, Romania
- Department of Dermatology, Colentina Clinical Hospital, 021103 Bucharest, Romania
| | - Andreea Calinescu
- Department of Dermatology, Colentina Clinical Hospital, 021103 Bucharest, Romania
- Department of Physiology, ‘Carol Davila’ University of Medicine and Pharmacy, 050474 Bucharest, Romania
| | - Mihaela Balaban
- Department of Dermatology, Derma 360° Clinic, 011274 Bucharest, Romania
- Department of Biochemistry, ‘Carol Davila’ University of Medicine and Pharmacy, 050474 Bucharest, Romania
| | - Daniela Adriana Ion
- Department of Pathophysiology, ‘Carol Davila’ University of Medicine and Pharmacy, 050474 Bucharest, Romania
| | - Carmen Diaconu
- Department of Cellular and Molecular Pathology, ‘Stefan S. Nicolau’ Institute of Virology, 030304 Bucharest, Romania
| | - Coralia Bleotu
- Department of Cellular and Molecular Pathology, ‘Stefan S. Nicolau’ Institute of Virology, 030304 Bucharest, Romania
| | - Daniel Pirici
- Department of Pathology, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| | - Sabina Andrada Zurac
- Department of Pathology, Colentina Clinical Hospital, 021103 Bucharest, Romania
- Department of Pathology, ‘Carol Davila’ University of Medicine and Pharmacy, 050474 Bucharest, Romania
| | - Gabriela Turcu
- Department of Dermatology, Derma 360° Clinic, 011274 Bucharest, Romania
- Department of Dermatology, Colentina Clinical Hospital, 021103 Bucharest, Romania
- Department of Dermatology, ‘Carol Davila’ University of Medicine and Pharmacy, 050474 Bucharest, Romania
| |
Collapse
|
22
|
Antohe M, Nedelcu RI, Nichita L, Popp CG, Cioplea M, Brinzea A, Hodorogea A, Calinescu A, Balaban M, Ion DA, Diaconu C, Bleotu C, Pirici D, Zurac SA, Turcu G. Tumor infiltrating lymphocytes: The regulator of melanoma evolution. Oncol Lett 2019; 17:4155-4161. [PMID: 30944610 PMCID: PMC6444298 DOI: 10.3892/ol.2019.9940] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Accepted: 09/27/2018] [Indexed: 02/06/2023] Open
Abstract
Melanoma is the most severe type of skin cancer and its incidence has increased in the last decades. In the United States, it is the 6th most common cancer in both men and women. Prognosis for patients with melanoma depends on the stage of the disease at the time of diagnosis and it can be influenced by the immunologic response. Melanoma has been historically considered an immunogenic malignancy. It often contains great amount of immune cells (different subsets of T-cells, dendritic cells, macrophages, neutrophils, mast cells, B lymphocytes), which may reflect a continuous intercommunication between host and tumor. It is not established if tumor-infiltrating lymphocytes (TILs) are induced by tumor cells or by other components of the microenvironment or when they are a host direct immunologic reaction. It has been observed that in many cases, the presence of a dense TIL is associated with good prognosis. The pattern and activation state of the cells which constitute TIL is variable and modulates the clinical outcome. An important step in the understanding of tumor immunobiology is the analysis of the populations and subsets of immune cells that form TIL. Besides its prognostic significance, after approval of cytotoxic T lymphocyte antigen 4, programmed cell death-1 and programmed death-1 ligand antibodies for the treatment of melanoma, the assessment of immune infiltrate composition has become even more captivating, as it could provide new target molecules and new biomarkers for predicting the effect of the treatment and disease outcome in patients treated with immunotherapy. In this review we discuss current state of knowledge in the field of immune cells that infiltrate melanoma, resuming the potential of TIL components to become prognostic markers for natural evolution, for response to drugs or valuable targets for new medication.
Collapse
Affiliation(s)
- Mihaela Antohe
- Department of Pathophysiology, ‘Carol Davila’ University of Medicine and Pharmacy, 050474 Bucharest, Romania
- Department of Dermatology, Derma 360° Clinic, 011274 Bucharest, Romania
| | - Roxana Ioana Nedelcu
- Department of Pathophysiology, ‘Carol Davila’ University of Medicine and Pharmacy, 050474 Bucharest, Romania
- Department of Dermatology, Derma 360° Clinic, 011274 Bucharest, Romania
| | - Luciana Nichita
- Department of Pathology, Colentina Clinical Hospital, 021103 Bucharest, Romania
| | | | - Mirela Cioplea
- Department of Pathology, Colentina Clinical Hospital, 021103 Bucharest, Romania
| | - Alice Brinzea
- Department of Pathophysiology, ‘Carol Davila’ University of Medicine and Pharmacy, 050474 Bucharest, Romania
- National Institute for Infectious Diseases ‘Prof. Dr. Matei Balș’, Ambulatory Service, 021105 Bucharest, Romania
| | - Anastasia Hodorogea
- Department of Pathophysiology, ‘Carol Davila’ University of Medicine and Pharmacy, 050474 Bucharest, Romania
- Department of Dermatology, Colentina Clinical Hospital, 021103 Bucharest, Romania
| | - Andreea Calinescu
- Department of Dermatology, Colentina Clinical Hospital, 021103 Bucharest, Romania
- Department of Physiology, ‘Carol Davila’ University of Medicine and Pharmacy, 050474 Bucharest, Romania
| | - Mihaela Balaban
- Department of Dermatology, Derma 360° Clinic, 011274 Bucharest, Romania
- Department of Biochemistry, ‘Carol Davila’ University of Medicine and Pharmacy, 050474 Bucharest, Romania
| | - Daniela Adriana Ion
- Department of Pathophysiology, ‘Carol Davila’ University of Medicine and Pharmacy, 050474 Bucharest, Romania
| | - Carmen Diaconu
- Department of Cellular and Molecular Pathology, ‘Stefan S. Nicolau’ Institute of Virology, 030304 Bucharest, Romania
| | - Coralia Bleotu
- Department of Cellular and Molecular Pathology, ‘Stefan S. Nicolau’ Institute of Virology, 030304 Bucharest, Romania
| | - Daniel Pirici
- Department of Pathology, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| | - Sabina Andrada Zurac
- Department of Pathology, Colentina Clinical Hospital, 021103 Bucharest, Romania
- Department of Pathology, ‘Carol Davila’ University of Medicine and Pharmacy, 050474 Bucharest, Romania
| | - Gabriela Turcu
- Department of Dermatology, Derma 360° Clinic, 011274 Bucharest, Romania
- Department of Dermatology, Colentina Clinical Hospital, 021103 Bucharest, Romania
- Department of Dermatology, ‘Carol Davila’ University of Medicine and Pharmacy, 050474 Bucharest, Romania
| |
Collapse
|
23
|
Shi DB, Ma RR, Zhang H, Hou F, Guo XY, Gao P. GAGE7B promotes tumor metastasis and growth via activating the p38δ/pMAPKAPK2/pHSP27 pathway in gastric cancer. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2019; 38:124. [PMID: 30871606 PMCID: PMC6419436 DOI: 10.1186/s13046-019-1125-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Accepted: 03/01/2019] [Indexed: 01/01/2023]
Abstract
Background Gastric cancer is the second most common cause of cancer-related mortality; thus, the mechanisms underlying tumor metastasis and growth in gastric cancer need to be extensively explored. Methods Differentially expressed genes were examined in gastric cancer samples with lymph node metastasis (LNM) and without LNM using mRNA microarray and RT-qPCR. The effects of G antigen 7B (GAGE7B) on the metastasis, growth, and angiogenesis of gastric cancer were investigated in vitro and in vivo. GAGE7B protein expression was detected by immunohistochemical (IHC) analysis. Microarray, RT-qPCR, and western blot assays were performed to detect downstream target genes of GAGE7B. Dual-luciferase reporter and western blot assays were used to identify miRNAs that could negatively regulate GAGE7B. Results GAGE7B was significantly overexpressed in samples with LNM. High expression levels of GAGE7B were associated with advanced clinical stage and poor patient survival. GAGE7B dramatically enhanced the metastasis, growth, and angiogenesis ability of gastric cancer. GAGE7B was further demonstrated to promote the progression of gastric cancer by activating the p38δ/pMAPKAPK2/pHSP27 pathway. However, the GAGE7B-induced p38δ/pMAPKAPK2/pHSP27 pathway was inactivated by miR-30c, as the expression levels of both GAGE7B and p38δ were found to be directly suppressed by miR-30c. Intriguingly, GAGE7B was found to be a ceRNA for p38δ, as it activated the p38δ/pMAPKAPK2/pHSP27 pathway by competitively binding miR-30c. Conclusions GAGE7B may serve as a prognostic indicator in gastric cancer. GAGE7B significantly promotes gastric cancer progression by upregulating the p38δ/pMAPKAPK2/pHSP27 pathway, but it is negatively regulated by miR-30c. GAGE7B and miR-30c may be potential therapeutic targets in gastric cancer. Electronic supplementary material The online version of this article (10.1186/s13046-019-1125-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Duan-Bo Shi
- Department of Pathology, School of Medicine, Shandong University, Jinan, 250012, China.,Department of Pathology, Qilu Hospital, Shandong University, Jinan, 250012, China
| | - Ran-Ran Ma
- Department of Pathology, School of Medicine, Shandong University, Jinan, 250012, China.,Department of Pathology, Qilu Hospital, Shandong University, Jinan, 250012, China
| | - Hui Zhang
- Department of Pathology, School of Medicine, Shandong University, Jinan, 250012, China.,Department of Pathology, Qilu Hospital, Shandong University, Jinan, 250012, China
| | - Feng Hou
- Department of Pathology, School of Medicine, Shandong University, Jinan, 250012, China.,Department of Pathology, The Affiliated Hospital Of Qingdao University, Qingdao, 266071, China
| | - Xiang-Yu Guo
- Department of Pathology, School of Medicine, Shandong University, Jinan, 250012, China.,Department of Pathology, Qilu Hospital, Shandong University, Jinan, 250012, China
| | - Peng Gao
- Department of Pathology, School of Medicine, Shandong University, Jinan, 250012, China. .,Department of Pathology, Qilu Hospital, Shandong University, Jinan, 250012, China.
| |
Collapse
|
24
|
Lee JJ, Chu E. Recent Advances in the Clinical Development of Immune Checkpoint Blockade Therapy for Mismatch Repair Proficient (pMMR)/non-MSI-H Metastatic Colorectal Cancer. Clin Colorectal Cancer 2018; 17:258-273. [PMID: 30072278 PMCID: PMC6612427 DOI: 10.1016/j.clcc.2018.06.004] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Revised: 06/10/2018] [Accepted: 06/14/2018] [Indexed: 12/11/2022]
Abstract
Metastatic colorectal cancer (mCRC) continues to be associated with a poor prognosis, and there remains a significant unmet need for novel agents and treatment regimens. Major breakthroughs have been made with immune checkpoint blockade therapy in several disease types, including DNA mismatch repair deficient/microsatellite instability-high (MSI-H) tumors. To date, however, immune checkpoint monotherapy has not shown significant clinical activity in the treatment of patients with mismatch repair proficient (pMMR)/non-MSI-H mCRC. The immune resistance mechanisms in pMMR/non-MSI-H mCRC have not yet been clearly elucidated. Significant efforts are currently focused on identifying effective combination immunotherapy regimens for the treatment of patients with pMMR/non-MSI-H mCRC. The combination of atezolizumab with cobimetinib had shown promising clinical activity in an early-phase clinical trial. Unfortunately, the IMblaze 370 (COTEZO) phase III trial of atezolizumab/cobimetinib combination in patients with mCRC failed to show significant improvement in overall survival in patients treated with the atezolizumab/combimetinib combination in comparison with regorafenib alone. This review summarizes the recent major advances in the clinical development of immunotherapy regimens for patients with pMMR/non-MSI-H mCRC.
Collapse
Affiliation(s)
- James J Lee
- Division of Hematology-Oncology, Department of Medicine, Cancer Therapeutics Program, UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, PA.
| | - Edward Chu
- Division of Hematology-Oncology, Department of Medicine, Cancer Therapeutics Program, UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, PA
| |
Collapse
|
25
|
Yang YJ, Mai DJ, Dursch TJ, Olsen BD. Nucleopore-Inspired Polymer Hydrogels for Selective Biomolecular Transport. Biomacromolecules 2018; 19:3905-3916. [DOI: 10.1021/acs.biomac.8b00556] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Affiliation(s)
- Yun Jung Yang
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| | - Danielle J. Mai
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| | - Thomas J. Dursch
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| | - Bradley D. Olsen
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| |
Collapse
|
26
|
Shukla SA, Bachireddy P, Schilling B, Galonska C, Zhan Q, Bango C, Langer R, Lee PC, Gusenleitner D, Keskin DB, Babadi M, Mohammad A, Gnirke A, Clement K, Cartun ZJ, Van Allen EM, Miao D, Huang Y, Snyder A, Merghoub T, Wolchok JD, Garraway LA, Meissner A, Weber JS, Hacohen N, Neuberg D, Potts PR, Murphy GF, Lian CG, Schadendorf D, Hodi FS, Wu CJ. Cancer-Germline Antigen Expression Discriminates Clinical Outcome to CTLA-4 Blockade. Cell 2018; 173:624-633.e8. [PMID: 29656892 DOI: 10.1016/j.cell.2018.03.026] [Citation(s) in RCA: 126] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Revised: 12/11/2017] [Accepted: 03/13/2018] [Indexed: 02/07/2023]
Abstract
CTLA-4 immune checkpoint blockade is clinically effective in a subset of patients with metastatic melanoma. We identify a subcluster of MAGE-A cancer-germline antigens, located within a narrow 75 kb region of chromosome Xq28, that predicts resistance uniquely to blockade of CTLA-4, but not PD-1. We validate this gene expression signature in an independent anti-CTLA-4-treated cohort and show its specificity to the CTLA-4 pathway with two independent anti-PD-1-treated cohorts. Autophagy, a process critical for optimal anti-cancer immunity, has previously been shown to be suppressed by the MAGE-TRIM28 ubiquitin ligase in vitro. We now show that the expression of the key autophagosome component LC3B and other activators of autophagy are negatively associated with MAGE-A protein levels in human melanomas, including samples from patients with resistance to CTLA-4 blockade. Our findings implicate autophagy suppression in resistance to CTLA-4 blockade in melanoma, suggesting exploitation of autophagy induction for potential therapeutic synergy with CTLA-4 inhibitors.
Collapse
Affiliation(s)
- Sachet A Shukla
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA; Broad Institute, Cambridge, MA 02142, USA
| | - Pavan Bachireddy
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA; Broad Institute, Cambridge, MA 02142, USA; Department of Medicine, Brigham & Women's Hospital, Boston, MA 02115, USA
| | - Bastian Schilling
- Department of Dermatology, University Hospital, University Duisburg-Essen, 45147 Essen, Germany; German Cancer Consortium (DKTK), 69121 Heidelberg, Germany; Department of Dermatology, Venereology and Allergology, University Hospital Würzburg, 97080 Würzburg, Germany
| | | | - Qian Zhan
- Program in Dermatopathology, Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Clyde Bango
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA
| | - Rupert Langer
- Department of Pathology, University of Bern, 3012 Bern, Switzerland
| | - Patrick C Lee
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA
| | - Daniel Gusenleitner
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA
| | - Derin B Keskin
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA; Broad Institute, Cambridge, MA 02142, USA; Department of Medicine, Brigham & Women's Hospital, Boston, MA 02115, USA
| | | | | | | | - Kendell Clement
- Broad Institute, Cambridge, MA 02142, USA; Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA
| | - Zachary J Cartun
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA
| | - Eliezer M Van Allen
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA; Broad Institute, Cambridge, MA 02142, USA; Department of Medicine, Brigham & Women's Hospital, Boston, MA 02115, USA
| | - Diana Miao
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA; Broad Institute, Cambridge, MA 02142, USA
| | - Ying Huang
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA
| | - Alexandra Snyder
- Weill Cornell Medical College, New York, NY, USA; Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10016, USA
| | - Taha Merghoub
- Weill Cornell Medical College, New York, NY, USA; Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10016, USA
| | - Jedd D Wolchok
- Weill Cornell Medical College, New York, NY, USA; Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10016, USA
| | - Levi A Garraway
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA; Broad Institute, Cambridge, MA 02142, USA; Department of Medicine, Brigham & Women's Hospital, Boston, MA 02115, USA
| | - Alexander Meissner
- Broad Institute, Cambridge, MA 02142, USA; Max Planck Institute for Molecular Genetics, 14195 Berlin, Germany; Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA
| | - Jeffrey S Weber
- New York University Langone Medical Center, New York, NY 10016, USA
| | | | - Donna Neuberg
- Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
| | - Patrick R Potts
- Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, TN 38105-3678, USA
| | - George F Murphy
- Program in Dermatopathology, Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Christine G Lian
- Program in Dermatopathology, Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Dirk Schadendorf
- Department of Dermatology, University Hospital, University Duisburg-Essen, 45147 Essen, Germany; German Cancer Consortium (DKTK), 69121 Heidelberg, Germany
| | - F Stephen Hodi
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA; Department of Medicine, Brigham & Women's Hospital, Boston, MA 02115, USA
| | - Catherine J Wu
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA; Broad Institute, Cambridge, MA 02142, USA; Department of Medicine, Brigham & Women's Hospital, Boston, MA 02115, USA.
| |
Collapse
|
27
|
Ping Y, Liu C, Zhang Y. T-cell receptor-engineered T cells for cancer treatment: current status and future directions. Protein Cell 2018; 9:254-266. [PMID: 28108950 PMCID: PMC5829268 DOI: 10.1007/s13238-016-0367-1] [Citation(s) in RCA: 109] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Accepted: 12/16/2016] [Indexed: 12/20/2022] Open
Abstract
T-cell receptor (TCR)-engineered T cells are a novel option for adoptive cell therapy used for the treatment of several advanced forms of cancer. Work using TCR-engineered T cells began more than two decades ago, with numerous preclinical studies showing that such cells could mediate tumor lysis and eradication. The success of these trials provided the foundation for clinical trials, including recent clinical successes using TCR-engineered T cells to target New York esophageal squamous cell carcinoma (NY-ESO-1). These successes demonstrate the potential of this approach to treat cancer. In this review, we provide a perspective on the current and future applications of TCR-engineered T cells for the treatment of cancer. Our summary focuses on TCR activation and both pre-clinical and clinical applications of TCR-engineered T cells. We also discuss how to enhance the function of TCR-engineered T cells and prolong their longevity in the tumor microenvironment.
Collapse
Affiliation(s)
- Yu Ping
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Chaojun Liu
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Yi Zhang
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China.
- Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China.
- Engineering Key Laboratory for Cell Therapy of Henan Province, Zhengzhou, 450052, China.
| |
Collapse
|
28
|
Wada S, Yada E, Ohtake J, Sasada T. Personalized peptide vaccines for cancer therapy: current progress and state of the art. EXPERT REVIEW OF PRECISION MEDICINE AND DRUG DEVELOPMENT 2017. [DOI: 10.1080/23808993.2017.1403286] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Satoshi Wada
- Cancer Immunotherapy, Kanagawa Cancer Center, Asahi-ku, Yokohama, Japan
| | - Erica Yada
- Cancer Immunotherapy, Kanagawa Cancer Center, Asahi-ku, Yokohama, Japan
| | - Junya Ohtake
- Cancer Immunotherapy, Kanagawa Cancer Center, Asahi-ku, Yokohama, Japan
| | - Tetsuro Sasada
- Cancer Immunotherapy, Kanagawa Cancer Center, Asahi-ku, Yokohama, Japan
| |
Collapse
|
29
|
Abstract
PURPOSE OF REVIEW We will describe recently discovered smart aptamers with tumor specificity, with an emphasis on targeted delivery of novel therapeutic molecules, cancer-specific biomarkers, and immunotherapy. RECENT FINDINGS The development of cancer-specific aptamers has facilitated targeted delivery of potent therapeutic molecules to cancer cells without harming nontumoral cells. This specificity also makes it possible to discover novel cancer biomarkers. Furthermore, alternative immune-checkpoint blockade aptamers have been developed for combinational immunotherapy. SUMMARY Aptamers selected against cancer cells show cancer specificity, which has great potential for targeting. First, functionalizing targeted aptamers with therapeutic molecule payloads (e.g., small activating RNAs, antimitotic drugs, therapeutic antibodies, and peptides) facilitates successful delivery into cancer cells. This approach greatly improves the therapeutic index by minimizing side-effects in nontumoral cells. Second, cancer-specific proteins have been identified as cancer biomarkers through in-vitro and in-vivo selection, aptamer pull-down assays, and mass spectrometry. These newly discovered biomarkers improve therapeutic intervention and diagnostic specificity. In addition, the development of alternative immune-checkpoint blockade aptamers is suggested for use in combinational immunotherapeutic with current immune blockade regimens, to reduce the resistance and exhaustion of T cells in clinical trials. VIDEO ABSTRACT: http://links.lww.com/COON/A21.
Collapse
|
30
|
Van Tongelen A, Loriot A, De Smet C. Oncogenic roles of DNA hypomethylation through the activation of cancer-germline genes. Cancer Lett 2017; 396:130-137. [DOI: 10.1016/j.canlet.2017.03.029] [Citation(s) in RCA: 84] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2017] [Revised: 03/16/2017] [Accepted: 03/16/2017] [Indexed: 12/19/2022]
|
31
|
Abstract
The melanoma-associated antigen (MAGE) genes are known to be expressed in various kinds of tumors including lung cancer. Although they are studied as targets for immunotherapy and tools for early detection of lung cancer, the correlation between MAGE expression and the prognosis in lung cancer has not been clarified. In this study, we evaluated the relationship between MAGE A1-6 gene expression and the clinical prognosis in lung cancer. Bone marrow aspirations were performed in 60 patients who were diagnosed as lung cancer and underwent lung cancer surgery between 2007 and 2008. Each bone marrow was examined using nested reverse transcription- polymerase chain reaction (RT-PCR) with the MAGE common primer to detect MAGE A1-6. Overall survival rate, disease-free survival rate, recurrence, and distant metastasis were reviewed retrospectively. Survival periods were analyzed using SPSS ver. 20.0. Of the total 60 lung cancer patients, 9 patients (15%) had MAGE A1-6. MAGE A1-6-positive patients showed poor overall survival and overall disease-free survival rates (43.8 ± 26.1, 43.2 ± 26.9 months, respectively) compared with MAGE A1-6-negative patients (54.4 ± 17.2, 44.8 ± 22.1 months, respectively). No significant difference was shown in either survival rates. In conclusion, MAGE A1-6 expression of bone marrow in lung cancer patients correlated with poor survival rates. We suggest that MAGE A1-6 may be considered as a novel prognostic factor for lung cancer which leads to effective follow-up and treatment.
Collapse
Affiliation(s)
- Eunjue Yi
- Department of Thoracic and Cardiovascular Surgery, Seoul National University Bundang Hospital, Seongnam-si, Gyeonggi-do, Republic of Korea
| | - Ji-Eun Chang
- Department of Thoracic and Cardiovascular Surgery, Seoul National University Bundang Hospital, Seongnam-si, Gyeonggi-do, Republic of Korea
| | - Chosun Leem
- Respiratory Center, Seoul National University Bundang Hospital, Seongnam-si, Gyeonggi-do, Republic of Korea
| | - Chang-Ho Jeon
- Department of Laboratory Medicine, Catholic University of Daegu School of Medicine, Daegu, Republic of Korea
| | - Sanghoon Jheon
- Department of Thoracic and Cardiovascular Surgery, Seoul National University Bundang Hospital, Seongnam-si, Gyeonggi-do, Republic of Korea.,Department of Thoracic and Cardiovascular Surgery, Seoul National University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
32
|
Najjar YG, Ding F, Lin Y, VanderWeele R, Butterfield LH, Tarhini AA. Melanoma antigen-specific effector T cell cytokine secretion patterns in patients treated with ipilimumab. J Transl Med 2017; 15:39. [PMID: 28222797 PMCID: PMC5319167 DOI: 10.1186/s12967-017-1140-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Accepted: 02/04/2017] [Indexed: 12/11/2022] Open
Abstract
Background In a previously reported study, patients with regionally advanced melanoma were treated with neoadjuvant ipilimumab (ipi) (Tarhini in PLoS ONE 9(2): e87705, 3). Significant changes in circulating myeloid derived suppressor cells (MDSC), regulatory T cells (Treg) and peptide specific type I CD4+ and CD8+ T cells were noted at week 6 that correlated with clinical outcome. Characterization of antigen-specific effector T cell secreted cytokines may shed insights into ipi associated T cell activation and function. Methods Patients were treated with neoadjuvant ipi (10 mg/kg every 3 weeks ×2) administered intravenously before and after surgery. Peripheral blood mononuclear cells (PBMC) that were collected at baseline and week 6 (after ipi) were tested here. Each sample was divided into 5 groups and stimulated with controls or shared melanoma antigen overlapping peptide pools (NY-ESO 1, gp-100, MART-1). Secreted cytokines, chemokines and growth factors were assessed using Luminex. Cytokine expression levels between the 3 antigen groups were compared using the Wilcox rank-sum test. Results Seventeen cytokines were differentially expressed with stimulation by each antigen at baseline (p value <0.05): IL1β, MIP1β, IL1RA, VEGF, IL13, IL17, MIP1α, GM-CSF, MCP1, IL5, IL2R, IL4, IL10, IFNγ, TNFα, IL8 and IL2. At week 6, 15 cytokines were differentially expressed (p < 0.05): IL1β, VEGF, G-CSF, HGF, IL13, IL17, GM-CSF, MCP1, IL5, IL7, IL4, IL10, IFNγ, IL8 and IL2. Patients were later clustered based on cytokine expression levels at baseline and at week 6, and recurrence free survival (RFS) was compared. Clear differences in RFS were noted based on cytokine level clustering both at baseline and at week 6: Patients whose PBMCs secreted more cytokines in response to NY-ESO-1 showed a trend towards better RFS. Conclusions PBMCs of patients treated with ipi secreted significantly more cytokines, chemokines and growth factors in response to NY-ESO-1 than to gp-100 or MART-1. These cytokines belonged to different functional groups, including inflammatory, type 1, type 2 and regulatory, that warrant further study. Patients whose PBMCs secreted more cytokines (particularly in response to NY-ESO-1) tended to have better RFS, supporting further exploration in terms of therapeutic predictive value. Electronic supplementary material The online version of this article (doi:10.1186/s12967-017-1140-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Yana G Najjar
- University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Fei Ding
- Biostatistics Facility, University of Pittsburgh Cancer Institute, Pittsburgh, PA, USA
| | - Yan Lin
- Biostatistics Facility, University of Pittsburgh Cancer Institute, Pittsburgh, PA, USA
| | | | - Lisa H Butterfield
- University of Pittsburgh, University of Pittsburgh Cancer Institute, Pittsburgh, PA, USA
| | - Ahmad A Tarhini
- Division of Hematology-Oncology, Department of Medicine, University of Pittsburgh, University of Pittsburgh Cancer Institute, 5150 Centre Avenue (555), Pittsburgh, PA, 15232, USA.
| |
Collapse
|
33
|
MAGE-A is More Highly Expressed Than NY-ESO-1 in a Systematic Immunohistochemical Analysis of 3668 Cases. J Immunother 2016; 39:181-7. [PMID: 27070449 DOI: 10.1097/cji.0000000000000119] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Two cancer testis antigens, the New York esophageal squamous cell carcinoma-1 (NY-ESO-1) and the melanoma-antigen family A (MAGE-A), represent promising immunotherapy targets due to the low expression of these antigens in nonmalignant tissue. To assess overexpression patterns in various cancers, we performed a systematic immunohistochemical analysis for NY-ESO-1 and MAGE-A on tissue array samples of 3668 common epithelial carcinomas (CA) and germ cell tumors of high prevalence and mortality. Here, we find significantly higher expression of MAGE-A (>50% on tumor cells) compared with NY-ESO-1 in several CAs including cutaneous squamous cell carcinomas (SCC) (52.8%/2.8%), esophageal SCC (50%/0%), head and neck SCC (41.1%/<1%), bladder urothelial CA (40.4%/8.3%), cervical/anal SCC (37.5%/0%), lung SCC (34%/3.8%), lung adenocarcinomas (27.6%/3.9%), ovarian CA (26.4%/3.6%), endometrial CA (26.3%/1.3%), lung small cell CA (24.4%/2.4%), gastric adenocarcinomas (20%/4%), breast mucinous CA (19.3%/0%), hepatocellular CA (18.8%/1.2%), breast infiltrating ductal CA (16.4%/1.8%), colorectal adenocarcinomas (10.7%/<1%), cholangiocarcinomas (9.8%/0%), thymic CA (9%/4.5%), and mesotheliomas (7.9%/<1%). Furthermore, high expression of MAGE-A, but not NY-ESO-1, was seen in whole slide evaluations of an independent cohort of metastatic SCC (45.5%/3.6%) and metastatic CA (13.5%/0%) of various primaries with significantly higher expression of MAGE-A in metastatic SCC compared with other metastatic CA. MAGE-A is also more highly expressed in germ cell tumors, seminomas (69%/3.5%) and nonseminomas (40.1%/4.7%). In summary, MAGE-A is more highly expressed than NY-ESO-1 in a majority of human malignancies, and targeting MAGE-A may benefit a large number of patients.
Collapse
|
34
|
van der Werff ten Bosch J, van den Akker M. Genetic predisposition and hematopoietic malignancies in children: Primary immunodeficiency. Eur J Med Genet 2016; 59:647-653. [DOI: 10.1016/j.ejmg.2016.03.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2015] [Revised: 02/18/2016] [Accepted: 03/08/2016] [Indexed: 01/24/2023]
|
35
|
Salmaninejad A, Zamani MR, Pourvahedi M, Golchehre Z, Hosseini Bereshneh A, Rezaei N. Cancer/Testis Antigens: Expression, Regulation, Tumor Invasion, and Use in Immunotherapy of Cancers. Immunol Invest 2016; 45:619-40. [DOI: 10.1080/08820139.2016.1197241] [Citation(s) in RCA: 101] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
|
36
|
Bagchi S, Li S, Wang CR. CD1b-autoreactive T cells recognize phospholipid antigens and contribute to antitumor immunity against a CD1b + T cell lymphoma. Oncoimmunology 2016; 5:e1213932. [PMID: 27757307 DOI: 10.1080/2162402x.2016.1213932] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2016] [Revised: 07/08/2016] [Accepted: 07/11/2016] [Indexed: 12/21/2022] Open
Abstract
Adoptive immunotherapy for cancer treatment is an emerging field of study. Till now, several tumor-derived, peptide-specific T cell responses have been harnessed for treating cancers. However, the contribution of lipid-specific T cells in tumor immunity has been understudied. CD1 molecules, which present self- and foreign lipid antigens to T cells, are divided into group 1 (CD1a, CD1b, and CD1c) and group 2 (CD1d). Although the role of CD1d-restricted natural killer T cells (NKT) in several tumor models has been well established, the contribution of group 1 CD1-restricted T cells in tumor immunity remains obscure due to the lack of group 1 CD1 expression in mice. In this study, we used a double transgenic mouse model expressing human group 1 CD1 molecules (hCD1Tg) and a CD1b-restricted, self-lipid reactive T cell receptor (HJ1Tg) to study the potential role of group 1 CD1-restricted autoreactive T cells in antitumor response. We found that HJ1 T cells recognized phospholipids and responded more potently to lipid extracted from tumor cells than the equivalent amount of lipids extracted from normal cells. Additionally, the autoreactivity of HJ1 T cells was enhanced upon treatment with various intracellular toll-like receptor (TLR) agonists, including CpG oligodeoxynucleotides (ODN), R848, and poly (I:C). Interestingly, the adoptive transfer of HJ1 T cells conferred protection against the CD1b-transfected murine T cell lymphoma (RMA-S/CD1b) and CpG ODN enhanced the antitumor effect. Thus, this study, for the first time, demonstrates the antitumor potential of CD1b-autoreactive T cells and their potential use in adoptive immunotherapy.
Collapse
Affiliation(s)
- Sreya Bagchi
- Department of Microbiology and Immunology, Northwestern University , Chicago, IL, USA
| | - Sha Li
- Department of Microbiology and Immunology, Northwestern University, Chicago, IL, USA; Department of Microbiology, Cornell University, Ithaca, NY, USA
| | - Chyung-Ru Wang
- Department of Microbiology and Immunology, Northwestern University , Chicago, IL, USA
| |
Collapse
|
37
|
Tsukahara T, Hirohashi Y, Kanaseki T, Nakatsugawa M, Kubo T, Sato N, Torigoe T. Peptide vaccination therapy: Towards the next generation. Pathol Int 2016; 66:547-553. [PMID: 27435148 DOI: 10.1111/pin.12438] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2016] [Revised: 06/14/2016] [Accepted: 06/24/2016] [Indexed: 12/24/2022]
Affiliation(s)
- Tomohide Tsukahara
- Department of PathologySapporo Medical University School of Medicine South‐1 West‐17, Chuo‐ku Sapporo Japan
| | - Yoshihiko Hirohashi
- Department of PathologySapporo Medical University School of Medicine South‐1 West‐17, Chuo‐ku Sapporo Japan
| | - Takayuki Kanaseki
- Department of PathologySapporo Medical University School of Medicine South‐1 West‐17, Chuo‐ku Sapporo Japan
| | - Munehide Nakatsugawa
- Department of PathologySapporo Medical University School of Medicine South‐1 West‐17, Chuo‐ku Sapporo Japan
| | - Terufumi Kubo
- Department of PathologySapporo Medical University School of Medicine South‐1 West‐17, Chuo‐ku Sapporo Japan
| | - Noriyuki Sato
- Department of PathologySapporo Medical University School of Medicine South‐1 West‐17, Chuo‐ku Sapporo Japan
| | - Toshihiko Torigoe
- Department of PathologySapporo Medical University School of Medicine South‐1 West‐17, Chuo‐ku Sapporo Japan
| |
Collapse
|
38
|
Zoledronic acid induces dose-dependent increase of antigen-specific CD8 T-cell responses in combination with peptide/poly-IC vaccine. Vaccine 2016; 34:1275-81. [PMID: 26828454 DOI: 10.1016/j.vaccine.2016.01.026] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2015] [Revised: 01/06/2016] [Accepted: 01/17/2016] [Indexed: 01/26/2023]
Abstract
Zoledronic acid (ZA) is used for treating osteoporosis and for preventing skeletal fractures in cancer patients suffering from myeloma and prostate cancer. It is also reported to directly induce cancer cell apoptosis and indirectly modulate T-cell immune response as an antitumor agent. In this study, the effect of ZA following peptide/polyinosinic-polycytidylic acid (poly-IC) vaccination was investigated in a murine tumor model. The combination of ZA with peptide/poly-IC vaccine showed a synergistic effect on the induction of antigen-specific CD8 T-cell response. Three consecutive intravenous administrations of ZA was defined to induce the highest CD8 T-cell response. Further, total splenocyte counts and antigen-specific CD8 T-cell response gradually increased depending on the dose of ZA. In tumor-bearing mice, ZA showed a dose-dependent decrease of growth and prolonged survival. Treatment with ZA only decreased the number of CD11b(+)Gr1(+) myeloid cells in blood. Our results demonstrate that the use of ZA could improve antitumor immune responses induced by the peptide/poly-IC vaccine.
Collapse
|
39
|
Improving Multi-Epitope Long Peptide Vaccine Potency by Using a Strategy that Enhances CD4+ T Help in BALB/c Mice. PLoS One 2015; 10:e0142563. [PMID: 26556756 PMCID: PMC4640540 DOI: 10.1371/journal.pone.0142563] [Citation(s) in RCA: 119] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2015] [Accepted: 10/25/2015] [Indexed: 11/19/2022] Open
Abstract
Peptide-based vaccines are attractive approaches for cancer immunotherapy; but the success of these vaccines in clinical trials have been limited. Our goal is to improve immune responses and anti-tumor effects against a synthetic, multi-epitope, long peptide from rat Her2/neu (rHer2/neu) using the help of CD4+ T cells and appropriate adjuvant in a mouse tumor model. Female BALB/c mice were vaccinated with P5+435 multi-epitope long peptide that presents epitopes for cytotoxic T lymphocytes (CTL) in combination with a universal Pan DR epitope (PADRE) or CpG-oligodeoxynucleotides (CpG-ODNs) as a Toll-like receptor agonist adjuvant. The results show that vaccination with the multi-epitope long peptide in combination with the PADRE peptide and CpG-ODN induced expansion of subpopulations of CD4+ and CD8+ cells producing IFN-γ, the average tumor size in the vaccinated mice was less than that of the other groups, and tumor growth was inhibited in 40% of the mice in the vaccinated group. The mean survival time was 82.6 ± 1.25 days in mice vaccinated with P5+435 + CpG+ PADRE. Our results demonstrate that inclusion of PADRE and CpG with the peptide vaccine enhanced significant tumor specific-immune responses in vaccinated mice.
Collapse
|
40
|
Cannuyer J, Van Tongelen A, Loriot A, De Smet C. A gene expression signature identifying transient DNMT1 depletion as a causal factor of cancer-germline gene activation in melanoma. Clin Epigenetics 2015; 7:114. [PMID: 26504497 PMCID: PMC4620642 DOI: 10.1186/s13148-015-0147-4] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2015] [Accepted: 10/05/2015] [Indexed: 12/31/2022] Open
Abstract
Background Many human tumors show aberrant activation of a group of germline-specific genes, termed cancer-germline (CG) genes, several of which appear to exert oncogenic functions. Although activation of CG genes in tumors has been linked to promoter DNA demethylation, the mechanisms underlying this epigenetic alteration remain unclear. Two main processes have been proposed: awaking of a gametogenic program directing demethylation of target DNA sequences via specific regulators, or general deficiency of DNA methylation activities resulting from mis-targeting or down-regulation of the DNMT1 methyltransferase. Results By the analysis of transcriptomic data, we searched to identify gene expression changes associated with CG gene activation in melanoma cells. We found no evidence linking CG gene activation with differential expression of gametogenic regulators. Instead, CG gene activation correlated with decreased expression of a set of mitosis/division-related genes (ICCG genes). Interestingly, a similar gene expression signature was previously associated with depletion of DNMT1. Consistently, analysis of a large set of melanoma tissues revealed that DNMT1 expression levels were often lower in samples showing activation of multiple CG genes. Moreover, by using immortalized melanocytes and fibroblasts carrying an inducible anti-DNMT1 small hairpin RNA (shRNA), we demonstrate that transient depletion of DNMT1 can lead to long-term activation of CG genes and repression of ICCG genes at the same time. For one of the ICCG genes (CDCA7L), we found that its down-regulation in melanoma cells was associated with deposition of repressive chromatin marks, including H3K27me3. Conclusions Together, our observations point towards transient DNMT1 depletion as a causal factor of CG gene activation in vivo in melanoma. Electronic supplementary material The online version of this article (doi:10.1186/s13148-015-0147-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Julie Cannuyer
- Group of Genetics and Epigenetics, de Duve Institute, Université Catholique de Louvain, Brussels, Belgium
| | - Aurélie Van Tongelen
- Group of Genetics and Epigenetics, de Duve Institute, Université Catholique de Louvain, Brussels, Belgium
| | - Axelle Loriot
- Group of Genetics and Epigenetics, de Duve Institute, Université Catholique de Louvain, Brussels, Belgium
| | - Charles De Smet
- Group of Genetics and Epigenetics, de Duve Institute, Université Catholique de Louvain, Brussels, Belgium
| |
Collapse
|
41
|
Abstract
DNA vaccination has emerged as an attractive immunotherapeutic approach against cancer
due to its simplicity, stability, and safety. Results from numerous clinical trials have
demonstrated that DNA vaccines are well tolerated by patients and do not trigger major
adverse effects. DNA vaccines are also very cost effective and can be administered
repeatedly for long-term protection. Despite all the practical advantages, DNA vaccines
face challenges in inducing potent antigen specific cellular immune responses as a result
of immune tolerance against endogenous self-antigens in tumors. Strategies to enhance
immunogenicity of DNA vaccines against self-antigens have been investigated including
encoding of xenogeneic versions of antigens, fusion of antigens to molecules that activate
T cells or trigger associative recognition, priming with DNA vectors followed by boosting
with viral vector, and utilization of immunomodulatory molecules. This review will focus
on discussing strategies that circumvent immune tolerance and provide updates on findings
from recent clinical trials.
Collapse
Key Words
- APCs, antigen presenting cells
- CEA, carcinoembryonic antigen
- CIN, cervical intraepithelial neoplasia
- CT antigens, cancer-testis antigens
- CTLs, cytotoxic lymphocytes
- DNA vaccines
- DOM, fragment c domain
- EP, electroporation
- GITR, glucocorticoid-induced tumor necrosis factor receptor family-related genes
- HER2, Her2/neu
- HSP70, heat shock protein 70
- IFNs, interferons
- IRF, interferon regulatory factor
- Id, idiotype
- MHC, major histocompatibility complex
- Mam-A, Mammaglobin-A
- NHP, non-human primate
- PAP, Prostatic acid phosphatase
- PMED, particle mediated epidermal delivery
- PSMA, prostate-specific membrane antigen
- SCT, single-chain trimer
- STING, stimulator of interferon genes
- TAAs, tumor-associated antigens
- TBK1, Tank-binding kinase 1
- TLRs, Toll-like receptors
- TT, tetanus toxin
- Trp2, tyrosinase related protein 2
- cellular immune response
- hTERT, human telomerase reverse transcriptase
- humoral immune response
- immune tolerance
- phTERT, optimized full-length hTERT
- tumor antigens
- vaccine delivery
Collapse
Affiliation(s)
- Benjamin Yang
- a Department of Pathology ; Johns Hopkins University ; Baltimore , MD USA
| | | | | | | | | |
Collapse
|
42
|
Wang CY, Lin BL, Chen CH. An aptamer targeting shared tumor-specific peptide antigen of MAGE-A3 in multiple cancers. Int J Cancer 2015; 138:918-26. [PMID: 26314689 DOI: 10.1002/ijc.29826] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2015] [Accepted: 08/14/2015] [Indexed: 01/15/2023]
Abstract
A DNA aptamer was identified against the shared tumor-specific MAGE-A3111-125 peptide antigen. The dissociation constant between the aptamer and the peptide was measured at 57 nM. Binding of the aptamer to seven types of cancer cells, melanoma, breast, colorectal, liver, lung, pancreas and oral cancer, was confirmed with flow cytometry and fluorescence imaging. Cy3-conjugated aptamers signals were specifically localized to the surface of those cancer cells. The results indicate that the DNA aptamer against the shared tumor-specific MAGE-A3 peptide can be used in cancer cell targeting and has the potential for developing into new modalities for the diagnosis of multiple cancers.
Collapse
Affiliation(s)
- Chin-Yu Wang
- Genomics Research Center, Academia Sinica, Taipei, Taiwan
| | - Bai-Ling Lin
- Genomics Research Center, Academia Sinica, Taipei, Taiwan
| | | |
Collapse
|
43
|
[Radiation therapy and immunomodulation: Focus on experimental data]. Cancer Radiother 2015; 19:515-8. [PMID: 26293415 DOI: 10.1016/j.canrad.2015.05.018] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2015] [Accepted: 05/20/2015] [Indexed: 12/13/2022]
Abstract
The immunosuppressive effects of radiation therapy have long been the only ones considered. It has been demonstrated that exposure to ionizing radiation induces the release of tumour antigens which activates both the innate immune system and the adaptive immune response of the host. The purpose of tumour immunotherapy is based on the principle that reversal of tolerance to immunogenic tumours would be able to activate an immune response against tumour cells. Preclinical data and clinical studies early phase suggest a potential therapeutic benefit of immunotherapy combined with radiation therapy. The objective of this article is to review how tumour cells interact with the immune system and how ionizing radiation modulate this interaction and finally the combination of perspectives of immunotherapy and ionizing radiation by focusing on existing clinical data.
Collapse
|
44
|
Fortes C, Mastroeni S, Mannooranparampil TJ, Passarelli F, Zappalà A, Annessi G, Marino C, Caggiati A, Russo N, Michelozzi P. Tumor-infiltrating lymphocytes predict cutaneous melanoma survival. Melanoma Res 2015; 25:306-11. [PMID: 25933208 DOI: 10.1097/cmr.0000000000000164] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Understanding differences in survival across distinct subgroups of melanoma patients may help with the choice of types of therapy. Tumor-infiltrating lymphocytes (TILs) are considered a manifestation of the host immune response to tumor, but the role of TILs in melanoma mortality is controversial. The aim of this study was to investigate independent prognostic factors for melanoma mortality. We carried out a 10-year cohort study on 4133 melanoma patients from the same geographic area (Lazio) with primary cutaneous melanoma diagnosed between January 1998 and December 2008. The probability of survival was estimated using Kaplan-Meier methods and prognostic factors were evaluated by multivariate analysis (Cox proportional hazards model). The 10-year survival rate for melanoma decreased with increasing Breslow thickness (Pfor trend<0.0001) and with age (Pfor trend<0.0001) whereas survival increased with increasing levels of TILs (Pfor trend=0.0001). The 10-year survival rate for melanoma divided into TILs intensity as scanty, moderate, and marked was 88.0, 92.2, and 97.0%, respectively. In the multivariate Cox model, the presence of high levels of TILs in primary invasive melanomas was associated with a lower risk of melanoma death (hazard ratio 0.32; 95% confidence interval 0.13-0.82) after controlling for sex, age, Breslow thickness, histological type, mitotic rate, and ulceration. After including lymph node status in the multivariate analysis, the protective effect of marked TILs on melanoma mortality remained (hazard ratio 0.37; 95% confidence interval 0.15-0.94). The results of this study suggest that the immune microenvironment affects melanoma survival.
Collapse
Affiliation(s)
- Cristina Fortes
- aEpidemiology Unit bPathology Unit cOncology Unit dSurgery Department of Plastic Surgery eMedical Direction, IDI-IRCCS fDepartment of Epidemiology of the Regional Health Service, Rome, Italy
| | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
de Aquino MTP, Malhotra A, Mishra MK, Shanker A. Challenges and future perspectives of T cell immunotherapy in cancer. Immunol Lett 2015; 166:117-33. [PMID: 26096822 PMCID: PMC4499494 DOI: 10.1016/j.imlet.2015.05.018] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2015] [Revised: 05/10/2015] [Accepted: 05/27/2015] [Indexed: 12/15/2022]
Abstract
Since the formulation of the tumour immunosurveillance theory, considerable focus has been on enhancing the effectiveness of host antitumour immunity, particularly with respect to T cells. A cancer evades or alters the host immune response by various ways to ensure its development and survival. These include modifications of the immune cell metabolism and T cell signalling. An inhibitory cytokine milieu in the tumour microenvironment also leads to immune suppression and tumour progression within a host. This review traces the development in the field and attempts to summarize the hurdles that the approach of adoptive T cell immunotherapy against cancer faces, and discusses the conditions that must be improved to allow effective eradication of cancer.
Collapse
Affiliation(s)
- Maria Teresa P de Aquino
- Department of Biochemistry and Cancer Biology, School of Medicine, Meharry Medical College, Nashville, TN 37208, USA
| | - Anshu Malhotra
- Department of Biochemistry and Cancer Biology, School of Medicine, Meharry Medical College, Nashville, TN 37208, USA
| | - Manoj K Mishra
- Department of Biological Sciences, Alabama State University, Montgomery, AL 36101, USA
| | - Anil Shanker
- Department of Biochemistry and Cancer Biology, School of Medicine, Meharry Medical College, Nashville, TN 37208, USA; Tumor-Host Interactions Research Program, Vanderbilt-Ingram Cancer Center, Vanderbilt University, Nashville, TN 37232, USA.
| |
Collapse
|
46
|
Gupta SK, Jaitly T, Schmitz U, Schuler G, Wolkenhauer O, Vera J. Personalized cancer immunotherapy using Systems Medicine approaches. Brief Bioinform 2015; 17:453-67. [DOI: 10.1093/bib/bbv046] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2015] [Indexed: 12/27/2022] Open
|
47
|
Abstract
A number of consensuses regarding cancer immunology have recently emerged from both preclinical immunotherapy models and analysis of cancer patients. First and foremost, the natural state of endogenous tumor reactive T cells is characterized by general hyporesponsiveness or anergy. This is likely due to a number of mechanisms that tumors use to induce tolerance as they develop. While many of the newer generation vaccines can effectively transfer antigen to and activate dendritic cells, T-cell tolerance remains a major barrier that is difficult to overcome by vaccination alone. Preclinical models demonstrate that for poorly immunogenic tumors, once tolerance has been established, therapeutic vaccines alone are ineffective at curing animals with a significant established tumor burden. However, combination strategies of vaccination together with inhibitors of immunologic checkpoints and agonists for co-stimulatory pathways are proving capable of overcoming tolerance and generating significant anti-tumor responses even in cases of established metastatic cancer.
Collapse
Affiliation(s)
- Drew Pardoll
- Department of Oncology and Director Cancer Immunology Program, Johns Hopkins University School of Medicine and Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD.
| |
Collapse
|
48
|
Proteasome subtypes and regulators in the processing of antigenic peptides presented by class I molecules of the major histocompatibility complex. Biomolecules 2014; 4:994-1025. [PMID: 25412285 PMCID: PMC4279167 DOI: 10.3390/biom4040994] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2014] [Revised: 10/02/2014] [Accepted: 10/29/2014] [Indexed: 02/07/2023] Open
Abstract
The proteasome is responsible for the breakdown of cellular proteins. Proteins targeted for degradation are allowed inside the proteasome particle, where they are cleaved into small peptides and released in the cytosol to be degraded into amino acids. In vertebrates, some of these peptides escape degradation in the cytosol, are loaded onto class I molecules of the major histocompatibility complex (MHC) and displayed at the cell surface for scrutiny by the immune system. The proteasome therefore plays a key role for the immune system: it provides a continued sampling of intracellular proteins, so that CD8-positive T-lymphocytes can kill cells expressing viral or tumoral proteins. Consequently, the repertoire of peptides displayed by MHC class I molecules at the cell surface depends on proteasome activity, which may vary according to the presence of proteasome subtypes and regulators. Besides standard proteasomes, cells may contain immunoproteasomes, intermediate proteasomes and thymoproteasomes. Cells may also contain regulators of proteasome activity, such as the 19S, PA28 and PA200 regulators. Here, we review the effects of these proteasome subtypes and regulators on the production of antigenic peptides. We also discuss an unexpected function of the proteasome discovered through the study of antigenic peptides: its ability to splice peptides.
Collapse
|
49
|
Destexhe E, Grosdidier E, Baudson N, Forster R, Gerard C, Garçon N, Segal L. Non‐clinical safety evaluation of single and repeated intramuscular administrations of MAGE‐A3 Cancer Immunotherapeutic in rabbits and cynomolgus monkeys. J Appl Toxicol 2014; 35:717-28. [DOI: 10.1002/jat.3025] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2014] [Revised: 03/14/2014] [Accepted: 04/02/2014] [Indexed: 12/24/2022]
Affiliation(s)
- Eric Destexhe
- GlaxoSmithKline Vaccines 1330 Rixensart/Wavre Belgium
| | | | | | | | | | | | | |
Collapse
|
50
|
Benteyn D, Heirman C, Bonehill A, Thielemans K, Breckpot K. mRNA-based dendritic cell vaccines. Expert Rev Vaccines 2014; 14:161-76. [PMID: 25196947 DOI: 10.1586/14760584.2014.957684] [Citation(s) in RCA: 113] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Cancer immunotherapy has been proposed as a powerful treatment modality. Active immunotherapy aspires to stimulate the patient's immune system, particularly T cells. These cells can recognize and kill cancer cells and can form an immunological memory. Dendritic cells (DCs) are the professional antigen-presenting cells of our immune system. They take up and process antigens to present them to T cells. Consequently, DCs have been investigated as a means to stimulate cancer-specific T-cell responses. An efficient strategy to program DCs is the use of mRNA, a well-defined and safe molecule that can be easily generated at high purity. Importantly, vaccines consisting of mRNA-modified DCs showed promising results in clinical trials. Therefore, we will introduce cancer immunotherapy and DCs and give a detailed overview on the application of mRNA to generate cancer-fighting DC vaccines.
Collapse
Affiliation(s)
- Daphné Benteyn
- Laboratory of Molecular and Cellular Therapy, Department of Biomedical Sciences, Vrije Universiteit Brussel, Laarbeeklaan 103/E, 1090 Jette, Belgium
| | | | | | | | | |
Collapse
|