1
|
Liang C, Geng L, Dong Y, Zhang H. VEGF165b mutant can be used as a protein carrier to form a chimeric tumor vaccine with Mucin1 peptide to elicit an anti-tumor response. Mol Immunol 2024; 175:31-39. [PMID: 39298996 DOI: 10.1016/j.molimm.2024.09.009] [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: 08/05/2023] [Revised: 09/15/2024] [Accepted: 09/16/2024] [Indexed: 09/22/2024]
Abstract
Peptide-based anticancer vaccines have shown some efficacy in generating cancer-specific immune responses in various cancer studies, but clinical success is limited, one of the reasons is due to its prone degradation and weak immunogenicity. So some tumor epitope peptide vaccines often require coupling or forming fusion proteins with corresponding protein carriers to enhance their stability and immunogenicity. Given the scarcity of validated carriers for clinical trials, there is an urgent requirement for the development of novel protein carrier. Our previous work has demonstrated that VEGF165b mutant could be used as an effective immunization adjunct to enhance anti-tumor immune response. By analyzing and evaluating the gene structure of VEGF, we speculated that mVEGF165b has the potential to be utilized as a tumor peptide vaccine carrier. An mVEGF165b-MUC1 chimeric tumor vaccine was produced by fusing the MUC1 peptide ((MUC1, a T-cell epitope dominant peptide from Mucin1) to the C-terminus of mVEGF165b, expressing the fusing protein in pichia yeast, followed by purification with a HiTrap heparin affinity chromatography column. We found that immunizing mice with mVEGF165b-MUC1 fusion protein induced high-titer antibodies against VEGF in a preventive context, which in turn reduced the proportion of Tregs and further stimulated mice to produce T-cell responses specific to mucin1. The high-titer VEGF antibody stimulated by mVEGF165b also promoted tumor blood vessel maturation and facilitated T-cell infiltration. In conclusion,immunized with mVEGF165b-MUC1 protein are beneficial for eliciting immune responses targeting Mucin1, mVEGF165b have the potential to be utilized as a peptide tumor vaccine carrier.
Collapse
Affiliation(s)
- Chen Liang
- Synthetic Biology Engineering Lab of Henan Province, School of Life Sciences and Technology, Xinxiang Medical University, Xinxiang, Henan Province 453003, PR China
| | - Lujing Geng
- Institute of Applied Neurosciences, School of Life Sciences and Technology, Xinxiang Medical University, Xinxiang, Henan Province 453003, PR China
| | - Yifan Dong
- Institute of Applied Neurosciences, School of Life Sciences and Technology, Xinxiang Medical University, Xinxiang, Henan Province 453003, PR China
| | - Huiyong Zhang
- Synthetic Biology Engineering Lab of Henan Province, School of Life Sciences and Technology, Xinxiang Medical University, Xinxiang, Henan Province 453003, PR China.
| |
Collapse
|
2
|
Abdollahi P, Norseth HM, Schjesvold F. Advances and challenges in anti-cancer vaccines for multiple myeloma. Front Immunol 2024; 15:1411352. [PMID: 39161773 PMCID: PMC11331005 DOI: 10.3389/fimmu.2024.1411352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Accepted: 07/17/2024] [Indexed: 08/21/2024] Open
Abstract
Multiple myeloma (MM) is a hematological cancer marked by plasma cell accumulation in the bone marrow. Despite treatment advancements, MM remains incurable in most patients. MM-associated immune dysregulation fosters disease progression, prompting research into immunotherapy to combat the disease. An area of immunotherapy investigation is the design of myeloma vaccine therapy to reverse tumor-associated immune suppression and elicit tumor-specific immune responses to effectively target MM cells. This article reviews vaccine immunotherapy for MM, categorizing findings by antigen type and delivery method. Antigens include idiotype (Id), tumor-associated (TAA), tumor-specific (TSA), and whole tumor lysate. Myeloma vaccination has so far shown limited clinical efficacy. However, further studies are essential to optimize various aspects, including antigen and patient selection, vaccine timing and sequencing, and rational combinations with emerging MM treatments.
Collapse
Affiliation(s)
- Pegah Abdollahi
- Oslo Myeloma Center, Department of Hematology, Oslo University Hospital, Oslo, Norway
| | | | | |
Collapse
|
3
|
Jin W, Zhang M, Dong C, Huang L, Luo Q. The multifaceted role of MUC1 in tumor therapy resistance. Clin Exp Med 2023; 23:1441-1474. [PMID: 36564679 DOI: 10.1007/s10238-022-00978-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Accepted: 12/10/2022] [Indexed: 12/24/2022]
Abstract
Tumor therapeutic resistances are frequently linked to the recurrence and poor prognosis of cancers and have been a key bottleneck in clinical tumor treatment. Mucin1 (MUC1), a heterodimeric transmembrane glycoprotein, exhibits abnormally overexpression in a variety of human tumors and has been confirmed to be related to the formation of therapeutic resistance. In this review, the multifaceted roles of MUC1 in tumor therapy resistance are summarized from aspects of pan-cancer principles shared among therapies and individual mechanisms dependent on different therapies. Concretely, the common mechanisms of therapy resistance across cancers include interfering with gene expression, promoting genome instability, modifying tumor microenvironment, enhancing cancer heterogeneity and stemness, and activating evasion and metastasis. Moreover, the individual mechanisms of therapy resistance in chemotherapy, radiotherapy, and biotherapy are introduced. Last but not least, MUC1-involved therapy resistance in different types of cancers and MUC1-related clinical trials are summarized.
Collapse
Affiliation(s)
- Weiqiu Jin
- Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, 200025, China
- Department of Histoembryology, Genetics and Developmental Biology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Key Laboratory of Reproductive Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Mengwei Zhang
- Department of Histoembryology, Genetics and Developmental Biology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Key Laboratory of Reproductive Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Department of Liver Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Changzi Dong
- Department of Bioengineering, School of Engineering and Science, University of Pennsylvania, Philadelphia, 19104, USA
| | - Lei Huang
- Department of Histoembryology, Genetics and Developmental Biology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Key Laboratory of Reproductive Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
- Innovative Research Team of High-Level Local Universities in Shanghai, Shanghai, China.
| | - Qingquan Luo
- Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, 200025, China.
| |
Collapse
|
4
|
Liu Z, Yang C, Liu X, Xu X, Zhao X, Fu R. Therapeutic strategies to enhance immune response induced by multiple myeloma cells. Front Immunol 2023; 14:1169541. [PMID: 37275861 PMCID: PMC10232766 DOI: 10.3389/fimmu.2023.1169541] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2023] [Accepted: 05/08/2023] [Indexed: 06/07/2023] Open
Abstract
Multiple myeloma (MM)as a haematological malignancy is still incurable. In addition to the presence of somatic genetic mutations in myeloma patients, the presence of immunosuppressive microenvironment greatly affects the outcome of treatment. Although the discovery of immunotherapy makes it possible to break the risk of high toxicity and side effects of traditional chemotherapeutic drugs, there are still obstacles of ineffective treatment or disease recurrence. In this review, we discuss therapeutic strategies to further enhance the specific anti-tumor immune response by activating the immunogenicity of MM cells themselves. New ideas for future myeloma therapeutic approaches are provided.
Collapse
|
5
|
Lanier OL, Pérez-Herrero E, Andrea APD, Bahrami K, Lee E, Ward DM, Ayala-Suárez N, Rodríguez-Méndez SM, Peppas NA. Immunotherapy approaches for hematological cancers. iScience 2022; 25:105326. [PMID: 36325064 PMCID: PMC9619355 DOI: 10.1016/j.isci.2022.105326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Hematological cancers such as leukemia, lymphoma, and multiple myeloma have traditionally been treated with chemo and radiotherapy approaches. Introduction of immunotherapies for treatment of these diseases has led to patient remissions that would not have been possible with traditional approaches. In this critical review we identify main disease characteristics, symptoms, and current treatment options. Five common immunotherapies, namely checkpoint inhibitors, vaccines, cell-based therapies, antibodies, and oncolytic viruses, are described, and their applications in hematological cancers are critically discussed.
Collapse
Affiliation(s)
- Olivia L. Lanier
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, USA
- Institute for Biomaterials, Drug Delivery, and Regenerative Medicine, The University of Texas at Austin, Austin, TX, USA
| | - Edgar Pérez-Herrero
- Departamento de Ingeniería Química y Tecnología Farmacéutica, Universidad de La Laguna, La Laguna, 38206 Tenerife, Spain
- Instituto Universitario de Bio-Orgánica Antonio González, Universidad de La Laguna, La Laguna, 38206 Tenerife, Spain
- Instituto Universitario de Tecnologías Biomédicas, Universidad de La Laguna, La Laguna, 38200 Tenerife, Spain
| | - Abielle P. D.’ Andrea
- Department of Chemical Engineering, The University of Texas at Austin, Austin, TX, USA
- Institute for Biomaterials, Drug Delivery, and Regenerative Medicine, The University of Texas at Austin, Austin, TX, USA
| | - Kiana Bahrami
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, USA
- Institute for Biomaterials, Drug Delivery, and Regenerative Medicine, The University of Texas at Austin, Austin, TX, USA
| | - Elaine Lee
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, USA
- Institute for Biomaterials, Drug Delivery, and Regenerative Medicine, The University of Texas at Austin, Austin, TX, USA
| | - Deidra M. Ward
- Department of Chemical Engineering, The University of Texas at Austin, Austin, TX, USA
- Institute for Biomaterials, Drug Delivery, and Regenerative Medicine, The University of Texas at Austin, Austin, TX, USA
| | - Nilaya Ayala-Suárez
- Instituto Universitario de Bio-Orgánica Antonio González, Universidad de La Laguna, La Laguna, 38206 Tenerife, Spain
| | - Sheyla M. Rodríguez-Méndez
- Instituto Universitario de Bio-Orgánica Antonio González, Universidad de La Laguna, La Laguna, 38206 Tenerife, Spain
| | - Nicholas A. Peppas
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, USA
- Department of Chemical Engineering, The University of Texas at Austin, Austin, TX, USA
- Institute for Biomaterials, Drug Delivery, and Regenerative Medicine, The University of Texas at Austin, Austin, TX, USA
- Division of Molecular Pharmaceutics and Drug Delivery, College of Pharmacy, The University of Texas at Austin, Austin, TX, USA
- Department of Pediatrics, Dell Medical School, The University of Texas at Austin, Austin, TX, USA
- Department of Surgery and Perioperative Care, Dell Medical School, The University of Texas at Austin, Austin, TX, USA
| |
Collapse
|
6
|
Boussi LS, Avigan ZM, Rosenblatt J. Immunotherapy for the treatment of multiple myeloma. Front Immunol 2022; 13:1027385. [PMID: 36389674 PMCID: PMC9649817 DOI: 10.3389/fimmu.2022.1027385] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Accepted: 10/12/2022] [Indexed: 12/05/2022] Open
Abstract
Despite advances in treatment for multiple myeloma, the majority of patients ultimately develop relapsed disease marked by immune evasion and resistance to standard therapy. Immunotherapy has emerged as a powerful tool for tumor-directed cytotoxicity with the unique potential to induce immune memory to reduce the risk of relapse. Understanding the specific mechanisms of immune dysregulation and dysfunction in advanced myeloma is critical to the development of further therapies that produce a durable response. Adoptive cellular therapy, most strikingly CAR T cell therapy, has demonstrated dramatic responses in the setting of refractory disease. Understanding the factors that contribute to immune evasion and the mechanisms of response and resistance to therapy will be critical to developing the next generation of adoptive cellular therapies, informing novel combination therapy, and determining the optimal time to incorporate immune therapy in the treatment of myeloma.
Collapse
Affiliation(s)
- Leora S. Boussi
- Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA, United States
| | - Zachary M. Avigan
- Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA, United States
| | - Jacalyn Rosenblatt
- Division of Hematology, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA, United States
| |
Collapse
|
7
|
Dima D, Jiang D, Singh DJ, Hasipek M, Shah HS, Ullah F, Khouri J, Maciejewski JP, Jha BK. Multiple Myeloma Therapy: Emerging Trends and Challenges. Cancers (Basel) 2022; 14:cancers14174082. [PMID: 36077618 PMCID: PMC9454959 DOI: 10.3390/cancers14174082] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 08/16/2022] [Accepted: 08/18/2022] [Indexed: 11/16/2022] Open
Abstract
Multiple myeloma (MM) is a complex hematologic malignancy characterized by the uncontrolled proliferation of clonal plasma cells in the bone marrow that secrete large amounts of immunoglobulins and other non-functional proteins. Despite decades of progress and several landmark therapeutic advancements, MM remains incurable in most cases. Standard of care frontline therapies have limited durable efficacy, with the majority of patients eventually relapsing, either early or later. Induced drug resistance via up-modulations of signaling cascades that circumvent the effect of drugs and the emergence of genetically heterogeneous sub-clones are the major causes of the relapsed-refractory state of MM. Cytopenias from cumulative treatment toxicity and disease refractoriness limit therapeutic options, hence creating an urgent need for innovative approaches effective against highly heterogeneous myeloma cell populations. Here, we present a comprehensive overview of the current and future treatment paradigm of MM, and highlight the gaps in therapeutic translations of recent advances in targeted therapy and immunotherapy. We also discuss the therapeutic potential of emerging preclinical research in multiple myeloma.
Collapse
Affiliation(s)
- Danai Dima
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH 44195, USA
- Center for Immunotherapy and Precision Immuno-Oncology, Lerner Research Institute, Cleveland, OH 44195, USA
- Department of Hematology and Medical Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Dongxu Jiang
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH 44195, USA
- Center for Immunotherapy and Precision Immuno-Oncology, Lerner Research Institute, Cleveland, OH 44195, USA
| | - Divya Jyoti Singh
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH 44195, USA
- Center for Immunotherapy and Precision Immuno-Oncology, Lerner Research Institute, Cleveland, OH 44195, USA
| | - Metis Hasipek
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Haikoo S. Shah
- Department of Hematology and Medical Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Fauzia Ullah
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Jack Khouri
- Department of Hematology and Medical Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH 44195, USA
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH 44106, USA
- Cleveland Clinic Lerner College of Medicine, Cleveland, OH 44195, USA
| | - Jaroslaw P. Maciejewski
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH 44195, USA
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH 44106, USA
- Cleveland Clinic Lerner College of Medicine, Cleveland, OH 44195, USA
| | - Babal K. Jha
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH 44195, USA
- Center for Immunotherapy and Precision Immuno-Oncology, Lerner Research Institute, Cleveland, OH 44195, USA
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH 44106, USA
- Cleveland Clinic Lerner College of Medicine, Cleveland, OH 44195, USA
- Correspondence:
| |
Collapse
|
8
|
Odales J, Servín-Blanco R, Martínez-Cortés F, Guzman Valle J, Domínguez-Romero AN, Gevorkian G, Manoutcharian K. Antitumor efficacy of MUC1-derived variable epitope library treatments in a mouse model of breast cancer. Vaccine 2022; 40:4796-4805. [PMID: 35788294 DOI: 10.1016/j.vaccine.2022.06.062] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2021] [Revised: 04/13/2022] [Accepted: 06/21/2022] [Indexed: 11/15/2022]
Abstract
The identification of novel targets for cancer immunotherapy and the development of new vaccine immunogens are subjects of permanent interest. MUC1 is an overexpressed antigen found in most tumors, and its overexpression correlates with poor prognosis. Many attempts to direct the immune response against MUC1 in tumor cells have failed, including several clinical trials. We have previously developed an innovative Variable Epitope Library (VEL) vaccine platform that carries massively substituted mutant variants of defined epitopes or epitope regions as an alternative to using wild-type peptide sequences-based immunogens. Here, two murine MUC1-derived epitopes equivalent to the previously tested in cancer immunotherapy human MUC1 regions were used to generate VELs. We observed that vaccination with the 23L VEL immunogens, encompassing the entire signal peptide region of MUC1, reduces the tumor area compared to the wild-type sequence treatment. Contrastingly, vaccination with the MUC1 signal peptide-derived predicted CD8++ T cell epitope-based VEL, 9MUC1spL, showed similar tumor area reduction as the wild-type treatment; however, a decrease in lung metastasis after 9MUC1spL treatment was observed. In addition, vaccination induced a large pool of CD8+ T cells which recognized most variant epitopes from 9MUC1spL. Also, we generated MUC1 variable number tandem repeat (VNTR)-based VELs that reduced the metastatic burden when dendritic cells and M13 recombinant bacteriophages were used as vaccine carriers. Collectively, our data demonstrate the immunogenic and antitumor properties of MUC1 signal peptide- and VNTR-derived VEL immunogens.
Collapse
Affiliation(s)
- Josué Odales
- Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México (UNAM), AP 70228, Ciudad Universitaria, México DF 04510, MÉXICO
| | - Rodolfo Servín-Blanco
- Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México (UNAM), AP 70228, Ciudad Universitaria, México DF 04510, MÉXICO
| | - Fernando Martínez-Cortés
- Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México (UNAM), AP 70228, Ciudad Universitaria, México DF 04510, MÉXICO
| | - Jesus Guzman Valle
- Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México (UNAM), AP 70228, Ciudad Universitaria, México DF 04510, MÉXICO
| | - Allan Noé Domínguez-Romero
- Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México (UNAM), AP 70228, Ciudad Universitaria, México DF 04510, MÉXICO
| | - Goar Gevorkian
- Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México (UNAM), AP 70228, Ciudad Universitaria, México DF 04510, MÉXICO
| | - Karen Manoutcharian
- Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México (UNAM), AP 70228, Ciudad Universitaria, México DF 04510, MÉXICO.
| |
Collapse
|
9
|
Rekad Z, Izzi V, Lamba R, Ciais D, Van Obberghen-Schilling E. The Alternative Matrisome: alternative splicing of ECM proteins in development, homeostasis and tumor progression. Matrix Biol 2022; 111:26-52. [DOI: 10.1016/j.matbio.2022.05.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2022] [Revised: 04/19/2022] [Accepted: 05/04/2022] [Indexed: 12/14/2022]
|
10
|
Pritam M, Singh G, Kumar R, Singh SP. Screening of potential antigens from whole proteome and development of multi-epitope vaccine against Rhizopus delemar using immunoinformatics approaches. J Biomol Struct Dyn 2022; 41:2118-2145. [PMID: 35067195 DOI: 10.1080/07391102.2022.2028676] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Mucormycosis is a deadly fungal disease mainly caused by Rhizopus oryzae (strain 99-880), also known as Rhizopus delemar. Previously, mucormycosis occurs in immunocompromised patients of diabetes mellitus, cancer, organ transplant, etc. But there was a drastic increase in mucormycosis cases in the ongoing COVID-19 pandemic. Despite several available therapies and antifungal treatments, the mortality rate of mucormycosis is about more than 50%. Currently, there is no vaccine available in the market for mucormycosis that urgently needs to develop a potential vaccine against mucormycosis with high efficacy. In the present study, we have screened 4 genome-derived predicted antigens (GDPA) through sequential filtration of the whole proteome of R. delemar using different benchmarked bioinformatics tools. These 4 GDPA along with 4 randomly selected experimentally reported antigens (ERA) were sourced for prediction of B- and T- cell epitopes and utilized in designing of two potential multi-epitope vaccine candidates which can induce both innate and adaptive immunity against R. delemar. Besides these, comparative immune simulation studies and in silico cloning were performed using L. lactis as an expression system for their possible uses as oral vaccines. This is the first multi-epitope vaccine designed against R. delemar through systematic pipelined reverse vaccinology and immunoinformatic approaches. Although the wet-lab based experimental validation of designed vaccines is required before testing in the preclinical model, the current study will significantly help in reducing the cost of experimentation as well as improving the efficacy of vaccine therapy against mucormycosis and other pathogenic diseases.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Manisha Pritam
- Amity Institute of Biotechnology, Amity University Uttar Pradesh, Lucknow, India
| | - Garima Singh
- Amity Institute of Biotechnology, Amity University Uttar Pradesh, Lucknow, India
| | - Rajnish Kumar
- Amity Institute of Biotechnology, Amity University Uttar Pradesh, Lucknow, India
| | | |
Collapse
|
11
|
In Silico Model Estimates the Clinical Trial Outcome of Cancer Vaccines. Cells 2021; 10:cells10113048. [PMID: 34831269 PMCID: PMC8616443 DOI: 10.3390/cells10113048] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 10/27/2021] [Accepted: 11/03/2021] [Indexed: 12/22/2022] Open
Abstract
Over 30 years after the first cancer vaccine clinical trial (CT), scientists still search the missing link between immunogenicity and clinical responses. A predictor able to estimate the outcome of cancer vaccine CTs would greatly benefit vaccine development. Published results of 94 CTs with 64 therapeutic vaccines were collected. We found that preselection of CT subjects based on a single matching HLA allele does not increase immune response rates (IRR) compared with non-preselected CTs (median 60% vs. 57%, p = 0.4490). A representative in silico model population (MP) comprising HLA-genotyped subjects was used to retrospectively calculate in silico IRRs of CTs based on the percentage of MP-subjects having epitope(s) predicted to bind ≥ 1–4 autologous HLA allele(s). We found that in vitro measured IRRs correlated with the frequency of predicted multiple autologous allele-binding epitopes (AUC 0.63–0.79). Subgroup analysis of multi-antigen targeting vaccine CTs revealed correlation between clinical response rates (CRRs) and predicted multi-epitope IRRs when HLA threshold was ≥ 3 (r = 0.7463, p = 0.0004) but not for single HLA allele-binding epitopes (r = 0.2865, p = 0.2491). Our results suggest that CRR depends on the induction of broad T-cell responses and both IRR and CRR can be predicted when epitopes binding to multiple autologous HLAs are considered.
Collapse
|
12
|
Marczynski M, Kimna C, Lieleg O. Purified mucins in drug delivery research. Adv Drug Deliv Rev 2021; 178:113845. [PMID: 34166760 DOI: 10.1016/j.addr.2021.113845] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 06/02/2021] [Accepted: 06/16/2021] [Indexed: 12/20/2022]
Abstract
One of the main challenges in the field of drug delivery remains the development of strategies to efficiently transport pharmaceuticals across mucus barriers, which regulate the passage and retention of molecules and particles in all luminal spaces of the body. A thorough understanding of the molecular mechanisms, which govern such selective permeability, is key for achieving efficient translocation of drugs and drug carriers. For this purpose, model systems based on purified mucins can contribute valuable information. In this review, we summarize advances that were made in the field of drug delivery research with such mucin-based model systems: First, we give an overview of mucin purification procedures and discuss the suitability of model systems reconstituted from purified mucins to mimic native mucus. Then, we summarize techniques to study mucin binding. Finally, we highlight approaches that made use of mucins as building blocks for drug delivery platforms or employ mucins as active compounds.
Collapse
|
13
|
Lee DH, Choi S, Park Y, Jin HS. Mucin1 and Mucin16: Therapeutic Targets for Cancer Therapy. Pharmaceuticals (Basel) 2021; 14:ph14101053. [PMID: 34681277 PMCID: PMC8537522 DOI: 10.3390/ph14101053] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 10/13/2021] [Accepted: 10/14/2021] [Indexed: 01/18/2023] Open
Abstract
The mucin (MUC) family is a group of highly glycosylated macromolecules that are abundantly expressed in mammalian epithelial cells. MUC proteins contribute to the formation of the mucus barrier and thus have protective functions against infection. Interestingly, some MUC proteins are aberrantly expressed in cancer cells and are involved in cancer development and progression, including cell growth, proliferation, the inhibition of apoptosis, chemoresistance, metabolic reprogramming, and immune evasion. With their unique biological and structural features, MUC proteins have been considered promising therapeutic targets and also biomarkers for human cancer. In this review, we discuss the biological roles of the transmembrane mucins MUC1 and MUC16 in the context of hallmarks of cancer and current efforts to develop MUC1- and MUC16-targeted therapies.
Collapse
Affiliation(s)
- Dong-Hee Lee
- Department of Convergence Medicine, Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Korea;
| | - Seunghyun Choi
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Korea;
| | - Yoon Park
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Korea;
- Correspondence: (Y.P.); (H.-s.J.)
| | - Hyung-seung Jin
- Department of Convergence Medicine, Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Korea;
- Correspondence: (Y.P.); (H.-s.J.)
| |
Collapse
|
14
|
Wang X, Shirazi F, Yan W, Liu X, Wang H, Orlowski RZ, Wang H. Mucin 20 modulates proteasome capacity through c-Met signalling to increase carfilzomib sensitivity in mantle cell lymphoma. J Cell Mol Med 2021; 25:10164-10174. [PMID: 34651428 PMCID: PMC8572801 DOI: 10.1111/jcmm.16953] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2021] [Revised: 09/08/2021] [Accepted: 09/11/2021] [Indexed: 11/28/2022] Open
Abstract
Mantle cell lymphoma (MCL) is a haematologic malignancy. The proteasome inhibitor (PI) bortezomib has been approved to treat MCL, but resistance has emerged through mechanisms that remain unclear. This study aimed to explore the mechanism of PI resistance in MCL and identify new targets for this patient subgroup. Carfilzomib‐resistant (CR) MCL cell lines and primary samples were used for both in vitro and in vivo experiments to identify gene expression and explore their related signalling pathways. We first identified mucin 20 (MUC20) suppression in carfilzomib‐resistant MCL models. MUC20 overexpression sensitized cells to carfilzomib in vitro and in vivo. MUC20 expression was inversely related to activation of c‐Met and the downstream p44/42 MAPK pathway. c‐Met activation with hepatocyte growth factor (HGF) induced PI resistance, while c‐Met inhibition restored PI sensitivity. Carfilzomib resistance and depressed MUC20 expression were associated with enhanced proteasome activity and higher expression of proteassemblin (POMP), a chaperone for catalytically active proteasome assembly. c‐Met and POMP were associated through binding and induction of MAPK‐regulated ELK1 to the POMP promoter. Our data reveal that c‐Met signalling activation enhanced proteasome capacity as a mechanism of PI resistance, and MUC20 expression may be a useful biomarker for PI therapy.
Collapse
Affiliation(s)
- Xiaobin Wang
- The Departments of Hematology, Shengjing Hospital, China Medical University, Shenyang, China
| | - Fazal Shirazi
- The Departments of Lymphoma/Myeloma, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Wei Yan
- The Departments of Hematology, Shengjing Hospital, China Medical University, Shenyang, China
| | - Xiaoyu Liu
- The Departments of Hematology, Shengjing Hospital, China Medical University, Shenyang, China
| | - Hua Wang
- The Departments of Lymphoma/Myeloma, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Robert Z Orlowski
- The Departments of Lymphoma/Myeloma, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Huihan Wang
- The Departments of Hematology, Shengjing Hospital, China Medical University, Shenyang, China
| |
Collapse
|
15
|
Monjezi MR, Fouladseresht H, Farjadian S, Gharesi-Fard B, Khosropanah S, Doroudchi M. T Cell Proliferative Responses and IgG Antibodies to β2GPI in Patients with Diabetes and Atherosclerosis. Endocr Metab Immune Disord Drug Targets 2021; 21:495-503. [PMID: 32368987 DOI: 10.2174/1871530320666200505115850] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2020] [Revised: 03/15/2020] [Accepted: 03/20/2020] [Indexed: 11/22/2022]
Abstract
BACKGROUND Diabetes increases the risk of myocardial infarction (MI) by 2 to 3 folds. Tlymphocytes play a role in atherosclerosis, which is the main pathology behind MI. Cellular immune responses to beta-2 glycoprotein I (β2GPI) are shown in carotid atherosclerosis. OBJECTIVE To investigate the self-reactive, β2GPI-specific T-lymphocytes in patients with and without diabetes and atherosclerosis. METHODS Collectively, 164 subjects with and without diabetes that underwent coronary angiography were divided into four groups based on their diabetes status and coronary stenosis. Group I=Diabetic with ≥50% stenosis: A+D+ (n=66); Group II=Non-diabetic with ≥50% stenosis, A+D- (n=39); Group III=Diabetic with <50% stenosis: A-D+ (n=28); and Group IV=Non-diabetic with <50% stenosis: AD- (n=31). All groups were evaluated for anti-β2GPI IgG antibody by ELISA method. Then, PBMCs were isolated from 18 subjects and were stimulated with β2GPI-derived peptides to assess their proliferation in accordance with their HLA-DRB1 alleles. RESULTS Mean β2GPI IgG levels were higher in groups with ≥50% stenosis (A+) compared to those with <50% stenosis (A-), (P=0.02). The co-presence of diabetes in A+ individuals increased mean β2GPI-specific IgG. Auto-reactive β2GPI-specific T cells were detected in the repertoire of T-lymphocytes in all groups. β2GPI-peptides showed promiscuous restriction by various HLADRB1. CONCLUSION β2GPI is the target of cellular and humoral immune responses in patients with atherosclerosis. Since the T cell responses but not antibodies were detectable in A-D+ and A-D- groups, it is reasonable to assume that cellular responses preceded the humoral responses. Post-translation modifications of β2GPI under oxidative and glycemic stresses may have increased the IgG levels in patients with diabetes. Finally, identification of antigens that trigger immuno-pathogenesis in atherosclerosis and diabetes may help the development of immunomodulation methods to prevent or treat these debilitating diseases.
Collapse
Affiliation(s)
- Mohammad R Monjezi
- Department of Immunology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Hamed Fouladseresht
- Department of Immunology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Shirin Farjadian
- Department of Immunology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Behrouz Gharesi-Fard
- Department of Immunology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Shahdad Khosropanah
- Department of Cardiology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mehrnoosh Doroudchi
- Department of Immunology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
16
|
Brockhausen I, Melamed J. Mucins as anti-cancer targets: perspectives of the glycobiologist. Glycoconj J 2021; 38:459-474. [PMID: 33704667 DOI: 10.1007/s10719-021-09986-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 02/22/2021] [Accepted: 02/26/2021] [Indexed: 12/11/2022]
Abstract
Mucins are highly O-glycosylated glycoproteins that carry a heterogenous variety of O-glycan structures. Tumor cells tend to overexpress specific mucins, such as the cell surface mucins MUC1 and MUC4 that are engaged in signaling and cell growth, and exhibit abnormal glycosylation. In particular, the Tn and T antigens and their sialylated forms are common in cancer mucins. We review herein methods chosen to use cancer-associated glycans and mucins as targets for the design of anti-cancer immunotherapies. Mucin peptides from the glycosylated and transmembrane domains have been combined with immune-stimulating adjuvants in a wide variety of approaches to produce anti-tumor antibodies and vaccines. These mucin conjugates have been tested on cancer cells in vitro and in mice with significant successes in stimulating anti-tumor responses. The clinical trials in humans, however, have shown limited success in extending survival. It seems critical that the individual-specific epitope expression of cancer mucins is considered in future therapies to result in lasting anti-tumor responses.
Collapse
Affiliation(s)
- Inka Brockhausen
- Biomedical and Molecular Sciences, Queen's University, 18 Stuart St, Kingston, ON, K7L 3N6, Canada.
| | - Jacob Melamed
- Biomedical and Molecular Sciences, Queen's University, 18 Stuart St, Kingston, ON, K7L 3N6, Canada
| |
Collapse
|
17
|
Liegel J, Weinstock M, Rosenblatt J, Avigan D. Vaccination as Immunotherapy in Hematologic Malignancies. J Clin Oncol 2021; 39:433-443. [PMID: 33434056 DOI: 10.1200/jco.20.01706] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Affiliation(s)
- Jessica Liegel
- Beth Israel Deaconess Medical Center, Department of Medicine, Division of Hematology and Hematologic Malignancies, Harvard Medical School, Boston, MA
| | - Matthew Weinstock
- Beth Israel Deaconess Medical Center, Department of Medicine, Division of Hematology and Hematologic Malignancies, Harvard Medical School, Boston, MA
| | - Jacalyn Rosenblatt
- Beth Israel Deaconess Medical Center, Department of Medicine, Division of Hematology and Hematologic Malignancies, Harvard Medical School, Boston, MA
| | - David Avigan
- Beth Israel Deaconess Medical Center, Department of Medicine, Division of Hematology and Hematologic Malignancies, Harvard Medical School, Boston, MA
| |
Collapse
|
18
|
Chen XR, Yuan HH, Guo JH, Zhang WY, Li QQ, Huang GD, Zhang YJ, Jiang B, Liu F. A signal peptide derived from Hsp60 induces protective cytotoxic T lymphocyte immunity against lymphoid malignancies independently of TAP and classical MHC-I. Cancer Lett 2020; 494:47-57. [PMID: 32829008 DOI: 10.1016/j.canlet.2020.08.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2020] [Revised: 08/04/2020] [Accepted: 08/14/2020] [Indexed: 10/23/2022]
Abstract
Hsp60sp, a signal peptide derived from the leader sequence of heat shock protein 60 kDa (Hsp60), is a Qa-1/HLA-E-binding peptide. We previously showed that Hsp60sp-specific CD8+ T cells are involved in the immunoregulation of autoimmune diseases by controlling the response of self-reactive lymphocytes. Here, we report that Hsp60sp-specific CD8+ T cells killed malignant lymphocytes in vitro independently of transporter associated with antigen processing (TAP) and classical MHC-I expression. Induction of this cytotoxic T lymphocyte (CTL) response in vivo, either by adoptive transfer of in vitro-amplified CTLs or peptide-loaded dendritic cell immunization, resulted in effective control of lymphoid tumors, including TAP- or classical MHC-I-deficient cells. Hsp60sp-specific immune activation combined with programmed cell death protein 1 (PD-1) blocking synergistically restrained mouse lymphoma development. Importantly, Hsp60sp-specific CD8+ T cells did not negatively affect normal tissues and cells. Our data suggest that Hsp60sp-based immunotherapy is an inviting strategy to control lymphoid malignancies.
Collapse
Affiliation(s)
- Xun-Rui Chen
- Oncology Department, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 201900, China
| | - Hai-Hua Yuan
- Oncology Department, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 201900, China
| | - Jia-Hui Guo
- Oncology Department, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 201900, China
| | - Wen-Ying Zhang
- Oncology Department, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 201900, China
| | - Qian-Qian Li
- Oncology Department, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 201900, China
| | - Guo-Ding Huang
- Oncology Department, Hainan Western Central Hospital, Danzhou, 571700, Hainan Province, China
| | - Yan-Jie Zhang
- Oncology Department, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 201900, China.
| | - Bin Jiang
- Oncology Department, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 201900, China.
| | - Feng Liu
- Oncology Department, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 201900, China; Oncology Department, Hainan Western Central Hospital, Danzhou, 571700, Hainan Province, China.
| |
Collapse
|
19
|
Frederiksen LSF, Zhang Y, Foged C, Thakur A. The Long Road Toward COVID-19 Herd Immunity: Vaccine Platform Technologies and Mass Immunization Strategies. Front Immunol 2020; 11:1817. [PMID: 32793245 PMCID: PMC7385234 DOI: 10.3389/fimmu.2020.01817] [Citation(s) in RCA: 151] [Impact Index Per Article: 30.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2020] [Accepted: 07/07/2020] [Indexed: 12/14/2022] Open
Abstract
There is an urgent need for effective countermeasures against the current emergence and accelerating expansion of coronavirus disease 2019 (COVID-19), caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Induction of herd immunity by mass vaccination has been a very successful strategy for preventing the spread of many infectious diseases, hence protecting the most vulnerable population groups unable to develop immunity, for example individuals with immunodeficiencies or a weakened immune system due to underlying medical or debilitating conditions. Therefore, vaccination represents one of the most promising counter-pandemic measures to COVID-19. However, to date, no licensed vaccine exists, neither for SARS-CoV-2 nor for the closely related SARS-CoV or Middle East respiratory syndrome-CoV. In addition, a few vaccine candidates have only recently entered human clinical trials, which hampers the progress in tackling COVID-19 infection. Here, we discuss potential prophylactic interventions for SARS-CoV-2 with a focus on the challenges existing for vaccine development, and we review pre-clinical progress and ongoing human clinical trials of COVID-19 vaccine candidates. Although COVID-19 vaccine development is currently accelerated via so-called fast-track programs, vaccines may not be timely available to have an impact on the first wave of the ongoing COVID-19 pandemic. Nevertheless, COVID-19 vaccines will be essential in the future for reducing morbidity and mortality and inducing herd immunity, if SARS-CoV-2 becomes established in the population like for example influenza virus.
Collapse
Affiliation(s)
| | - Yibang Zhang
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.,Department of Pharmaceutics, School of Pharmacy, Jiangsu University, Zhenjiang, China
| | - Camilla Foged
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Aneesh Thakur
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
20
|
Abstract
Cellular immunotherapy for myeloma has the unique potential both to potently kill the malignant clone and to evoke a memory response to protect from relapse. Understanding the complex interactions between the malignant clone and the microenvironment that promote immune escape is critical to evoke effective antimyeloma immunity. Tremendous progress has been made in the area of cancer vaccines and adoptive T-cell therapy in recent years. Careful study of the mechanisms of response and of immune escape will be critical to developing novel combination therapies and ultimately to improve outcomes for patients with myeloma.
Collapse
|
21
|
miR-485-5p inhibits the progression of breast cancer cells by negatively regulating MUC1. Breast Cancer 2020; 27:765-775. [PMID: 32144736 DOI: 10.1007/s12282-020-01075-2] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Accepted: 02/25/2020] [Indexed: 12/14/2022]
Abstract
OBJECTIVE To investigate the mechanism of miR-485-5p inhibiting breast cancer cells by targeting MUC1. METHODS Differentially expressed genes (DEGs) in breast cancer tissues were analyzed using breast cancer tissue microarrays (TMA) in the GEO database. Differential expression of MUC1 in breast cancer tissue samples was detected by TCGA database. qRT-PCR was used to detect the expression of MUC1 and miR-485-5p in human normal breast epithelial cell lines and human breast cancer cell lines. Bioinformatics was applied to analyze targeted binding site of miR-485-5p and MUC1 and their targeted relationship was identified by dual luciferase assay. The proliferation ability of breast cancer cells was detected by CCK-8 assay. Cell apoptosis was detected by flow cytometry. The ability of cell migration was measured by scratch healing test. Transwell assay was used to detect the invasion ability of cells. The protein expression levels of MUC1 and EMT-related molecules (E-cadherin, N-cadherin and Vimentin) were detected by Western blot. RESULTS MUC1 was highly expressed in breast cancer tissue samples and breast cancer cell lines, while miR-485-5p was lowly expressed. Overexpression of miR-485-5p inhibits cell viability and invasion and migration of breast cancer cell line MCF-7 and promotes apoptosis. The same results were obtained by silencing the expression of MUC1. MiR-485-5p targets to bind to the 3'-UTR region of MUC1 and negatively regulates the expression of MUC1. Overexpressing MUC1 while overexpressing miR-485-5p reversed the inhibitory effect of miR-485-5p on breast cancer and inhibited EMT. CONCLUSION MiR-485-5p can down-regulate the expression of MUC1, thus inhibit the proliferation, invasion and migration of breast cancer cells and promote cell apoptosis.
Collapse
|
22
|
Utilizing VEGF165b mutant as an effective immunization adjunct to augment antitumor immune response. Vaccine 2019; 37:2090-2098. [DOI: 10.1016/j.vaccine.2019.02.055] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2018] [Revised: 01/22/2019] [Accepted: 02/22/2019] [Indexed: 12/20/2022]
|
23
|
Vaccine therapy in hematologic malignancies. Blood 2018; 131:2640-2650. [DOI: 10.1182/blood-2017-11-785873] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Accepted: 02/04/2018] [Indexed: 02/06/2023] Open
Abstract
Abstract
Immune-based therapy has emerged as a paradigm shift in cancer therapy with dramatic responses observed in previously incurable disease. Cancer vaccines are being developed to disrupt tumor-associated tolerance and activate and selectively expand tumor-specific lymphocytes within the native effector cell repertoire while maintaining immune-regulatory protection against autoimmunity. Although individual antigen approaches result in immune response with a suggestion of clinical effect in some settings, broader efficacy may be dependent on presentation of multiple antigens that capture clonal diversity presented in the context of functionally potent antigen-presenting cells. The use of whole cell–based strategies such as dendritic cell/tumor fusions have yielded provocative results in single-arm studies and are currently being explored in multicenter randomized trials. The posttransplant setting is a potentially promising platform for vaccination due to cytoreduction and relative depletion of inhibitory accessory cells fostering greater immune responsiveness. Integration of these efforts with other immunotherapeutic strategies and agents that target the tumor microenvironment is being studied in an effort to generate durable immunologic responses with clinically meaningful impact on disease.
Collapse
|
24
|
Nahas MR, Rosenblatt J, Lazarus HM, Avigan D. Anti-cancer vaccine therapy for hematologic malignancies: An evolving era. Blood Rev 2018; 32:312-325. [PMID: 29475779 DOI: 10.1016/j.blre.2018.02.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2017] [Revised: 02/08/2018] [Accepted: 02/13/2018] [Indexed: 12/19/2022]
Abstract
The potential promise of therapeutic vaccination as effective therapy for hematologic malignancies is supported by the observation that allogeneic hematopoietic cell transplantation is curative for a subset of patients due to the graft-versus-tumor effect mediated by alloreactive lymphocytes. Tumor vaccines are being explored as a therapeutic strategy to re-educate host immunity to recognize and target malignant cells through the activation and expansion of effector cell populations. Via several mechanisms, tumor cells induce T cell dysfunction and senescence, amplifying and maintaining tumor cell immunosuppressive effects, resulting in failure of clinical trials of tumor vaccines and adoptive T cell therapies. The fundamental premise of successful vaccine design involves the introduction of tumor-associated antigens in the context of effective antigen presentation so that tolerance can be reversed and a productive response can be generated. With the increasing understanding of the role of both the tumor and tumor microenvironment in fostering immune tolerance, vaccine therapy is being explored in the context of immunomodulatory therapies. The most effective strategy may be to use combination therapies such as anti-cancer vaccines with checkpoint blockade to target critical aspects of this environment in an effort to prevent the re-establishment of tumor tolerance while limiting toxicity associated with autoimmunity.
Collapse
Affiliation(s)
- Myrna R Nahas
- Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA.
| | - Jacalyn Rosenblatt
- Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Hillard M Lazarus
- Department of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - David Avigan
- Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA
| |
Collapse
|
25
|
Kumar SK, Anderson KC. Immune Therapies in Multiple Myeloma. Clin Cancer Res 2018; 22:5453-5460. [PMID: 28151713 DOI: 10.1158/1078-0432.ccr-16-0868] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2016] [Revised: 09/20/2016] [Accepted: 09/22/2016] [Indexed: 11/16/2022]
Abstract
Treatment paradigms have changed rapidly for multiple myeloma, and immune therapies have taken center stage. Advances in therapies for myeloma have led to a dramatic improvement in the survival of patients with this incurable malignancy. The immune system is significantly impaired in patients with myeloma as a result of the disease leading to suppression of normal plasma cells as well the negative effects on cellular immunity. Given this scenario, immune approaches have not been successful until recently. Monoclonal antibodies directed against CD38 (daratumumab) and SLAMF7 (elotuzumab) are already in the clinic, and several other antibodies directed against different plasma cell antigens are under evaluation. Although immune checkpoint inhibition with PD-1 inhibitors had no clinical efficacy when the inhibitors were used as single agents, it has led to some dramatic results when the inhibitors are combined with immunomodulatory drugs such as lenalidomide and pomalidomide. Vaccination strategies have shown in vivo immune responses but no clear clinical efficacy. Newer approaches to vaccination with multiple antigens, used in combinations with immunomodulatory drugs and in the setting of minimal residual disease, have all increased possibility of this approach succeeding. Ex vivo effector cell expansion also appears to be promising and is in clinical trials. Finally, a chimeric antigen receptor T-cell approach appears to have some promise based on isolated reports of success and remains an area of intense investigation. Immune-based approaches can potentially augment or even supplant some of the current approaches and, given the low toxicity profile, may hold great potential in the early treatment of precursor-stage diseases. Clin Cancer Res; 22(22); 5453-60. ©2016 AACR SEE ALL ARTICLES IN THIS CCR FOCUS SECTION, "MULTIPLE MYELOMA MULTIPLYING THERAPIES".
Collapse
Affiliation(s)
- Shaji K Kumar
- Division of Hematology, Mayo Clinic, Rochester, Minnesota.
| | | |
Collapse
|
26
|
Abstract
PURPOSE OF REVIEW Here, we explore the significant progress made in the treatment of multiple myeloma, focusing on immunotherapy and the promise it has offered to patients suffering from advanced disease. RECENT FINDINGS Multiple myeloma, a B-cell malignancy, is characterized by unregulated plasma cell growth in the bone marrow as well as strong immunosuppression in the tumor microenvironment. mAbs targeting tumor antigens overcome this, increasing T-cell activation, multiple myeloma cell death, and depth of response. Similarly, adoptive T-cell therapy aims to engineer or isolate tumor-specific T cells for a targeted approach. Finally, peptide and dendritic cell/tumor fusion vaccines reeducate the immune system, expanding the immune response and generating long-term memory to prevent relapse of disease. Many of these approaches have been combined with existing therapies to enhance antitumor immunity. SUMMARY Immunotherapeutic approaches have remarkably changed the treatment paradigm for multiple myeloma, and encouraging patient responses have warranted further investigation into mAbs, adoptive T-cell therapy, vaccines, and combination therapy.
Collapse
|
27
|
Kumar S. Emerging options in multiple myeloma: targeted, immune, and epigenetic therapies. HEMATOLOGY. AMERICAN SOCIETY OF HEMATOLOGY. EDUCATION PROGRAM 2017; 2017:518-524. [PMID: 29222300 PMCID: PMC6142613 DOI: 10.1182/asheducation-2017.1.518] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/31/2023]
Abstract
Considerable progress has been made in the treatment of multiple myeloma in the past decade with median survival for the disease improving significantly. This has come through a combination of better understanding of the disease biology and coordinated research into new treatment approaches including better supportive care. However, patients eventually become refractory to available treatments and succumb to the disease, highlighting the need to develop new treatment approaches. The genetic heterogeneity in the disease and clonal evolution under treatment pressure underlie the development of resistance, underscoring the need to develop more effective therapies that can eradicate the disease at initial treatment as well as the need for new classes of drugs with varying mechanisms of action. To this end, there has been intense focus on exploring novel approaches to therapy including small-molecule inhibitors targeting specific abnormalities, immune therapies including monoclonal antibodies and adaptive T-cell therapy, as well as epigenetic approaches. Although many of these drugs are in the early stages of clinical development, the early data appear to be very promising. Many of these drugs can be safely and effectively combined with the current treatment classes such as proteasome inhibitors and immunomodulatory drugs, further enhancing the treatment options for myeloma.
Collapse
Affiliation(s)
- Shaji Kumar
- Division of Hematology and Internal Medicine, Mayo Clinic, Rochester, MN
| |
Collapse
|
28
|
Nabavinia MS, Gholoobi A, Charbgoo F, Nabavinia M, Ramezani M, Abnous K. Anti-MUC1 aptamer: A potential opportunity for cancer treatment. Med Res Rev 2017; 37:1518-1539. [PMID: 28759115 DOI: 10.1002/med.21462] [Citation(s) in RCA: 91] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Revised: 06/19/2017] [Accepted: 06/30/2017] [Indexed: 01/10/2023]
Abstract
Mucin 1 (MUC1) is a protein usually found on the apical surface of most normal secretory epithelial cells. However, in most adenocarcinomas, MUC1 is overexpressed, so that it not only appears over the entire cell surface, but is also shed as MUC1 fragments into the blood stream. These phenomena pinpoint MUC1 as a potential target for the diagnosis and treatment of cancer; consequently, interest has increased in MUC1 as a molecular target for overcoming cancer therapy challenges. MUC1 currently ranks second among 75 antigen candidates for cancer vaccines, and different antibodies or aptamers against MUC1 protein are proving useful for tracing cancer cells in the emerging field of targeted delivery. The unique properties of MUC1 aptamers as novel targeting agents, and the revolutionary role that MUC1 now plays in cancer therapy, are the focus of this review. Recent advancements in MUC1-targeted cancer therapy are also assessed.
Collapse
Affiliation(s)
- Maryam Sadat Nabavinia
- Department of Pharmaceutical Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Aida Gholoobi
- Department of Modern Sciences and Technologies, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Fahimeh Charbgoo
- Department of Pharmaceutical Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.,Pharmaceutical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | - Mohammad Ramezani
- Department of Pharmaceutical Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.,Nanotechnology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Khalil Abnous
- Pharmaceutical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.,Department of Medicinal Chemistry, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
29
|
Abstract
Hematological malignancies manifest as lymphoma, leukemia, and myeloma, and remain a burden on society. From initial therapy to endless relapse-related treatment, societal burden is felt not only in the context of healthcare cost, but also in the compromised quality of life of patients. Long-term therapeutic strategies have become the standard in keeping hematological malignancies at bay as these cancers develop resistance to each round of therapy with time. As a result, there is a continual need for the development of new drugs to combat resistant disease in order to prolong patient life, if not to produce a cure. This review aims to summarize advances in targeting lymphoma, leukemia, and myeloma through both cutting-edge and well established platforms. Current standard of treatment will be reviewed for these malignancies and emphasis will be made on new therapy development in the areas of antibody engineering, epigenetic small molecule inhibiting drugs, vaccine development, and chimeric antigen receptor cell engineering. In addition, platforms for the delivery of these and other drugs will be reviewed including antibody-drug conjugates, micro- and nanoparticles, and multimodal hydrogels. Lastly, we propose that tissue engineered constructs for hematological malignancies are the missing link in targeted drug discovery alongside mouse and patient-derived xenograft models.
Collapse
|
30
|
Tang Y, Cui X, Xiao H, Qi S, Hu X, Yu Q, Shi G, Zhang X, Gu J, Yu Y, Wang L, Li Y. Binding of circulating anti-MUC1 antibody and serum MUC1 antigen in stage IV breast cancer. Mol Med Rep 2017; 15:2659-2664. [PMID: 28447743 DOI: 10.3892/mmr.2017.6323] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2015] [Accepted: 11/21/2016] [Indexed: 11/05/2022] Open
Abstract
The present study aimed to investigate the binding of circulating mucin 1 (MUC1) antibody with serum MUC1 antigen in stage IV breast cancer. Serum samples of 61 patients with stage IV breast cancer and 64 patients with early-stage breast cancer were collected. The anti‑MUC1 antibody (IgG) and MUC1 antigen (cancer antigen 15‑3; Ca15‑3) were detected using an indirect enzyme-linked immunosorbent assay (I‑ELISA) and ELISA, respectively. The MUC1 IgG affinity was detected using a urea degradation combining ELISA. Western blot analysis and an inhibition test were performed for verification of the binding of anti‑MUC1 IgG with MUC1 antigen, and their correlation was analyzed. The results showed that there was a negative correlation between anti‑MUC1 IgG and CA15‑3 antigen in stage IV breast cancer when positive CA15‑3 antigen and/or anti‑MUC1 IgG were selected (r=‑0.417; P=0.0044). The positive anti‑MUC1 IgG with positive Ca15‑3 antigen was more common in stage IV breast cancer, compared with early‑stage breast cancer (χ2=4.629; P=0.031), however, Ca15‑3 antigen positivity was higher in stage IV breast cancer, compared with early‑stage breast cancer (χ2=10.58; P=0.001). Anti‑MUC1 IgG was able to bind to the MUC1 antigen in stage IV breast cancer. No differences in the 8R-MUCPT inhibition ratio were found between the two groups (P=0.778), and there were no differences in the affinity of anti‑MUC1 IgG (P=0.873). In stage IV breast cancer, circulating anti‑MUC1 antibody was found to bind serum MUC1 antigen, although their compatibility was low. No significant difference was found in the affinity of the anti‑MUC1 antibody between stage IV breast cancer and early‑stage breast cancer.
Collapse
Affiliation(s)
- Yan Tang
- Department of Oncology, The Second Hospital, Jilin University, Changchun, Jilin 130041, P.R. China
| | - Xiuxia Cui
- Department of Teaching and Scientific Research, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Han Xiao
- Department of Oncology, The Second Hospital, Jilin University, Changchun, Jilin 130041, P.R. China
| | - Shengkun Qi
- Department of Immunology, Norman Bethune Medical College, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Xiaoping Hu
- Department of Molecular Biology, Norman Bethune Medical College, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Qiong Yu
- Department of Oncology, The Second Hospital, Jilin University, Changchun, Jilin 130041, P.R. China
| | - Guang Shi
- Department of Oncology, The Second Hospital, Jilin University, Changchun, Jilin 130041, P.R. China
| | - Xue Zhang
- Department of Oncology, The Second Hospital, Jilin University, Changchun, Jilin 130041, P.R. China
| | - Jiaying Gu
- Department of Oncology, The Second Hospital, Jilin University, Changchun, Jilin 130041, P.R. China
| | - Yongli Yu
- Department of Immunology, Norman Bethune Medical College, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Liying Wang
- Department of Molecular Biology, Norman Bethune Medical College, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Yarong Li
- Department of Oncology, The Second Hospital, Jilin University, Changchun, Jilin 130041, P.R. China
| |
Collapse
|
31
|
Rao CV, Janakiram NB, Mohammed A. Molecular Pathways: Mucins and Drug Delivery in Cancer. Clin Cancer Res 2017; 23:1373-1378. [PMID: 28039261 PMCID: PMC6038927 DOI: 10.1158/1078-0432.ccr-16-0862] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2016] [Revised: 11/29/2016] [Accepted: 11/30/2016] [Indexed: 12/12/2022]
Abstract
Over the past few decades, clinical and preclinical studies have clearly demonstrated the role of mucins in tumor development. It is well established that mucins form a barrier impeding drug access to target sites, leading to cancer chemoresistance. Recently gained knowledge regarding core enzyme synthesis has opened avenues to explore the possibility of disrupting mucin synthesis to improve drug efficacy. Cancer cells exploit aberrant mucin synthesis to efficiently mask the epithelial cells and ensure survival under hostile tumor microenvironment conditions. However, O-glycan synthesis enzyme core 2 beta 1,6 N-acetylglucosaminyltransferase (GCNT3/C2GnT-2) is overexpressed in Kras-driven mouse and human cancer, and inhibition of GCNT3 has been shown to disrupt mucin synthesis. This previously unrecognized developmental pathway might be responsible for aberrant mucin biosynthesis and chemoresistance. In this Molecular Pathways article, we briefly discuss the potential role of mucin synthesis in cancers, ways to improve drug delivery and disrupt mucin mesh to overcome chemoresistance by targeting mucin synthesis, and the unique opportunity to target the GCNT3 pathway for the prevention and treatment of cancers. Clin Cancer Res; 23(6); 1373-8. ©2016 AACR.
Collapse
Affiliation(s)
- Chinthalapally V Rao
- Center for Cancer Prevention and Drug Development, Hematology and Oncology Section, Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma.
| | - Naveena B Janakiram
- Center for Cancer Prevention and Drug Development, Hematology and Oncology Section, Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Altaf Mohammed
- Center for Cancer Prevention and Drug Development, Hematology and Oncology Section, Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma.
| |
Collapse
|
32
|
Monoclonal antibody therapy in multiple myeloma. Leukemia 2017; 31:1039-1047. [PMID: 28210004 DOI: 10.1038/leu.2017.60] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2016] [Revised: 01/14/2017] [Accepted: 02/06/2017] [Indexed: 02/07/2023]
Abstract
The therapeutic landscape of multiple myeloma (MM) has evolved spectacularly over the past decade with the discovery and validation of proteasome inhibitors and immunomodulatory agents as highly active agents, both in front-line therapy as well as in the relapse and maintenance settings. Although previous attempts to apply available monoclonal antibodies (Mabs) to the treatment of patients with MM has until recently been disappointing, novel targets specifically explored in the context of MM have recently lead to the first approvals of Mabs for the treatment of patients with MM. We have performed a literature search to identify preclinical targeting of MM, including in vitro and in vivo models using monoclonal antibodies, as well as clinical trials of monoclonal antibodies in patients with MM. Sources used were peer-reviewed publications, congress abstracts and on-line clinical trials data (such as clinicaltrials.gov). Several targets have been evaluated in preclinical models and a growing number of agents are being evaluated in clinical trials, as single agents or in combination and under various antibody formats. Two agents, targeting for the first time CD38 and SLAMF7, respectively, have recently been approved for the treatment of patients with MM. The recent approval of these two antibodies is expected to have a strong impact on treatment modalities and outcome in patients with MM, including both transplant eligible and elderly patients.
Collapse
|
33
|
Chung C. Role of Immunotherapy in Targeting the Bone Marrow Microenvironment in Multiple Myeloma: An Evolving Therapeutic Strategy. Pharmacotherapy 2017; 37:129-143. [PMID: 27870103 DOI: 10.1002/phar.1871] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Multiple myeloma (referred to henceforth as myeloma) is a B-cell malignancy characterized by unregulated growth of plasma cells in the bone marrow. The treatment paradigm for myeloma underwent significant evolution in the last decade, with an improved understanding of the pathogenesis of the disease as well as the development of therapeutic agents that target not only the tumor cells but also their microenvironment. Despite these therapeutic advances, the prognosis of patients with relapsed or refractory myeloma remains poor. Accordingly, a need exists for new therapeutic avenues that can overcome resistance to current therapies and improve survival outcomes. In addition, myeloma is associated with progressive immune dysregulation, with defects in T-cell immunity, natural killer cell function, and the antigen-presenting capacity of dendritic cells, resulting in a tumor microenvironment that promotes disease tolerance and progression. Together, the immunosuppressive microenvironment and oncogenic mutations activate signaling networks that promote myeloma cell survival. Immunotherapy incorporates novel treatment options (e.g., monoclonal antibodies, antibody-drug conjugates, chimeric antigen receptor T-cell therapy, immune checkpoint inhibitors, bispecific antibodies, and tumor vaccines) either alone or in combination with existing lines of therapies (e.g., immunomodulatory agents, proteasome inhibitors, and histone deacetylase inhibitors) to enhance the host anti myeloma immunity within the bone marrow microenvironment and improve clinical response. Following the U.S. Food and Drug Administration approval of daratumumab and elotuzumab in 2015, more immunotherapeutic agents are expected to be become available as valuable treatment options in the near future. This review provides a basic understanding of the role of immunotherapy in modulating the bone marrow tumor microenvironment and its role in the treatment of myeloma. Clinical efficacy and safety of recently approved therapeutic monoclonal antibodies (daratumumab, elotuzumab) are discussed, along with the therapeutic potential of emerging immunotherapies (antibody-drug conjugates, chimeric antigen receptor T-cell therapy, tumor vaccines, and immune checkpoint inhibitors).
Collapse
|
34
|
PD-1/PD-L1 expression in extra-medullary lesions of multiple myeloma. Leuk Res 2016; 49:98-101. [DOI: 10.1016/j.leukres.2016.09.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2016] [Revised: 08/13/2016] [Accepted: 09/04/2016] [Indexed: 12/18/2022]
|
35
|
Sousa AM, Grandgenett PM, David L, Almeida R, Hollingsworth MA, Santos-Silva F. Reflections on MUC1 glycoprotein: the hidden potential of isoforms in carcinogenesis. APMIS 2016; 124:913-924. [PMID: 27538373 DOI: 10.1111/apm.12587] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Accepted: 07/03/2016] [Indexed: 12/13/2022]
Abstract
Mucin 1 (MUC1) has been described as the renaissance molecule due to the large set of functions it displays in both normal and neoplastic cells. This membrane-tethered glycoprotein is overexpressed and aberrantly glycosylated in most epithelial cancers, being involved in several processes related with malignant phenotype acquisition. With a highly polymorphic structure, both in the polypeptide and glycan counterparts, MUC1 variability has been associated with susceptibility to several diseases, including cancer. Biochemical features and biological functions have been characterized upon the full-length MUC1 protein, remaining to clarify the real impact on cell dynamics of the plethora of MUC1 isoforms. This review aims to encompass a detailed characterization of MUC1 role in carcinogenesis, highlighting recent findings in cell differentiation and uncovering new evidences of MUC1 isoforms involvement in malignant phenotype.
Collapse
Affiliation(s)
- Andreia M Sousa
- i3S-Institute of Research and Innovation in Health, University of Porto, Porto, Portugal. .,IPATIMUP-Institute of Molecular Pathology and Immunology of the University of Porto, Porto, Portugal.
| | - Paul M Grandgenett
- Eppley Institute for Research in Cancer and Allied Disease, Omaha, NE, USA
| | - Leonor David
- i3S-Institute of Research and Innovation in Health, University of Porto, Porto, Portugal.,IPATIMUP-Institute of Molecular Pathology and Immunology of the University of Porto, Porto, Portugal.,Faculty of Medicine of the University of Porto, Porto, Portugal
| | - Raquel Almeida
- i3S-Institute of Research and Innovation in Health, University of Porto, Porto, Portugal.,IPATIMUP-Institute of Molecular Pathology and Immunology of the University of Porto, Porto, Portugal.,Faculty of Medicine of the University of Porto, Porto, Portugal.,Department of Biology, Faculty of Sciences of the University of Porto, Porto, Portugal
| | | | - Filipe Santos-Silva
- i3S-Institute of Research and Innovation in Health, University of Porto, Porto, Portugal.,IPATIMUP-Institute of Molecular Pathology and Immunology of the University of Porto, Porto, Portugal.,Faculty of Medicine of the University of Porto, Porto, Portugal
| |
Collapse
|
36
|
Aberrant Glycosylation of Anchor-Optimized MUC1 Peptides Can Enhance Antigen Binding Affinity and Reverse Tolerance to Cytotoxic T Lymphocytes. Biomolecules 2016; 6:biom6030031. [PMID: 27367740 PMCID: PMC5039417 DOI: 10.3390/biom6030031] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2016] [Revised: 06/07/2016] [Accepted: 06/07/2016] [Indexed: 12/22/2022] Open
Abstract
Cancer vaccines have often failed to live up to their promise, although recent results with checkpoint inhibitors are reviving hopes that they will soon fulfill their promise. Although mutation-specific vaccines are under development, there is still high interest in an off-the-shelf vaccine to a ubiquitous antigen, such as MUC1, which is aberrantly expressed on most solid and many hematological tumors, including more than 90% of breast carcinomas. Clinical trials for MUC1 have shown variable success, likely because of immunological tolerance to a self-antigen and to poor immunogenicity of tandem repeat peptides. We hypothesized that MUC1 peptides could be optimized, relying on heteroclitic optimizations of potential anchor amino acids with and without tumor-specific glycosylation of the peptides. We have identified novel MUC1 class I peptides that bind to HLA-A*0201 molecules with significantly higher affinity and function than the native MUC1 peptides. These peptides elicited CTLs from normal donors, as well as breast cancer patients, which were highly effective in killing MUC1-expressing MCF-7 breast cancer cells. Each peptide elicited lytic responses in greater than 6/8 of normal individuals and 3/3 breast cancer patients. The CTLs generated against the glycosylated-anchor modified peptides cross reacted with the native MUC1 peptide, STAPPVHNV, suggesting these analog peptides may offer substantial improvement in the design of epitope-based vaccines.
Collapse
|
37
|
Abstract
Mucin1 (MUC1) is a transmembrane oncogenic protein that plays a central role in malignant transformation and disease evolution, including cell proliferation, survival, self-renewal, and metastatic invasion. MUC1 has been shown to interact with diverse effectors such as β-catenin, receptor tyrosine kinases, and c-Abl, which are of importance in the pathogenesis of various hematological malignancies. In myeloid leukemia, MUC1 has been shown to have an essential role in leukemia stem-cell function, the induction of reactive oxygen species (ROS), and the promotion of terminal myeloid differentiation. As such, MUC1 is an attractive therapeutic target in hematologic malignancies. Targeting MUC1 has been shown to be an effective approach for inducing cell death in tumor in in vivo and in vitro models. Furthermore, MUC1 inhibition is synergistic with other therapeutic agents in the treatment of hematologic disorders. This review will explore the role of MUC1 in hematological malignancies, and strategies for targeting this oncoprotein.
Collapse
Affiliation(s)
- Dina Stroopinsky
- a Beth Israel Deaconess Medical Center , Harvard Medical School , Boston , MA , USA
| | - Donald Kufe
- b Dana Farber Cancer Institute, Harvard Medical School , Boston , MA , USA
| | - David Avigan
- a Beth Israel Deaconess Medical Center , Harvard Medical School , Boston , MA , USA
| |
Collapse
|
38
|
Nahas MR, Avigan D. Challenges in vaccine therapy in hematological malignancies and strategies to overcome them. Expert Opin Biol Ther 2016; 16:1093-104. [DOI: 10.1080/14712598.2016.1190828] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
39
|
Abstract
One of the strategies to enhance immune response against tumors has been the use of vaccines against tumor-associated antigens (TAAs). MUC1 is a TAA that is overexpressed in many malignancies being linked to worse prognosis. Moreover, tumor MUC1 is hypoglycosylated revealing new epitopes that are antigenic and potential T-cell targets. TG4010 is a recombinant viral vaccine targeting MUC1, also encoding for IL-2. TG4010 has been tested in Phase I-II trials demonstrating a consistent safety profile with mild local reactions as main side effect. These studies have confirmed immune responses to the vaccine product. Clinical efficacy has been observed mainly in patients with non-small-cell lung cancer in combination with chemotherapy. Peripheral activated NK cells are currently being validated as biomarkers of response.
Collapse
Affiliation(s)
- Edurne Arriola
- Southampton NIHR Experimental Cancer Medicine Centre, Faculty of Medicine, University of Southampton Tremona Road, Southampton SO16 6YD, UK
- University Hospital Southampton NHS Foundation Trust, Tremona Road, Southampton SO16 6YD, UK
| | - Christian Ottensmeier
- Southampton NIHR Experimental Cancer Medicine Centre, Faculty of Medicine, University of Southampton Tremona Road, Southampton SO16 6YD, UK
- University Hospital Southampton NHS Foundation Trust, Tremona Road, Southampton SO16 6YD, UK
| |
Collapse
|
40
|
Fang F, Zhao Q, Sui Z, Liang Y, Jiang H, Yang K, Liang Z, Zhang L, Zhang Y. Glycan Moieties as Bait to Fish Plasma Membrane Proteins. Anal Chem 2016; 88:5065-71. [DOI: 10.1021/acs.analchem.6b01082] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Fei Fang
- National Chromatographic R. & A. Center, Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Science, Dalian, Liaoning 116023, China
- University of Chinese Academy of Sciences, Beijing 100039, China
| | - Qun Zhao
- National Chromatographic R. & A. Center, Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Science, Dalian, Liaoning 116023, China
| | - Zhigang Sui
- National Chromatographic R. & A. Center, Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Science, Dalian, Liaoning 116023, China
| | - Yu Liang
- National Chromatographic R. & A. Center, Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Science, Dalian, Liaoning 116023, China
| | - Hao Jiang
- National Chromatographic R. & A. Center, Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Science, Dalian, Liaoning 116023, China
- University of Chinese Academy of Sciences, Beijing 100039, China
| | - Kaiguang Yang
- National Chromatographic R. & A. Center, Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Science, Dalian, Liaoning 116023, China
| | - Zhen Liang
- National Chromatographic R. & A. Center, Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Science, Dalian, Liaoning 116023, China
| | - Lihua Zhang
- National Chromatographic R. & A. Center, Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Science, Dalian, Liaoning 116023, China
| | - Yukui Zhang
- National Chromatographic R. & A. Center, Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Science, Dalian, Liaoning 116023, China
| |
Collapse
|
41
|
Lakshminarayanan V, Supekar NT, Wei J, McCurry DB, Dueck AC, Kosiorek HE, Trivedi PP, Bradley JM, Madsen CS, Pathangey LB, Hoelzinger DB, Wolfert MA, Boons GJ, Cohen PA, Gendler SJ. MUC1 Vaccines, Comprised of Glycosylated or Non-Glycosylated Peptides or Tumor-Derived MUC1, Can Circumvent Immunoediting to Control Tumor Growth in MUC1 Transgenic Mice. PLoS One 2016; 11:e0145920. [PMID: 26788922 PMCID: PMC4720451 DOI: 10.1371/journal.pone.0145920] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2015] [Accepted: 12/05/2015] [Indexed: 01/21/2023] Open
Abstract
It remains challenging to produce decisive vaccines against MUC1, a tumor-associated antigen widely expressed by pancreas, breast and other tumors. Employing clinically relevant mouse models, we ruled out such causes as irreversible T-cell tolerance, inadequate avidity, and failure of T-cells to recognize aberrantly glycosylated tumor MUC1. Instead, every tested MUC1 preparation, even non-glycosylated synthetic 9mer peptides, induced interferon gamma-producing CD4+ and CD8+ T-cells that recognized glycosylated variants including tumor-associated MUC1. Vaccination with synthetic peptides conferred protection as long as vaccination was repeated post tumor challenge. Failure to revaccinate post challenge was associated with down-regulated tumor MUC1 and MHC molecules. Surprisingly, direct admixture of MUC1-expressing tumor with MUC1-hyperimmune T-cells could not prevent tumor outgrowth or MUC1 immunoediting, whereas ex vivo activation of the hyperimmune T-cells prior to tumor admixture rendered them curative. Therefore, surrogate T-cell preactivation outside the tumor bed, either in culture or by repetitive vaccination, can overcome tumor escape.
Collapse
Affiliation(s)
- Vani Lakshminarayanan
- Department of Immunology, Mayo Clinic in Arizona, Scottsdale, AZ, United States of America
| | - Nitin T. Supekar
- Complex Carbohydrate Research Center, University of Georgia, Athens, GA, United States of America
| | - Jie Wei
- Department of Immunology, Mayo Clinic in Arizona, Scottsdale, AZ, United States of America
| | - Dustin B. McCurry
- Hematology/Oncology, Mayo Clinic in Arizona, Scottsdale, AZ, United States of America
| | - Amylou C. Dueck
- Biostatistics, Mayo Clinic in Arizona, Scottsdale, AZ, United States of America
| | - Heidi E. Kosiorek
- Biostatistics, Mayo Clinic in Arizona, Scottsdale, AZ, United States of America
| | - Priyanka P. Trivedi
- Department of Immunology, Mayo Clinic in Arizona, Scottsdale, AZ, United States of America
| | - Judy M. Bradley
- Department of Immunology, Mayo Clinic in Arizona, Scottsdale, AZ, United States of America
| | - Cathy S. Madsen
- Department of Immunology, Mayo Clinic in Arizona, Scottsdale, AZ, United States of America
| | - Latha B. Pathangey
- Department of Immunology, Mayo Clinic in Arizona, Scottsdale, AZ, United States of America
| | | | - Margreet A. Wolfert
- Complex Carbohydrate Research Center, University of Georgia, Athens, GA, United States of America
| | - Geert-Jan Boons
- Complex Carbohydrate Research Center, University of Georgia, Athens, GA, United States of America
- * E-mail: (SJG); (PAC); (GJB)
| | - Peter A. Cohen
- Department of Immunology, Mayo Clinic in Arizona, Scottsdale, AZ, United States of America
- Hematology/Oncology, Mayo Clinic in Arizona, Scottsdale, AZ, United States of America
- * E-mail: (SJG); (PAC); (GJB)
| | - Sandra J. Gendler
- Department of Immunology, Mayo Clinic in Arizona, Scottsdale, AZ, United States of America
- Department of Biochemistry/Molecular Biology, Mayo Clinic in Arizona, Scottsdale, AZ, United States of America
- Hematology/Oncology, Mayo Clinic in Arizona, Scottsdale, AZ, United States of America
- * E-mail: (SJG); (PAC); (GJB)
| |
Collapse
|
42
|
Rivalland G, Loveland B, Mitchell P. Update on Mucin-1 immunotherapy in cancer: a clinical perspective. Expert Opin Biol Ther 2015; 15:1773-87. [PMID: 26453294 DOI: 10.1517/14712598.2015.1088519] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
INTRODUCTION Mucin 1 (MUC1) is particularly well suited as a cancer immunotherapy target due to the elevated protein expression and aberrant forms associated with malignancy. A variety of therapeutic strategies have been explored, including antibodies intended to induce cancer cell destruction, and vaccinations with peptides, tumor extracts, and gene expression systems. AREAS COVERED MUC1 immunotherapeutic strategies have included vaccination with peptide sequences, glycan molecules, viruses, and dendritic cells, monoclonal antibodies and monoclonal antibody conjugates. Here we review the relevant clinical trials in each field of immunotherapy with particular focus on large and recently published trials. EXPERT OPINION Long clinical experience in the trial setting has reduced concerns of immunotherapy associated toxicities and inappropriate immune responses, with the main limitation (common to many experimental approaches) being a lack of clinical efficacy. However, there have been sufficient treatment-associated responses to justify continued pursuit of MUC1 targeted immunotherapies. The focus now should be on application to the relevant cancers under appropriate circumstances and combination with the emerging non-specific immunotherapy approaches such as the PD-1 pathway inhibitors.
Collapse
Affiliation(s)
- Gareth Rivalland
- a 1 Austin Health, Olivia Newton-John Cancer and Wellness Centre , Studley Rd, Heidelberg VIC 3084, Australia
| | - Bruce Loveland
- b 2 Burnet Institute, Centre for Biomedical Research , Melbourne VIC 3004, Australia
| | - Paul Mitchell
- c 3 Austin Health, Level 4, Olivia Newton-John Cancer and Wellness Centre , Studley Rd, Heidelberg VIC 3084, Australia +613 94 96 57 63 ; +613 94 57 66 98 ;
| |
Collapse
|
43
|
Lendvai N, Cohen AD, Cho HJ. Beyond consolidation: auto-SCT and immunotherapy for plasma cell myeloma. Bone Marrow Transplant 2015; 50:770-80. [PMID: 25751647 DOI: 10.1038/bmt.2015.5] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2014] [Accepted: 12/31/2014] [Indexed: 12/15/2022]
Abstract
Autologous hematopoietic cell transplantation (auto-HCT) is the standard consolidation therapy for plasma cell myeloma patients following induction therapy. Auto-HCT improves disease-free survival (DFS), but is generally not curative. The allogeneic HCT experience demonstrated that T-cell immunotherapy can confer long-term DFS. Preclinical and clinical data indicate that myeloma-associated Ags elicit humoral and cellular immune responses (IRs) in myeloma patients. These findings strongly suggest that the immunotherapeutic strategies, including immune checkpoint inhibitors, therapeutic cancer vaccines and adoptive cellular therapies, are promising avenues of clinical research that may be most applicable in the minimal residual disease state following auto-HCT. These strategies are designed to prime or augment antimyeloma IRs and promote a 'host-vs-myeloma' effect that may result in durable DFS. Innovative clinical trials investigating immune checkpoint inhibitors and cancer vaccines have demonstrated that robust immunity against myeloma-associated Ags can be elicited in the setting of auto-HCT. A diverse array of immunotherapeutic strategies have entered clinical trials in myeloma, including PD-1/PD-L1 inhibitors, DC/myeloma cell fusion vaccines and adoptive chimeric Ag receptor T-cell therapy, and further investigation of combinations of immunologic and pharmaceutical agents are expected in the near future. In this review, we will discuss the preclinical data supporting immunotherapy in auto-HCT for myeloma, clinical investigation of these strategies and the future prospects of immunotherapy in pursuit of the goal of curative therapy.
Collapse
Affiliation(s)
- N Lendvai
- 1] Myeloma Service, Memorial Sloan-Kettering Cancer Center, New York, NY, USA [2] Department of Medicine, Weill Medical College of Cornell University, New York, NY, USA
| | - A D Cohen
- Division of Hematology/Oncology, Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
| | - H J Cho
- Multiple Myeloma Service, Division of Hematology and Medical Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
44
|
Abstract
Signal peptide (SP) domains have a common motif but also sequence specific features. This knowledge was mainly ignored by immunologists who considered SP as generic, short-lived, targeting sequences. Consequently, while SP-derived MHC class I, class II and HLA-E epitopes have been isolated, their use as antigen-specific vaccine candidates (VCs) was mostly neglected. Recently we demonstrated the rational of selecting entire SP domains as multi-epitope long peptide VCs based on their high T and B-cell epitope densities. This review summarizes preclinical and clinical results demonstrating the various advantages of human SP domain VCs derived from both bacterial and tumor antigens. Such vaccine design provides for a straightforward, yet unique immunotherapeutic means of generating robust, non-toxic, diversified, combined antigen-specific CD4+/CD8+ T/B-cell immunity, irrespective of patient HLA repertoire also in disease associated transporter-associated with antigen processing (TAP) deficiencies. Subsequent clinical trials will further assess the full potential of this approach.
Collapse
Key Words
- ADCC, antibody-dependent cell-mediated cytotoxicity
- AE, adverse events
- APC, antigen presenting cells
- DC, dendritic cells
- ER, endoplasmic reticulum
- ImMucin
- LP, long peptide
- MHC
- MHC, major histocompatibility complex
- MM, multiple myeloma
- MUC1
- PBMC, peripheral blood mononuclear cells
- SP, signal peptide
- SPP, signal peptide peptidase
- SPase, signal peptidase
- T-cell
- TAA, tumor associated antigen
- TAP, transporter-associated with antigen processing
- VC, vaccine candidate
- antibodies
- cancer
- hGM-CSF, human granulocyte-macrophage colony-stimulating factor
- long peptide
- signal peptide
- tuberculosis
- vaccine
Collapse
|