1
|
Zhang D, Jiang L, Yu F, Yan P, Liu Y, Wu Y, Yang X. PepT1-targeted nanodrug based on co-assembly of anti-inflammatory peptide and immunosuppressant for combined treatment of acute and chronic DSS-induced ColitiS. Front Pharmacol 2024; 15:1442876. [PMID: 39211778 PMCID: PMC11357942 DOI: 10.3389/fphar.2024.1442876] [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: 06/03/2024] [Accepted: 07/31/2024] [Indexed: 09/04/2024] Open
Abstract
Introduction Inflammatory bowel disease (IBD), as a chronic and recurrent inflammatory bowel diseases with limited therapeutic outcomes, is characterized by immune disorders and intestinal barrier dysfunction. Currently, the most medications used to cure IBD in clinic just temporarily induce and maintain remission with poor response rates and limited outcomes. Therefore, it is urgently necessary to develop an appropriate therapeutic candidate with preferable efficacy and less adverse reaction for curing IBD. Methods Five groups of mice were utilized: control that received saline, DSS group (mice received 2.5% DSS or 4% DSS), KPV group (mice received KPV), FK506 group (mice received FK506) and NPs groups (mice received NPs). The effect of NP on the inflammatory factors of macrophage was evaluated using CCK-8, quantitative polymerase chain reaction (PCR), Elisa and Western blot (WB). Immunofluorescent staining revealed the targeting relationship between NP and Petp-1. Immunohistochemistry staining showed the effect of NP on tight junction proteins. Moreover, in vivo animal experiments confirmed that NPs reduced inflammatory levels in IBD. Results and Discussion After administering with NPs, mice with DSS-induced acute or chronic colitis exhibited significant improvement in body weight, colon length, and disease activity index, decreased the level of the factors associated with oxidative stress (MPO, NO and ROS) and the inflammatory cytokines (TNF-α, IL-1β and IL-6), which implied that NPs could ameliorate murine colitis effectively. Furthermore, treating by NPs revealed a notable reduction of the expressions of CD68 and CD3, restoring the expression levels of tight junction proteins (Claudin-5, Occludin-1, and ZO-1) were significantly restored, surpassing those observed in the KPV and FK506 groups. which indicated that NPs can reduce inflammation and enhance epithelial barrier integrity by decreasing the infiltration of macrophages and T-lymphocytes. Collectively, those results demonstrated the effectively therapeutic outcome after using NPs in both acute and chronic colitis, suggesting that the newly co-assembled of NPs can be as a potential therapeutic candidate for colitis.
Collapse
Affiliation(s)
- Daifang Zhang
- Department of Cardiovascular Surgery, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- Metabolic Vascular Disease Key Laboratory of Sichuan Province, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Longqi Jiang
- Metabolic Vascular Disease Key Laboratory of Sichuan Province, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- Department of Vascular Surgery, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Fengxu Yu
- Department of Cardiovascular Surgery, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- Metabolic Vascular Disease Key Laboratory of Sichuan Province, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Pijun Yan
- Metabolic Vascular Disease Key Laboratory of Sichuan Province, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Yong Liu
- Metabolic Vascular Disease Key Laboratory of Sichuan Province, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- Department of Vascular Surgery, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- Key Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province (Collaborative Innovation Center for Prevention of Cardiovascular Diseases), Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China
| | - Ya Wu
- Metabolic Vascular Disease Key Laboratory of Sichuan Province, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- Department of Vascular Surgery, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- Key Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province (Collaborative Innovation Center for Prevention of Cardiovascular Diseases), Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China
| | - Xi Yang
- Department of Vascular Surgery, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- Key Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province (Collaborative Innovation Center for Prevention of Cardiovascular Diseases), Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China
| |
Collapse
|
2
|
Wang QT, Liu YX, Wang J, Wang H. Advances in Cancer Nanovaccines: Harnessing Nanotechnology for Broadening Cancer Immune Response. ChemMedChem 2023; 18:e202200673. [PMID: 37088719 DOI: 10.1002/cmdc.202200673] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 04/20/2023] [Accepted: 04/20/2023] [Indexed: 04/25/2023]
Abstract
Many advances have been made recently in the field of cancer immunotherapy, particularly with the development of treatments such as immune checkpoint inhibitors and adoptive cellular immunotherapy. The efficacy of immunotherapy is limited, however, owing to high levels of tumor heterogeneity and the immunosuppressive environments of advanced malignant tumors. Therefore, therapeutic anticancer vaccines have gradually become powerful tools for inducing valid antitumor immune responses and regulating the immune microenvironment. Tumor vaccines loaded in nanocarriers have become an indispensable delivery platform for tumor treatment because of their enhanced stability, targeting capability, and high level of safety. Through a unique design, cancer nanovaccines activate innate immunity and tumor-specific immunity simultaneously. For example, the design of cancer vaccines can incorporate strategies such as enhancing the stability and targeting of tumor antigens, combining effective adjuvants, cytokines, and immune microenvironment regulators, and promoting the maturation and cross-presentation of antigen-presenting cells (APCs). In this review, we discuss the design and preparation of nanovaccines for remodeling tumor antigen immunogenicity and regulating the immunosuppressive microenvironment.
Collapse
Affiliation(s)
- Qian-Ting Wang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology (NCNST), Beijing, 100190, P. R. China
| | - Yi-Xuan Liu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology (NCNST), Beijing, 100190, P. R. China
- University of the Chinese Academy of Sciences (UCAS), Beijing, 100049, P. R. China
| | - Jie Wang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology (NCNST), Beijing, 100190, P. R. China
| | - Hao Wang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology (NCNST), Beijing, 100190, P. R. China
| |
Collapse
|
3
|
Shanshal M, Caimi PF, Adjei AA, Ma WW. T-Cell Engagers in Solid Cancers-Current Landscape and Future Directions. Cancers (Basel) 2023; 15:2824. [PMID: 37345160 DOI: 10.3390/cancers15102824] [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: 04/05/2023] [Revised: 05/15/2023] [Accepted: 05/15/2023] [Indexed: 06/23/2023] Open
Abstract
Monoclonal antibody treatment initially heralded an era of molecularly targeted therapy in oncology and is now widely applied in modulating anti-cancer immunity by targeting programmed cell receptors (PD-1, PD-L1), cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) and, more recently, lymphocyte-activation gene 3 (LAG3). Chimeric antigen receptor T-cell therapy (CAR-T) recently proved to be a valid approach to inducing anti-cancer immunity by directly modifying the host's immune cells. However, such cell-based therapy requires extensive resources such as leukapheresis, ex vivo modification and expansion of cytotoxic T-cells and current Good Manufacturing Practice (cGMP) laboratories and presents significant logistical challenges. Bi-/trispecific antibody technology is a novel pharmaceutical approach to facilitate the engagement of effector immune cells to potentially multiple cancer epitopes, e.g., the recently approved blinatumomab. This opens the opportunity to develop 'off-the-shelf' anti-cancer agents that achieve similar and/or complementary anti-cancer effects as those of modified immune cell therapy. The majority of bi-/trispecific antibodies target the tumor-associated antigens (TAA) located on the extracellular surface of cancer cells. The extracellular antigens represent just a small percentage of known TAAs and are often associated with higher toxicities because some of them are expressed on normal cells (off-target toxicity). In contrast, the targeting of intracellular TAAs such as mutant RAS and TP53 may lead to fewer off-target toxicities while still achieving the desired antitumor efficacy (on-target toxicity). Here, we provide a comprehensive review on the emerging field of bi-/tri-specific T-cell engagers and potential therapeutic opportunities.
Collapse
Affiliation(s)
| | | | | | - Wen Wee Ma
- Cleveland Clinic, Cleveland, OH 44195, USA
| |
Collapse
|
4
|
Kumbhari A, Rose D, Lee PP, Kim PS. A minimal model of T cell avidity may identify subtherapeutic vaccine schedules. Math Biosci 2021; 334:108556. [PMID: 33539903 DOI: 10.1016/j.mbs.2021.108556] [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: 12/08/2020] [Revised: 01/24/2021] [Accepted: 01/25/2021] [Indexed: 11/17/2022]
Abstract
T cells protect the body from cancer by recognising tumour-associated antigens. Recognising these antigens depends on multiple factors, one of which is T cell avidity, i.e., the total interaction strength between a T cell and a cancer cell. While both high- and low-avidity T cells can kill cancer cells, durable anti-cancer immune responses require the selection of high-avidity T cells. Previous experimentation with anti-cancer vaccines, however, has shown that most vaccines elicit low-avidity T cells. Optimising vaccine schedules may remedy this by preferentially selecting high-avidity T cells. Here, we use mathematical modelling to develop a simple, phenomenological model of avidity selection that may identify vaccine schedules that disproportionately favour low-avidity T cells. We calibrate our model to our prior, more complex model, and then validate it against several experimental data sets. We find that the sensitivity of the model's parameters change with vaccine dosage, which allows us to use a patient's data and clinical history to screen for suitable vaccine strategies.
Collapse
Affiliation(s)
- Adarsh Kumbhari
- School of Mathematics and Statistics, University of Sydney, Sydney, NSW, Australia
| | - Danya Rose
- School of Mathematics and Statistics, University of Sydney, Sydney, NSW, Australia
| | - Peter P Lee
- Department of Immuno-Oncology, City of Hope and Beckman Research Institute, Duarte, CA, USA
| | - Peter S Kim
- School of Mathematics and Statistics, University of Sydney, Sydney, NSW, Australia.
| |
Collapse
|
5
|
Hashemzadeh N, Dolatkhah M, Adibkia K, Aghanejad A, Barzegar-Jalali M, Omidi Y, Barar J. Recent advances in breast cancer immunotherapy: The promising impact of nanomedicines. Life Sci 2021; 271:119110. [PMID: 33513401 DOI: 10.1016/j.lfs.2021.119110] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 01/13/2021] [Accepted: 01/20/2021] [Indexed: 12/29/2022]
Abstract
Breast cancer (BC) is one of the prevalent cancers among women. Generally, the treatment of BC is mostly based on several prominent strategies, including chemotherapy, surgery, endocrine therapy, molecular targeted therapy, and radiation. Owing to the growing knowledge about the complexity of BC pathobiology, immunotherapy as a promising treatment modality has substantially improved the patients' care in the clinic. Immunotherapy is used to harness the patient's immune system to recognize and battle devious cancer cells. As a novel therapy approach, this emerging strategy targets the key molecular entities of tumor tissue. To achieve maximal therapeutic impacts, the dynamic interplay between cancer and immune cells needs to be fully comprehended. The key molecular machinery of solid tumors can be targeted by nanoscale immunomedicines. While discussing the potential biomarkers involved in the initiation and progression of BC, we aimed to provide comprehensive insights into the immunotherapy and articulate the recent advances in terms of the therapeutic strategies used to control this disease, including immune checkpoint inhibitors, vaccines, chimeric antigen receptor T cells therapy, and nanomedicines.
Collapse
Affiliation(s)
- Nastaran Hashemzadeh
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran; Student Research Committee, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Pharmaceutics, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mitra Dolatkhah
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran; Student Research Committee, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Pharmaceutics, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Khosro Adibkia
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Pharmaceutics, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ayuob Aghanejad
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Barzegar-Jalali
- Department of Pharmaceutics, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Yadollah Omidi
- Department of Pharmaceutical Sciences, College of Pharmacy, Nova Southeastern University, Fort Lauderdale, FL 33328, USA
| | - Jaleh Barar
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Pharmaceutics, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
6
|
Buonaguro L, Tagliamonte M. Selecting Target Antigens for Cancer Vaccine Development. Vaccines (Basel) 2020; 8:vaccines8040615. [PMID: 33080888 PMCID: PMC7711972 DOI: 10.3390/vaccines8040615] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 10/12/2020] [Accepted: 10/13/2020] [Indexed: 12/11/2022] Open
Abstract
One of the principal goals of cancer immunotherapy is the development of efficient therapeutic cancer vaccines that are able to elicit an effector as well as memory T cell response specific to tumor antigens. In recent years, the attention has been focused on the personalization of cancer vaccines. However, the efficacy of therapeutic cancer vaccines is still disappointing despite the large number of vaccine strategies targeting different tumors that have been evaluated in recent years. While the preclinical data have frequently shown encouraging results, clinical trials have not provided satisfactory data to date. The main reason for such failures is the complexity of identifying specific target tumor antigens that should be unique or overexpressed only by the tumor cells compared to normal cells. Most of the tumor antigens included in cancer vaccines are non-mutated overexpressed self-antigens, eliciting mainly T cells with low-affinity T cell receptors (TCR) unable to mediate an effective anti-tumor response. In this review, the target tumor antigens employed in recent years in the development of therapeutic cancer vaccine strategies are described, along with potential new classes of tumor antigens such as the human endogenous retroviral elements (HERVs), unconventional antigens, and/or heteroclitic peptides.
Collapse
|
7
|
Mu W, Chu Q, Liu Y, Zhang N. A Review on Nano-Based Drug Delivery System for Cancer Chemoimmunotherapy. NANO-MICRO LETTERS 2020; 12:142. [PMID: 34138136 PMCID: PMC7770879 DOI: 10.1007/s40820-020-00482-6] [Citation(s) in RCA: 132] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/26/2020] [Accepted: 06/11/2020] [Indexed: 05/11/2023]
Abstract
Although notable progress has been made on novel cancer treatments, the overall survival rate and therapeutic effects are still unsatisfactory for cancer patients. Chemoimmunotherapy, combining chemotherapeutics and immunotherapeutic drugs, has emerged as a promising approach for cancer treatment, with the advantages of cooperating two kinds of treatment mechanism, reducing the dosage of the drug and enhancing therapeutic effect. Moreover, nano-based drug delivery system (NDDS) was applied to encapsulate chemotherapeutic agents and exhibited outstanding properties such as targeted delivery, tumor microenvironment response and site-specific release. Several nanocarriers have been approved in clinical cancer chemotherapy and showed significant improvement in therapeutic efficiency compared with traditional formulations, such as liposomes (Doxil®, Lipusu®), nanoparticles (Abraxane®) and micelles (Genexol-PM®). The applications of NDDS to chemoimmunotherapy would be a powerful strategy for future cancer treatment, which could greatly enhance the therapeutic efficacy, reduce the side effects and optimize the clinical outcomes of cancer patients. Herein, the current approaches of cancer immunotherapy and chemoimmunotherapy were discussed, and recent advances of NDDS applied for chemoimmunotherapy were further reviewed.
Collapse
Affiliation(s)
- Weiwei Mu
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Wenhuaxi Road, Jinan, 250012, People's Republic of China
| | - Qihui Chu
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Wenhuaxi Road, Jinan, 250012, People's Republic of China
| | - Yongjun Liu
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Wenhuaxi Road, Jinan, 250012, People's Republic of China
| | - Na Zhang
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Wenhuaxi Road, Jinan, 250012, People's Republic of China.
| |
Collapse
|
8
|
Fromer MW, Scoggins CR. Article Commentary: Cancer Immunotherapy for the General Surgeon. Am Surg 2020. [DOI: 10.1177/000313482008600423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Progress in the arena of cancer immunotherapy has been immense in recent years. The fact remains that most of the cancer resections in the United States are performed by general surgeons and not oncologic specialists. A busy practice in general surgery will invariably make it difficult to keep pace with such rapid advancement. This review offers a concise summary of the major concepts and trials that have driven the immunotherapy revolution and their implications for surgeons who deliver cancer care.
Collapse
Affiliation(s)
- Marc W. Fromer
- From the Division of Surgical Oncology, Hiram C. Polk, Jr., MD Department of Surgery, University of Louisville School of Medicine, Louisville, Kentucky
| | - Charles R. Scoggins
- From the Division of Surgical Oncology, Hiram C. Polk, Jr., MD Department of Surgery, University of Louisville School of Medicine, Louisville, Kentucky
| |
Collapse
|
9
|
Kumbhari A, Kim PS, Lee PP. Optimisation of anti-cancer peptide vaccines to preferentially elicit high-avidity T cells. J Theor Biol 2020; 486:110067. [DOI: 10.1016/j.jtbi.2019.110067] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Revised: 09/24/2019] [Accepted: 11/01/2019] [Indexed: 10/25/2022]
|
10
|
van den Bulk J, Verdegaal EM, de Miranda NF. Cancer immunotherapy: broadening the scope of targetable tumours. Open Biol 2019; 8:rsob.180037. [PMID: 29875199 PMCID: PMC6030119 DOI: 10.1098/rsob.180037] [Citation(s) in RCA: 149] [Impact Index Per Article: 29.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Accepted: 05/11/2018] [Indexed: 12/12/2022] Open
Abstract
Cancer immunotherapy has experienced remarkable advances in recent years. Striking clinical responses have been achieved for several types of solid cancers (e.g. melanoma, non-small cell lung cancer, bladder cancer and mismatch repair-deficient cancers) after treatment of patients with T-cell checkpoint blockade therapies. These have been shown to be particularly effective in the treatment of cancers with high mutation burden, which places tumour-mutated antigens (neo-antigens) centre stage as targets of tumour immunity and cancer immunotherapy. With current technologies, neo-antigens can be identified in a short period of time, which may support the development of complementary, personalized approaches that increase the number of tumours amenable to immunotherapeutic intervention. In addition to reviewing the state of the art in cancer immunotherapy, we discuss potential avenues that can bring the immunotherapy revolution to a broader patient group including cancers with low mutation burden.
Collapse
|
11
|
Kissick HT. Is It Possible to Develop Cancer Vaccines to Neoantigens, What Are the Major Challenges, and How Can These Be Overcome? Neoantigens as Vaccine Targets for Cancer. Cold Spring Harb Perspect Biol 2018; 10:cshperspect.a033704. [PMID: 29254981 DOI: 10.1101/cshperspect.a033704] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Recent work by several groups has undoubtedly shown that we can produce cancer vaccines targeting neoantigens. However, each vaccine is essentially a single-use, patient-specific product, making this approach resource-intensive. For this reason, it is important to ask whether this approach will be any more successful than what has been attempted during the last 30 years using vaccines targeting self-epitopes. Here, we discuss what might be expected from neoantigen vaccines based on our experience in chronic viral infections, and how this new approach may be applied to cancer immunotherapy.
Collapse
Affiliation(s)
- Haydn T Kissick
- Departments of Urology and Microbiology and Immunology, Emory University School of Medicine, Atlanta, Georgia 30322
| |
Collapse
|
12
|
Hos BJ, Tondini E, van Kasteren SI, Ossendorp F. Approaches to Improve Chemically Defined Synthetic Peptide Vaccines. Front Immunol 2018; 9:884. [PMID: 29755468 PMCID: PMC5932164 DOI: 10.3389/fimmu.2018.00884] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Accepted: 04/10/2018] [Indexed: 12/22/2022] Open
Abstract
Progress made in peptide-based vaccinations to induce T-cell-dependent immune responses against cancer has invigorated the search for optimal vaccine modalities. Design of new vaccine strategies intrinsically depends on the knowledge of antigen handling and optimal epitope presentation in both major histocompatibility complex class I and -II molecules by professional antigen-presenting cells to induce robust CD8 and CD4 T-cell responses. Although there is a steady increase in the understanding of the underlying mechanisms that bridges innate and adaptive immunology, many questions remain to be answered. Moreover, we are in the early stage of exploiting this knowledge to clinical advantage. Several adaptations of peptide-based vaccines like peptide-adjuvant conjugates have been explored and showed beneficial outcomes in preclinical models; but in the clinical trials conducted so far, mixed results were obtained. A major limiting factor to unravel antigen handling mechanistically is the lack of tools to efficiently track peptide vaccines at the molecular and (sub)cellular level. In this mini-review, we will discuss options to develop molecular tools for improving, as well as studying, peptide-based vaccines.
Collapse
Affiliation(s)
- Brett J Hos
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, Netherlands
| | - Elena Tondini
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, Netherlands
| | - Sander I van Kasteren
- Leiden Institute of Chemistry, The Institute for Chemical Immunology, Leiden University, Leiden, Netherlands
| | - Ferry Ossendorp
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, Netherlands
| |
Collapse
|
13
|
Therapeutic cancer vaccines: From initial findings to prospects. Immunol Lett 2018; 196:11-21. [DOI: 10.1016/j.imlet.2018.01.011] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2017] [Revised: 12/30/2017] [Accepted: 01/24/2018] [Indexed: 12/15/2022]
|
14
|
Patel PM, Ottensmeier CH, Mulatero C, Lorigan P, Plummer R, Pandha H, Elsheikh S, Hadjimichael E, Villasanti N, Adams SE, Cunnell M, Metheringham RL, Brentville VA, Machado L, Daniels I, Gijon M, Hannaman D, Durrant LG. Targeting gp100 and TRP-2 with a DNA vaccine: Incorporating T cell epitopes with a human IgG1 antibody induces potent T cell responses that are associated with favourable clinical outcome in a phase I/II trial. Oncoimmunology 2018; 7:e1433516. [PMID: 29872563 PMCID: PMC5980353 DOI: 10.1080/2162402x.2018.1433516] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Revised: 01/22/2018] [Accepted: 01/22/2018] [Indexed: 02/08/2023] Open
Abstract
A DNA vaccine, SCIB1, incorporating two CD8 and two CD4 epitopes from TRP-2/gp100 was evaluated in patients with metastatic melanoma. Each patient received SCIB1 via intramuscular injection with electroporation. The trial was designed to find the safest dose of SCIB1 which induced immune/clinical responses in patients with or without tumour. Fifteen patients with tumor received SCIB1 doses of 0.4-8 mg whilst 20 fully-resected patients received 2-8 mg doses. Twelve patients elected to continue immunization every 3 months for up to 39 months. SCIB1 induced dose-dependent T cell responses in 88% of patients with no serious adverse effects or dose limiting toxicities. The intensity of the T cell responses was significantly higher in patients receiving 4 mg doses without tumor when compared to those with tumor (p < 0.01). In contrast, patients with tumor showed a significantly higher response to the 8 mg dose than the 4 mg dose (p < 0.03) but there was no significant difference in the patients without tumor. One of 15 patients with measurable disease showed an objective tumor response and 7/15 showed stable disease. 5/20 fully-resected patients have experienced disease recurrence but all remained alive at the cut-off date with a median observation time of 37 months. A positive clinical outcome was associated with MHC-I and MHC-II expression on tumors prior to therapy (p = 0.027). We conclude that SCIB1 is well tolerated and stimulates potent T cell responses in melanoma patients. It deserves further evaluation as a single agent adjuvant therapy or in combination with checkpoint inhibitors in advanced disease.
Collapse
Affiliation(s)
- Poulam M Patel
- Academic Department of Clinical Oncology, Division of Cancer & Stem Cells, University of Nottingham, Nottingham, UK
| | - Christian H Ottensmeier
- Southampton Experimental Cancer Medicine Centre and Southampton University Hospitals, Faculty of Medicine, Southampton, UK
| | | | - Paul Lorigan
- Institute of Cancer Sciences, University of Manchester, The Christie NHS Foundation Trust, Manchester, UK
| | - Ruth Plummer
- Northern Institute for Cancer Research, Medical School, University of Newcastle-upon-Tyne and Wear, UK
| | - Hardev Pandha
- Faculty of Health and Medical Sciences, University of Surrey, Guildford, Surrey, UK
| | - Somaia Elsheikh
- University of Nottingham, School of Medicine Queen's Medical Centre, Nottingham, UK
| | | | - Naty Villasanti
- University of Nottingham, School of Medicine Queen's Medical Centre, Nottingham, UK
| | - Sally E Adams
- Scancell Limited, Academic Department of Clinical Oncology, University of Nottingham, Nottingham, UK
| | - Michelle Cunnell
- Academic Department of Clinical Oncology, Division of Cancer & Stem Cells, University of Nottingham, Nottingham, UK
| | - Rachael L Metheringham
- Scancell Limited, Academic Department of Clinical Oncology, University of Nottingham, Nottingham, UK
| | - Victoria A Brentville
- Scancell Limited, Academic Department of Clinical Oncology, University of Nottingham, Nottingham, UK
| | - Lee Machado
- Scancell Limited, Academic Department of Clinical Oncology, University of Nottingham, Nottingham, UK
| | - Ian Daniels
- Scancell Limited, Academic Department of Clinical Oncology, University of Nottingham, Nottingham, UK
| | - Mohamed Gijon
- Scancell Limited, Academic Department of Clinical Oncology, University of Nottingham, Nottingham, UK
| | | | - Lindy G Durrant
- Academic Department of Clinical Oncology, Division of Cancer & Stem Cells, University of Nottingham, Nottingham, UK.,Scancell Limited, Academic Department of Clinical Oncology, University of Nottingham, Nottingham, UK
| |
Collapse
|
15
|
Kupcova Skalnikova H, Cizkova J, Cervenka J, Vodicka P. Advances in Proteomic Techniques for Cytokine Analysis: Focus on Melanoma Research. Int J Mol Sci 2017; 18:E2697. [PMID: 29236046 PMCID: PMC5751298 DOI: 10.3390/ijms18122697] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Revised: 12/07/2017] [Accepted: 12/08/2017] [Indexed: 12/16/2022] Open
Abstract
Melanoma is a skin cancer with permanently increasing incidence and resistance to therapies in advanced stages. Reports of spontaneous regression and tumour infiltration with T-lymphocytes makes melanoma candidate for immunotherapies. Cytokines are key factors regulating immune response and intercellular communication in tumour microenvironment. Cytokines may be used in therapy of melanoma to modulate immune response. Cytokines also possess diagnostic and prognostic potential and cytokine production may reflect effects of immunotherapies. The purpose of this review is to give an overview of recent advances in proteomic techniques for the detection and quantification of cytokines in melanoma research. Approaches covered span from mass spectrometry to immunoassays for single molecule detection (ELISA, western blot), multiplex assays (chemiluminescent, bead-based (Luminex) and planar antibody arrays), ultrasensitive techniques (Singulex, Simoa, immuno-PCR, proximity ligation/extension assay, immunomagnetic reduction assay), to analyses of single cells producing cytokines (ELISpot, flow cytometry, mass cytometry and emerging techniques for single cell secretomics). Although this review is focused mainly on cancer and particularly melanoma, the discussed techniques are in general applicable to broad research field of biology and medicine, including stem cells, development, aging, immunology and intercellular communication.
Collapse
Affiliation(s)
- Helena Kupcova Skalnikova
- Laboratory of Applied Proteome Analyses, Institute of Animal Physiology and Genetics, Czech Academy of Sciences, Rumburska 89, 27721 Libechov, Czech Republic.
| | - Jana Cizkova
- Laboratory of Applied Proteome Analyses, Institute of Animal Physiology and Genetics, Czech Academy of Sciences, Rumburska 89, 27721 Libechov, Czech Republic.
- Department of Veterinary Sciences, Faculty of Agrobiology, Food and Natural Resources, Czech University of Life Sciences, Kamycka 129, 16500 Prague, Czech Republic.
| | - Jakub Cervenka
- Laboratory of Applied Proteome Analyses, Institute of Animal Physiology and Genetics, Czech Academy of Sciences, Rumburska 89, 27721 Libechov, Czech Republic.
- Department of Cell Biology, Faculty of Science, Charles University, Vinicna 7, 12843 Prague 4, Czech Republic.
| | - Petr Vodicka
- Laboratory of Applied Proteome Analyses, Institute of Animal Physiology and Genetics, Czech Academy of Sciences, Rumburska 89, 27721 Libechov, Czech Republic.
| |
Collapse
|
16
|
Ni D, Qing S, Ding H, Yue H, Yu D, Wang S, Luo N, Su Z, Wei W, Ma G. Biomimetically Engineered Demi-Bacteria Potentiate Vaccination against Cancer. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2017; 4:1700083. [PMID: 29051851 PMCID: PMC5644226 DOI: 10.1002/advs.201700083] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Revised: 04/21/2017] [Indexed: 05/04/2023]
Abstract
Failure in enhancing antigen immunogenicity has limited the development of cancer vaccine. Inspired by effective immune responses toward microorganisms, demi-bacteria (DB) from Bacillus are engineered as carriers for cancer vaccines. The explored hydrothermal treatment enables the Bacillus to preserve optimal pathogen morphology with intrinsic mannose receptor agonist. Meanwhile, the treated Bacillus can be further endowed with ideal hollow/porous structure for efficient accommodation of antigen and adjuvant, such as CpG. Therefore, this optimal engineered nanoarchitecture allows multiple immunostimulatory elements integrate in a pattern closely resembling that of bacterial pathogens. Such pathogen mimicry greatly enhances antigen uptake and cross-presentation, resulting in stronger immune activation suitable for cancer vaccines. Indeed, DB-based biomimetic vaccination in mice induces synergistic cellular and humoral immune responses, achieving potent therapeutic and preventive effects against cancer. Application of microorganism-sourced materials thus presents new opportunities for potent cancer therapy.
Collapse
Affiliation(s)
- Dezhi Ni
- State Key Laboratory of Biochemical EngineeringInstitute of Process EngineeringChinese Academy of Sciences1 North 2nd StreetZhongguancun, Haidian DistrictBeijing100190P. R. China
| | - Shuang Qing
- State Key Laboratory of Biochemical EngineeringInstitute of Process EngineeringChinese Academy of Sciences1 North 2nd StreetZhongguancun, Haidian DistrictBeijing100190P. R. China
- University of Chinese Academy of SciencesNo. 19A Yuquan RoadBeijing100049P. R. China
| | - Hui Ding
- University of Chinese Academy of SciencesNo. 19A Yuquan RoadBeijing100049P. R. China
| | - Hua Yue
- State Key Laboratory of Biochemical EngineeringInstitute of Process EngineeringChinese Academy of Sciences1 North 2nd StreetZhongguancun, Haidian DistrictBeijing100190P. R. China
| | - Di Yu
- Molecular Immunomodulation LaboratorySchool of Biomedical SciencesMonash UniversityClaytonVictoria3800Australia
| | - Shuang Wang
- State Key Laboratory of Biochemical EngineeringInstitute of Process EngineeringChinese Academy of Sciences1 North 2nd StreetZhongguancun, Haidian DistrictBeijing100190P. R. China
- University of Chinese Academy of SciencesNo. 19A Yuquan RoadBeijing100049P. R. China
| | - Nana Luo
- State Key Laboratory of Biochemical EngineeringInstitute of Process EngineeringChinese Academy of Sciences1 North 2nd StreetZhongguancun, Haidian DistrictBeijing100190P. R. China
| | - Zhiguo Su
- State Key Laboratory of Biochemical EngineeringInstitute of Process EngineeringChinese Academy of Sciences1 North 2nd StreetZhongguancun, Haidian DistrictBeijing100190P. R. China
| | - Wei Wei
- State Key Laboratory of Biochemical EngineeringInstitute of Process EngineeringChinese Academy of Sciences1 North 2nd StreetZhongguancun, Haidian DistrictBeijing100190P. R. China
| | - Guanghui Ma
- State Key Laboratory of Biochemical EngineeringInstitute of Process EngineeringChinese Academy of Sciences1 North 2nd StreetZhongguancun, Haidian DistrictBeijing100190P. R. China
- University of Chinese Academy of SciencesNo. 19A Yuquan RoadBeijing100049P. R. China
- Jiangsu National Synergetic Innovation Center for Advanced MaterialsNanjing211816P. R. China
| |
Collapse
|
17
|
|
18
|
Young GJ, Bi WL, Wu WW, Johanns TM, Dunn GP, Dunn IF. Management of intracranial melanomas in the era of precision medicine. Oncotarget 2017; 8:89326-89347. [PMID: 29179523 PMCID: PMC5687693 DOI: 10.18632/oncotarget.19223] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2016] [Accepted: 04/24/2017] [Indexed: 01/08/2023] Open
Abstract
Melanoma is the most lethal of skin cancers, in part because of its proclivity for rapid and distant metastasis. It is also potentially the most neurotropic cancer in terms of probability of CNS metastasis from the primary lesion. Despite surgical resection and radiotherapy, prognosis remains guarded for patients with brain metastases. Over the past five years, a new domain of personalized therapy has emerged for advanced melanoma patients with the introduction of BRAF and other MAP kinase pathway inhibitors, immunotherapy, and combinatory therapeutic strategies. By targeting critical cellular signaling pathways and unleashing the adaptive immune response against tumor antigens, a subset of melanoma patients have demonstrated remarkable responses to these treatments. Over time, acquired resistance to these modalities inexorably develops, providing new challenges to overcome. We review the rapidly evolving terrain for intracranial melanoma treatment, address likely and potential mechanisms of resistance, as well as evaluate promising future therapeutic approaches currently under clinical investigation.
Collapse
Affiliation(s)
- Grace J Young
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Wenya Linda Bi
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.,Department of Cancer Biology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Winona W Wu
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Tanner M Johanns
- Division of Medical Oncology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO, USA.,Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, St. Louis, MO, USA
| | - Gavin P Dunn
- Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, St. Louis, MO, USA.,Department of Neurological Surgery, Washington University School of Medicine, St. Louis, MO, USA
| | - Ian F Dunn
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
19
|
Kee D, McArthur G. Immunotherapy of melanoma. Eur J Surg Oncol 2017; 43:594-603. [DOI: 10.1016/j.ejso.2016.07.014] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2016] [Revised: 07/14/2016] [Accepted: 07/25/2016] [Indexed: 12/31/2022] Open
|
20
|
Merlo A, Dalla Santa S, Dolcetti R, Zanovello P, Rosato A. Reverse immunoediting: When immunity is edited by antigen. Immunol Lett 2016; 175:16-20. [PMID: 27131431 DOI: 10.1016/j.imlet.2016.04.015] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2016] [Accepted: 04/22/2016] [Indexed: 12/30/2022]
Abstract
Immune selective pressure occurring during cancer immunoediting shapes tumor features revealed at clinical presentation. However, in the "Escape" phase, the tumor itself has the chance to influence the immunological response. Therefore, the capacity of the immune response to sculpt the tumor characteristics is only one side of the coin and even the opposite is likely true, i.e. that an antigen can shape the immune response in a sort of "reverse immunoediting". This reciprocal modeling probably occurs continuously, whenever the immune system encounters a tumor/foreign antigen, and can be operative in the pathogen/immune system interplay, thus possibly permeating the protective immunity as a whole. In line with this view, the characterization of a T cell response as well as the design of both active and passive immunotherapy strategies should also take into account all Ag features (type, load and presentation). Overall, we suggest that the "reverse immunoediting" hypothesis could help to dissect the complex interplay between antigens and the immune repertoire, and to improve the outcome of immunotherapeutic approaches, where T cell responses are manipulated and reprogrammed.
Collapse
Affiliation(s)
- Anna Merlo
- Veneto Institute of Oncology IOV-IRCCS, Via Gattamelata, 64, 35128 Padua, Italy
| | - Silvia Dalla Santa
- Department of Surgery, Oncology and Gastroenterology, Via Gattamelata, 64, University of Padua, 35128 Padua, Italy
| | - Riccardo Dolcetti
- CRO-IRCCS, National Cancer Institute, Via F. Gallini, 2, 33081 Aviano, PN, Italy; University of Queensland Diamantina Institute, Translational Research Institute, Brisbane, Queensland 4102, Australia
| | - Paola Zanovello
- Veneto Institute of Oncology IOV-IRCCS, Via Gattamelata, 64, 35128 Padua, Italy; Department of Surgery, Oncology and Gastroenterology, Via Gattamelata, 64, University of Padua, 35128 Padua, Italy
| | - Antonio Rosato
- Veneto Institute of Oncology IOV-IRCCS, Via Gattamelata, 64, 35128 Padua, Italy; Department of Surgery, Oncology and Gastroenterology, Via Gattamelata, 64, University of Padua, 35128 Padua, Italy.
| |
Collapse
|
21
|
Makinen SR, Zhu Q, Davis HL, Weeratna RD. CpG-mediated augmentation of CD8+ T-cell responses in mice is attenuated by a water-in-oil emulsion (Montanide ISA-51) but enhanced by an oil-in-water emulsion (IDRI SE). Int Immunol 2016; 28:453-61. [PMID: 27055469 DOI: 10.1093/intimm/dxw017] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Accepted: 03/22/2016] [Indexed: 12/14/2022] Open
Abstract
Adjuvants are a key component in enhancing immunogenicity of vaccines and play a vital role in facilitating the induction of the correct type of immunity required for each vaccine to be optimally efficacious. Several different adjuvants are found in licensed vaccines, and many others are in pre-clinical or clinical testing. Agonists for TLRs are potent activators of the innate immune system and some, such as CpG (TLR9 agonist), are particularly good for promoting cellular immunity because of the induction of Th1 cytokines. Emulsions that have both delivery and adjuvant properties are classified as water-in-oil (W/O) or oil-in-water (O/W) formulations. The W/O emulsion Montanide ISA-51, often combined with CpG, has been widely tested in cancer vaccine clinical trials. Squalene-based O/W emulsions are in licensed influenza vaccines, and T-cell responses have been assessed pre-clinically. No clinical study has compared the two types of emulsions, and the continued use of W/O with CpG in cancer vaccines may be because the lack of single adjuvant controls has masked the interference issue. These findings may have important implications for the development of vaccines where T-cell immunity is considered essential, such as those for cancer and chronic infections. Using particulate (hepatitis B surface antigen) and soluble protein (ovalbumin) antigen, we show in mice that a W/O emulsion (ISA-51) abrogates CpG-mediated augmentation of CD8(+) T-cell responses, whereas a squalene-based O/W emulsion significantly enhanced them.
Collapse
Affiliation(s)
- Shawn R Makinen
- Pfizer Vaccine Immunotherapeutics, Ottawa Laboratories, 340 Terry Fox Drive, Suite 200, Ottawa, Ontario K2K 3A2, Canada
| | - Qin Zhu
- Pfizer Vaccine Immunotherapeutics, Ottawa Laboratories, 340 Terry Fox Drive, Suite 200, Ottawa, Ontario K2K 3A2, Canada
| | - Heather L Davis
- Pfizer Vaccine Immunotherapeutics, Ottawa Laboratories, 340 Terry Fox Drive, Suite 200, Ottawa, Ontario K2K 3A2, Canada
| | - Risini D Weeratna
- Pfizer Vaccine Immunotherapeutics, Ottawa Laboratories, 340 Terry Fox Drive, Suite 200, Ottawa, Ontario K2K 3A2, Canada
| |
Collapse
|
22
|
Schneble E, Clifton GT, Hale DF, Peoples GE. Peptide-Based Cancer Vaccine Strategies and Clinical Results. Methods Mol Biol 2016; 1403:797-817. [PMID: 27076168 DOI: 10.1007/978-1-4939-3387-7_46] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
Active cancer immunotherapy is an exciting and developing field in oncology research. Peptide vaccines, the use of isolated immunogenic tumor-associated antigen (TAA) epitopes to generate an anticancer immune response, are an attractive option as they are easily produced and administered with minimal toxicity. Multiple TAA-derived peptides have been identified and evaluated with various vaccine strategies currently in clinical testing. Research suggests that utilizing vaccines in patients with minimal-residual disease may be a more effective strategy compared to targeting patients with widely metastatic disease as it avoids the immune suppression and tolerance associated with higher volumes of more established disease. Clinical trials also suggest that vaccines may need to be tailored and administered to specific cancer subtypes to achieve maximum efficacy. Additionally, numerous immunomodulators now in research and development show potential synergy with peptide vaccines. Our group has focused on a simpler, single-peptide strategy largely from the HER2/neu protein. We will discuss our experience thus far as well as review other peptide vaccine strategies that have shown clinical efficacy.
Collapse
Affiliation(s)
- Erika Schneble
- Cancer Insight, LLC, 600 Navarro Street, Suite 500, San Antonio, TX, 78205, USA.
| | - G Travis Clifton
- Cancer Insight, LLC, 600 Navarro Street, Suite 500, San Antonio, TX, 78205, USA
- Department of Surgical Oncology, MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX, 77030, USA
| | - Diane F Hale
- Cancer Insight, LLC, 600 Navarro Street, Suite 500, San Antonio, TX, 78205, USA
- San Antonio Military Medical Center, 3551 Roger Brooke Drive, San Antonio, TX, 78234, USA
| | - George E Peoples
- Cancer Insight, LLC, 600 Navarro Street, Suite 500, San Antonio, TX, 78205, USA
| |
Collapse
|
23
|
Goyal S, Silk AW, Tian S, Mehnert J, Danish S, Ranjan S, Kaufman HL. Clinical Management of Multiple Melanoma Brain Metastases: A Systematic Review. JAMA Oncol 2015; 1:668-76. [PMID: 26181286 PMCID: PMC5726801 DOI: 10.1001/jamaoncol.2015.1206] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
IMPORTANCE The treatment of multiple brain metastases (MBM) from melanoma is controversial and includes surgical resection, stereotactic radiosurgery (SRS), and whole-brain radiation therapy (WBRT). Several new classes of agents have revolutionized the treatment of metastatic melanoma, allowing some subsets of patients to have long-term survival. Given this, management of MBM from melanoma is continually evolving. OBJECTIVE To review the current evidence regarding the treatment of MBM from melanoma. EVIDENCE REVIEW The PubMed database was searched using combinations of search terms and synonyms for melanoma, brain metastases, radiation, chemotherapy, immunotherapy, and targeted therapy published between January 1, 1995, and January 1, 2015. Articles were selected for inclusion on the basis of targeted keyword searches, manual review of bibliographies, and whether the article was a clinical trial, large observational study, or retrospective study focusing on melanoma brain metastases. Of 2243 articles initially identified, 110 were selected for full review. Of these, the most pertinent 73 articles were included. FINDINGS Patients with newly diagnosed MBM can be treated with various modalities, either alone or in combination. Level 1 evidence supports the use of SRS alone, WBRT, and SRS with WBRT. Although the addition of WBRT to SRS improves the overall brain relapse rate, WBRT has no significant impact on overall survival and has detrimental neurocognitive outcomes. Cytotoxic chemotherapy has largely been ineffective; targeted therapies and immunotherapies have been reported to have high response rates and deserve further attention in larger clinical trials. Further studies are needed to fully evaluate the efficacy of these novel regimens in combination with radiation therapy. CONCLUSIONS AND RELEVANCE At this time, the standard management for patients with MBM from melanoma includes SRS, WBRT, or a combination of both. Emerging data exist to support the notion that SRS in combination with targeted therapies or immune therapy may obviate the need for WBRT; prospective studies are required to fully evaluate the efficacy of these novel regimens in combination with radiation therapy.
Collapse
Affiliation(s)
- Sharad Goyal
- Department of Radiation Oncology, Rutgers Cancer Institute of New Jersey and Rutgers Robert Wood Johnson Medical School
| | - Ann W. Silk
- Division of Medical Oncology, Rutgers Cancer Institute of New Jersey and Rutgers Robert Wood Johnson Medical School
| | - Sibo Tian
- Department of Radiation Oncology, Rutgers Cancer Institute of New Jersey and Rutgers Robert Wood Johnson Medical School
| | - Janice Mehnert
- Division of Medical Oncology, Rutgers Cancer Institute of New Jersey and Rutgers Robert Wood Johnson Medical School
| | - Shabbar Danish
- Division of Surgical Oncology, Rutgers Cancer Institute of New Jersey and Rutgers Robert Wood Johnson Medical School
| | - Sinthu Ranjan
- Department of Radiation Oncology, Rutgers Cancer Institute of New Jersey and Rutgers Robert Wood Johnson Medical School
| | - Howard L. Kaufman
- Division of Surgical Oncology, Rutgers Cancer Institute of New Jersey and Rutgers Robert Wood Johnson Medical School
| |
Collapse
|
24
|
Kissick HT, Sanda MG. The role of active vaccination in cancer immunotherapy: lessons from clinical trials. Curr Opin Immunol 2015; 35:15-22. [PMID: 26050634 DOI: 10.1016/j.coi.2015.05.004] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2015] [Revised: 05/06/2015] [Accepted: 05/08/2015] [Indexed: 01/05/2023]
Abstract
In the past few years, a number of different immunotherapeutic strategies have shown impressive results in cancer patients. These successful approaches include blockade of immunosuppressive molecules like PD-1 and CTLA-4, adoptive transfer of patient derived and genetically modified T-cells, and vaccines that stimulate tumor antigen specific T-cells. However, several large vaccine trials recently failed to reach designated primary endpoints. In light of the success of other immunotherapeutic approaches, these negative results raise the questions of why vaccines have not generated a better response, and what the role of active vaccination will be moving forward in cancer immunotherapy.
Collapse
Affiliation(s)
- Haydn T Kissick
- Department of Urology, Emory University School of Medicine, United States; Department of Microbiology and Immunology, Emory University School of Medicine, United States.
| | - Martin G Sanda
- Department of Urology, Emory University School of Medicine, United States
| |
Collapse
|
25
|
Liu JK. Anti-cancer vaccines - a one-hit wonder? THE YALE JOURNAL OF BIOLOGY AND MEDICINE 2014; 87:481-9. [PMID: 25506282 PMCID: PMC4257035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Immunization against common bacterial and viral diseases has helped prevent millions of deaths worldwide. More recently, the concept of vaccination has been developed into a potentially novel strategy to treat and prevent cancer formation, progression, and spread. Over the past few years, a handful of anti-cancer vaccines have been licensed and approved for use in clinical practice, thus providing a breakthrough in the field. However, the path has not always been easy, with many hurdles that have had to be overcome in order to reach this point. Nevertheless, with more anti-cancer vaccines currently in development, there is still hope that they can eventually become routine tools used in the treatment and prevention of cancer in the future. This review will discuss in detail both types of anti-cancer vaccine presently used in clinical practice - therapeutic and preventive - before considering some of the more promising anti-cancer vaccines that are currently in development. Finally, the issue of side effects and the debate surrounding the overall cost-effectiveness of anti-cancer vaccines will be examined.
Collapse
|
26
|
Unger WW, Mayer CT, Engels S, Hesse C, Perdicchio M, Puttur F, Streng-Ouwehand I, Litjens M, Kalay H, Berod L, Sparwasser T, van Kooyk Y. Antigen targeting to dendritic cells combined with transient regulatory T cell inhibition results in long-term tumor regression. Oncoimmunology 2014; 4:e970462. [PMID: 26405564 PMCID: PMC4570108 DOI: 10.4161/21624011.2014.970462] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2014] [Revised: 09/24/2014] [Accepted: 09/25/2014] [Indexed: 12/20/2022] Open
Abstract
Therapeutic vaccinations against cancer are still largely ineffective. Major caveats are inefficient delivery of tumor antigens to dendritic cells (DCs) and excessive immune suppression by Foxp3+ regulatory T cells (Tregs), resulting in defective T cell priming and failure to induce tumor regression. To circumvent these problems we evaluated a novel combinatorial therapeutic strategy. We show that tumor antigen targeting to DC-SIGN in humanized hSIGN mice via glycans or specific antibodies induces superior T cell priming. Next, this targeted therapy was combined with transient Foxp3+ Treg depletion employing hSIGNxDEREG mice. While Treg depletion alone slightly delayed B16-OVA melanoma growth, only the combination therapy instigated long-term tumor regression in a substantial fraction of mice. This novel strategy resulted in optimal generation of antigen-specific activated CD8+ T cells which accumulated in regressing tumors. Notably, Treg depletion also allowed the local appearance of effector T cells specific for endogenous B16 antigens. This indicates that antitumor immune responses can be broadened by therapies aimed at controlling Tregs in tumor environments. Thus, transient inhibition of Treg-mediated immune suppression potentiates DC targeted antigen vaccination and tumor-specific immunity.
Collapse
Affiliation(s)
- Wendy Wj Unger
- Department of Molecular Cell Biology and Immunology; VU University Medical Center ; Amsterdam, The Netherlands
| | - Christian T Mayer
- Institute of Infection Immunology, TWINCORE, Centre for Experimental and Clinical Infection Research, a Joint Venture between the Medical School Hanover and the Helmholtz Centre for Infection Research , Hannover, Germany
| | - Steef Engels
- Department of Molecular Cell Biology and Immunology; VU University Medical Center ; Amsterdam, The Netherlands
| | - Christina Hesse
- Institute of Infection Immunology, TWINCORE, Centre for Experimental and Clinical Infection Research, a Joint Venture between the Medical School Hanover and the Helmholtz Centre for Infection Research , Hannover, Germany
| | - Maurizio Perdicchio
- Department of Molecular Cell Biology and Immunology; VU University Medical Center ; Amsterdam, The Netherlands
| | - Franz Puttur
- Institute of Infection Immunology, TWINCORE, Centre for Experimental and Clinical Infection Research, a Joint Venture between the Medical School Hanover and the Helmholtz Centre for Infection Research , Hannover, Germany
| | - Ingeborg Streng-Ouwehand
- Department of Molecular Cell Biology and Immunology; VU University Medical Center ; Amsterdam, The Netherlands
| | - Manja Litjens
- Department of Molecular Cell Biology and Immunology; VU University Medical Center ; Amsterdam, The Netherlands
| | - Hakan Kalay
- Department of Molecular Cell Biology and Immunology; VU University Medical Center ; Amsterdam, The Netherlands
| | - Luciana Berod
- Institute of Infection Immunology, TWINCORE, Centre for Experimental and Clinical Infection Research, a Joint Venture between the Medical School Hanover and the Helmholtz Centre for Infection Research , Hannover, Germany
| | - Tim Sparwasser
- Institute of Infection Immunology, TWINCORE, Centre for Experimental and Clinical Infection Research, a Joint Venture between the Medical School Hanover and the Helmholtz Centre for Infection Research , Hannover, Germany
| | - Yvette van Kooyk
- Department of Molecular Cell Biology and Immunology; VU University Medical Center ; Amsterdam, The Netherlands
| |
Collapse
|
27
|
Mansfield AS, Nevala WK, Lieser EAT, Leontovich AA, Markovic SN. The immunomodulatory effects of bevacizumab on systemic immunity in patients with metastatic melanoma. Oncoimmunology 2014; 2:e24436. [PMID: 23762809 PMCID: PMC3667915 DOI: 10.4161/onci.24436] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2013] [Revised: 03/15/2013] [Accepted: 03/25/2013] [Indexed: 11/19/2022] Open
Abstract
Metastatic melanoma is near-to-invariably a fatal disease. As novel therapeutic strategies against metastatic melanoma are urgently needed, we have tested a combinatorial regimen consisting of conventional chemotherapy coupled to bevacizumab, a monoclonal antibody that inhibit angiogenesis, demonstrating some clinical benefit. A preliminary assessment of one of our clinical trials points to a previously unrecognized immunomodulatory effect of bevacizumab. Herein, we evaluate the immunomodulatory effect of bevacizumab when administered together with conventional chemotherapy to patients with metastatic melanoma. To this aim, we measured the abundance of various lymphocyte subsets among peripheral blood mononuclear cells (PBMCs) as well as the circulating levels of 42 cytokines, chemokines and growth factors in patients with metastatic melanoma who received albumin-bound paclitaxel plus carboplatin, either as a standalone intervention (AC, 55 subjects) or combined with bevacizumab (ACB, 39 individuals), in the context of clinical trials N057e and N0775, respectively. Relative shifts in PBMC subsets and cytokine levels were calculated (relative to baseline levels) when patients underwent restaging evaluation after two cycles of therapy. The Mann-Whitney U test was used to compare responses between the groups. Bevacizumab failed to affect the TH1/TH2 cell ratio in this patient cohort. However, we observed a significant increase in CD8+ lymphocytes in patients who received ACB (+38%) but not in subjects treated with AC only (-10%) (p = 0.03). Moreover, circulating interleuikin-6 (IL-6) levels were reduced in patients treated with ACB (-42%) but not in individuals receiving AC only (28%) (p = 0.0018). Thus, the addition of bevacizumab to chemotherapy for the treatment of metastatic melanoma exerts immunomodulatory effects.
Collapse
Affiliation(s)
- Aaron S Mansfield
- Department of Oncology; Mayo Clinic; Rochester, MN USA ; Department of Internal Medicine; Division of Hematology; Mayo Clinic; Rochester, MN USA
| | | | | | | | | |
Collapse
|
28
|
Abstract
Augmented numbers of regulatory T cells contribute to the overall immunosuppression in tumor patients. Interleukin-2 has been widely used in the clinics in anticancer therapy, yet evidence has accumulated that the major drawback, limiting clinical efficacy, is the expansion of regulatory T cells, which aggravates immunosuppression.
Collapse
Affiliation(s)
- Marc Beyer
- Genomics and Immunoregulation; LIMES-Institute; University of Bonn; Bonn, Germany
| |
Collapse
|
29
|
Abstract
The potential for therapeutic efficacy of a melanoma vaccine has been evident preclinically for many years. In melanoma patients, vaccines have resulted in the induction of immune responses, although clinical benefit has not been clearly documented. The recent achievements with immune-checkpoint blockade have shown that immunotherapy can be a powerful tool in cancer therapy. With increased understanding of tumor immunity, the limitations of previous cancer vaccination approaches have become evident. Rapid progress in technologies that enable better vaccine design raise the expectation that these limitations can be overcome, thus leading to a clinically effective melanoma vaccine in the near future.
Collapse
Affiliation(s)
- Patrick A Ott
- Department of Medical Oncology, Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, MA 02215, USA; Melanoma Disease Center, Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, MA 02215, USA; Center for Immuno-Oncology, Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, MA 02215, USA; Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, 75 Francis Street, Boston, MA 02215, USA.
| | - Edward F Fritsch
- Department of Medical Oncology, Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, MA 02215, USA; Broad Institute of Harvard and MIT, 7 Cambridge Center, Cambridge, MA 02142, USA
| | - Catherine J Wu
- Department of Medical Oncology, Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, MA 02215, USA; Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, 75 Francis Street, Boston, MA 02215, USA; Cancer Vaccine Center, Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, MA 02215, USA
| | - Glenn Dranoff
- Department of Medical Oncology, Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, MA 02215, USA; Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, 75 Francis Street, Boston, MA 02215, USA; Cancer Vaccine Center, Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, MA 02215, USA
| |
Collapse
|
30
|
Guo C, Manjili MH, Subjeck JR, Sarkar D, Fisher PB, Wang XY. Therapeutic cancer vaccines: past, present, and future. Adv Cancer Res 2014; 119:421-75. [PMID: 23870514 DOI: 10.1016/b978-0-12-407190-2.00007-1] [Citation(s) in RCA: 365] [Impact Index Per Article: 36.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Therapeutic vaccines represent a viable option for active immunotherapy of cancers that aim to treat late stage disease by using a patient's own immune system. The promising results from clinical trials recently led to the approval of the first therapeutic cancer vaccine by the U.S. Food and Drug Administration. This major breakthrough not only provides a new treatment modality for cancer management but also paves the way for rationally designing and optimizing future vaccines with improved anticancer efficacy. Numerous vaccine strategies are currently being evaluated both preclinically and clinically. This review discusses therapeutic cancer vaccines from diverse platforms or targets as well as the preclinical and clinical studies employing these therapeutic vaccines. We also consider tumor-induced immune suppression that hinders the potency of therapeutic vaccines, and potential strategies to counteract these mechanisms for generating more robust and durable antitumor immune responses.
Collapse
Affiliation(s)
- Chunqing Guo
- Department of Human and Molecular Genetics, Virginia Commonwealth University School of Medicine, Richmond, Virginia, USA
| | | | | | | | | | | |
Collapse
|
31
|
Schlom J, Hodge JW, Palena C, Tsang KY, Jochems C, Greiner JW, Farsaci B, Madan RA, Heery CR, Gulley JL. Therapeutic cancer vaccines. Adv Cancer Res 2014; 121:67-124. [PMID: 24889529 PMCID: PMC6324585 DOI: 10.1016/b978-0-12-800249-0.00002-0] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Therapeutic cancer vaccines have the potential of being integrated in the therapy of numerous cancer types and stages. The wide spectrum of vaccine platforms and vaccine targets is reviewed along with the potential for development of vaccines to target cancer cell "stemness," the epithelial-to-mesenchymal transition (EMT) phenotype, and drug-resistant populations. Preclinical and recent clinical studies are now revealing how vaccines can optimally be used with other immune-based therapies such as checkpoint inhibitors, and so-called nonimmune-based therapeutics, radiation, hormonal therapy, and certain small molecule targeted therapies; it is now being revealed that many of these traditional therapies can lyse tumor cells in a manner as to further potentiate the host immune response, alter the phenotype of nonlysed tumor cells to render them more susceptible to T-cell lysis, and/or shift the balance of effector:regulatory cells in a manner to enhance vaccine efficacy. The importance of the tumor microenvironment, the appropriate patient population, and clinical trial endpoints is also discussed in the context of optimizing patient benefit from vaccine-mediated therapy.
Collapse
Affiliation(s)
- Jeffrey Schlom
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA.
| | - James W Hodge
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Claudia Palena
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Kwong-Yok Tsang
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Caroline Jochems
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - John W Greiner
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Benedetto Farsaci
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Ravi A Madan
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Christopher R Heery
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - James L Gulley
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
32
|
Abstract
Extensive research in the area of active-specific immunotherapy has led to the approval of the first therapeutic cancer vaccine sipuleucel-T (Provenge™) in 2010. Even though a major milestone for the field of cancer immunotherapy, many obstacles towards successful integration of vaccination strategies into the oncologists' armamentarium remain. This chapter discusses possible future perspectives for cancer vaccines as a treatment modality in oncology with special focus on biomarkers (response prediction and patient selection), requirements for clinical trial design, and combination therapies (standard of care and new molecular entities).Extensive research in the area of active-specific immunotherapy has led to the approval of the first therapeutic cancer vaccine sipuleucel-T (Provenge™) in 2010. Even though a major milestone for the field of cancer immunotherapy, many obstacles towards successful integration of vaccination strategies into the oncologists' armamentarium remain. This chapter discusses possible future perspectives for cancer vaccines as a treatment modality in oncology with special focus on biomarkers (response prediction and patient selection), requirements for clinical trial design, and combination therapies (standard of care and new molecular entities).
Collapse
Affiliation(s)
- Hauke Winter
- Laboratory of Clinical and Experimental Tumor Immunology, Department of Surgery, Grosshadern Medical Center, Ludwig-Maximilians-University Munich, Munich, Germany
| | | | | |
Collapse
|
33
|
Dillman RO, Depriest C, McClure SE. High-dose IL2 in metastatic melanoma: better survival in patients immunized with antigens from autologous tumor cell lines. Cancer Biother Radiopharm 2013; 29:53-7. [PMID: 24380630 DOI: 10.1089/cbr.2013.1565] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Abstract Various published data show that in patients with metastatic melanoma, high-dose interleukin-2 (IL2) is associated with 5-year survival rates of 15% from treatment initiation. We previously reported a median survival of 15.6 months, and a 20% 5-year survival rate for 150 patients who were treated with inpatient IL2 (Cancer Biother Radiopharm 2012;27:337). In the current study, we sought to determine whether treatment with active specific immunotherapy (ASI) with patient-specific tumor stem cell vaccines derived from autologous tumor cell (TC) lines contributed to the survival result. Existing databases revealed that 32/149 IL2-treated patients also received ASI, while 117 did not. ASI was given within 12 months of IL2 therapy in 19/32 patients. Patients who received IL2 plus ASI had better overall survival (p<0.001) with longer median survival (39.5 vs. 12.0 months) and a higher 5-year survival rate (39% vs. 13%). Survival was better even after exclusion of 55 IL2-alone patients who died before 12 months of follow-up (p=0.12). In subset analyses, survival was longer for 25 patients who received ASI after IL2 than for 7 who received ASI before IL2 (5-year survival 46% vs. 14%, p<0.001) and for 16 patients who received a dendritic cell/TC-based ASI compared with 16 injected with irradiated TC (p=0.17). This retrospective study suggests that receipt of IL2 followed by a patient-specific melanoma stem cell vaccine is associated with better survival than IL2 alone.
Collapse
Affiliation(s)
- Robert O Dillman
- 1 Hoag Institute for Research and Education , Newport Beach, California
| | | | | |
Collapse
|
34
|
Ye L, Fan J, Shi X, Tao Q, Ye D, Xian Z, Zeng X, Li Y, Feng M, Ju D. Tumor necrosis therapy antibody interleukin-2 fusion protein elicits prolonged and targeted antitumor effects in vivo. Appl Microbiol Biotechnol 2013; 98:4053-61. [DOI: 10.1007/s00253-013-5349-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2013] [Revised: 10/17/2013] [Accepted: 10/19/2013] [Indexed: 11/24/2022]
|
35
|
Batus M, Waheed S, Ruby C, Petersen L, Bines SD, Kaufman HL. Optimal management of metastatic melanoma: current strategies and future directions. Am J Clin Dermatol 2013; 14:179-94. [PMID: 23677693 PMCID: PMC3913474 DOI: 10.1007/s40257-013-0025-9] [Citation(s) in RCA: 70] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Melanoma is increasing in incidence and remains a major public health threat. Although the disease may be curable when identified early, advanced melanoma is characterized by widespread metastatic disease and a median survival of less than 10 months. In recent years, however, major advances in our understanding of the molecular nature of melanoma and the interaction of melanoma cells with the immune system have resulted in several new therapeutic strategies that are showing significant clinical benefit. Current therapeutic approaches include surgical resection of metastatic disease, chemotherapy, immunotherapy, and targeted therapy. Dacarbazine, interleukin-2, ipilimumab, and vemurafenib are now approved for the treatment of advanced melanoma. In addition, new combination chemotherapy regimens, monoclonal antibodies blocking the programmed death-1 (PD-1)/PD-ligand 1 pathway, and targeted therapy against CKIT, mitogen-activated protein/extracellular signal-regulated kinase (MEK), and other putative signaling pathways in melanoma are beginning to show promise in early-phase clinical trials. Further research on these modalities alone and in combination will likely be the focus of future clinical investigation and may impact the outcomes for patients with advanced melanoma.
Collapse
Affiliation(s)
- Marta Batus
- Rush University Melanoma Program and Departments of Medicine, General Surgery and Immunology and Microbiology, Rush University Medical Center, 1725 W. Harrison Street, Room 845, Chicago, IL 60612, USA
| | | | | | | | | | | |
Collapse
|
36
|
Alatrash G, Jakher H, Stafford PD, Mittendorf EA. Cancer immunotherapies, their safety and toxicity. Expert Opin Drug Saf 2013; 12:631-45. [DOI: 10.1517/14740338.2013.795944] [Citation(s) in RCA: 92] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
37
|
Filipazzi P, Pilla L, Mariani L, Patuzzo R, Castelli C, Camisaschi C, Maurichi A, Cova A, Rigamonti G, Giardino F, Di Florio A, Asioli M, Frati P, Sovena G, Squarcina P, Maio M, Danielli R, Chiarion-Sileni V, Villa A, Lombardo C, Tragni G, Santinami M, Parmiani G, Rivoltini L. Limited induction of tumor cross-reactive T cells without a measurable clinical benefit in early melanoma patients vaccinated with human leukocyte antigen class I-modified peptides. Clin Cancer Res 2012; 18:6485-96. [PMID: 23032742 DOI: 10.1158/1078-0432.ccr-12-1516] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE The progressive immune dysfunctions that occur in patients with advanced melanoma make them unlikely to efficiently respond to cancer vaccines. A multicenter randomized phase II trial was conducted to test whether immunization with modified HLA class I tumor peptides in the context of adjuvant therapy results in better immunologic responses and improved clinical outcomes in patients with early melanoma (stages IIB/C-III). EXPERIMENTAL DESIGN Forty-three patients were enrolled to undergo vaccination (n = 22) or observation (n = 21). The vaccine included four HLA-A*0201-restricted modified peptides (Melan-A/MART-1([27L]), gp100([210M]), NY-ESO-1([165V]), and Survivin([97M])) emulsified in Montanide ISA51 and injected subcutaneously in combination with cyclophosphamide (300 mg/m(2)) and low-dose IL-2 (3 × 10(6) IU). The immune responses were monitored using ex vivo IFN-γ-ELISpot, HLA/multimer staining, and in vitro short-term peptide sensitization assays. RESULTS Vaccination induced a rapid and persistent increase in specific effector memory CD8(+) T cells in 75% of the patients. However, this immunization was not associated with any significant increase in disease-free or overall survival as compared with the observation group. An extensive immunologic analysis revealed a significantly reduced cross-recognition of the corresponding native peptides and, most importantly, a limited ability to react to melanoma cells. CONCLUSIONS Adjuvant setting is an appealing approach for testing cancer vaccines because specific CD8(+) T cells can be efficiently induced in most vaccinated patients. However, the marginal antitumor activity of the T cells induced by modified peptides in this study largely accounts for the observed lack of benefit of vaccination. These findings suggest reconsidering this immunization strategy, particularly in early disease.
Collapse
Affiliation(s)
- Paola Filipazzi
- Unit of Immunotherapy of Human Tumors, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Berinstein NL, Karkada M, Morse MA, Nemunaitis JJ, Chatta G, Kaufman H, Odunsi K, Nigam R, Sammatur L, MacDonald LD, Weir GM, Stanford MM, Mansour M. First-in-man application of a novel therapeutic cancer vaccine formulation with the capacity to induce multi-functional T cell responses in ovarian, breast and prostate cancer patients. J Transl Med 2012; 10:156. [PMID: 22862954 PMCID: PMC3479010 DOI: 10.1186/1479-5876-10-156] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2012] [Accepted: 07/11/2012] [Indexed: 11/20/2022] Open
Abstract
Background DepoVaxTM is a novel non-emulsion depot-forming vaccine platform with the capacity to significantly enhance the immunogenicity of peptide cancer antigens. Naturally processed HLA-A2 restricted peptides presented by breast, ovarian and prostate cancer cells were used as antigens to create a therapeutic cancer vaccine, DPX-0907. Methods A phase I clinical study was designed to examine the safety and immune activating potential of DPX-0907 in advanced stage breast, ovarian and prostate cancer patients. A total of 23 late stage cancer patients were recruited and were divided into two dose/volume cohorts in a three immunization protocol. Results DPX-0907 was shown to be safe with injection site reactions being the most commonly reported adverse event. All breast cancer patients (3/3), most of ovarian (5/6) and one third of prostate (3/9) cancer patients exhibited detectable immune responses, resulting in a 61% immunological response rate. Immune responses were generally observed in patients with better disease control after their last prior treatment. Antigen-specific responses were detected in 73% of immune responders (44% of evaluable patients) after the first vaccination. In 83% of immune responders (50% of evaluable patients), peptide-specific T cell responses were detected at ≥2 time points post vaccination with 64% of the responders (39% of evaluable patients) showing evidence of immune persistence. Immune monitoring also demonstrated the generation of antigen-specific T cell memory with the ability to secrete multiple Type 1 cytokines. Conclusions The novel DepoVax formulation promotes multifunctional effector memory responses to peptide-based tumor associated antigens. The data supports the capacity of DPX-0907 to elicit Type-1 biased immune responses, warranting further clinical development of the vaccine. This study underscores the importance of applying vaccines in clinical settings in which patients are more likely to be immune competent. Trial Registration ClinicalTrials.gov NCT01095848
Collapse
|
39
|
Abstract
The inherent immunogenicity of melanoma and renal cell carcinoma (RCC) has made these tumors a focus of considerable research in vaccine development. Recent data from murine studies of immunosurveillance have highlighted the importance of both innate and adaptive immune responses in shaping a tumor's inherent susceptibility to immune surveillance and immunotherapy. Melanoma has been a useful model for the identification of tumor-associated antigens and a number of putative renal cell antigens have been described more recently. These antigens have been targeted using a variety of vaccine strategies, including protein- and peptide-based vaccines, recombinant antigen-expressing vectors, and whole cell vaccine approaches. While evidence for clinical benefit has been disappointing to date, several current phase III clinical trials are in progress based on promising results from phase II studies. Accumulating data suggest that the tumor microenvironment and mechanisms of immunological escape by established tumors are significant barriers that must be overcome before vaccine therapy can be fully realized. This review will discuss the basis for vaccine development, describe some of the more promising vaccine strategies in development, and mention some of the tumor escape mechanisms that block effective anti-tumor immunity for melanoma and RCC.
Collapse
|
40
|
Syed ON, Mandigo CE, Killory BD, Canoll P, Bruce JN. Cancer-testis and melanocyte-differentiation antigen expression in malignant glioma and meningioma. J Clin Neurosci 2012; 19:1016-21. [DOI: 10.1016/j.jocn.2011.10.008] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2011] [Accepted: 10/18/2011] [Indexed: 11/27/2022]
|
41
|
Zhou F, Hu J, Shao JH, Zou SB, Shen SL, Luo ZQ. Metronomic chemotherapy in combination with antiangiogenic treatment induces mosaic vascular reduction and tumor growth inhibition in hepatocellular carcinoma xenografts. J Cancer Res Clin Oncol 2012; 138:1879-90. [PMID: 22736027 DOI: 10.1007/s00432-012-1270-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2012] [Accepted: 06/13/2012] [Indexed: 12/11/2022]
Abstract
BACKGROUND In addition to sprouting angiogenesis, other mechanisms, such as mosaic tumor vessel formation, have been recognized to contribute to tumor vascularization. We sought to examine vascular alteration as well as tumor growth inhibition after treatment with antiangiogenic therapy, chemotherapy alone or in combination. METHODS Hepatocellular carcinoma cells (Hep3B) expressed green fluorescent protein were utilized to establish orthotopic xenograft model in nude mice. The formation and distribution of mosaic vessels was analyzed quantitatively by immunolabeling. Next, changes in tumor microcirculation and therapeutic effects on tumor growth were evaluated in several different treatment groups: control, conventional doxorubicin, metronomic doxorubicin, bevacizumab, bevacizumab plus conventional doxorubicin, and bevacizumab plus metronomic doxorubicin. In addition, we examined the effects of combined regimens on lung metastasis using a highly metastatic human hepatocellular carcinoma (HCCLM3) mouse model. RESULTS Approximately 62 % of the vessels were present in the central part or near the midsection of the tumor and were mosaic. Only the combined antiangiogenic treatment and chemotherapy (metronomic schedule, P = 0.00; conventional schedule, P = 0.02) had a significant effect on the degree of mosaic vasculature. Metronomic doxorubicin in combination with bevacizumab had an even more profound effect than bevacizumab plus conventional doxorubicin (P < 0.05) on tumor growth inhibition and survival. However, bevacizumab plus metronomic doxorubicin failed to inhibit lung metastasis compared with antiangiogenic monotherapy. CONCLUSIONS Metronomic chemotherapy in combination with antiangiogenic treatment results in the reduction of mosaic tumor vasculature, inhibition of tumor growth, and enhanced survival of mice. Further investigation of drug scheduling is required to optimize antitumor activity.
Collapse
MESH Headings
- Administration, Metronomic
- Angiogenesis Inhibitors/administration & dosage
- Animals
- Antibodies, Monoclonal, Humanized/administration & dosage
- Antineoplastic Combined Chemotherapy Protocols/therapeutic use
- Bevacizumab
- Carcinoma, Hepatocellular/drug therapy
- Carcinoma, Hepatocellular/pathology
- Cell Line, Tumor
- Doxorubicin/administration & dosage
- Drug Administration Schedule
- Humans
- Injections, Intravenous
- Liver Neoplasms/drug therapy
- Liver Neoplasms/pathology
- Lung Neoplasms/prevention & control
- Lung Neoplasms/secondary
- Male
- Mice
- Mice, Inbred BALB C
- Mice, Nude
- Neovascularization, Pathologic/pathology
- Neovascularization, Pathologic/prevention & control
- Survival Analysis
- Treatment Outcome
- Tumor Burden/drug effects
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Fan Zhou
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Nanchang University, 1 Mingde Road, Nanchang 330006, China.
| | | | | | | | | | | |
Collapse
|
42
|
Abstract
Despite significant scientific knowledge in the field of cancer immunology, therapeutic strategies using cancer vaccines to generate anti-tumor immunity have historically resulted in only modest clinical benefit. Disappointing results from prior cancer vaccine trials are likely due to multifactorial causes. Perhaps the most important is the role of inherent tumor-induced immune suppression and enhanced immunologic tolerance. Current research directed toward understanding the mechanisms of immunologic tolerance has led to the development of promising therapeutic immune regulatory antibodies that inhibit immunologic checkpoints and subsequently enhance immunologic anti-tumor activity. This review discusses the prior challenges associated with cancer vaccines and describes how, by breaking immune inhibition and facilitating immune stimulation, immune regulatory antibodies show great promise in the treatment of a variety of tumors.
Collapse
|
43
|
Schlom J. Therapeutic cancer vaccines: current status and moving forward. J Natl Cancer Inst 2012; 104:599-613. [PMID: 22395641 DOI: 10.1093/jnci/djs033] [Citation(s) in RCA: 198] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Concurrent with U.S. Food and Drug Administration (FDA) approval of the first therapeutic cancer vaccine, a wide spectrum of other cancer vaccine platforms that target a diverse range of tumor-associated antigens is currently being evaluated in randomized phase II and phase III trials. The profound influence of the tumor microenvironment and other immunosuppressive entities, however, can limit the effectiveness of these vaccines. Numerous strategies are currently being evaluated both preclinically and clinically to counteract these immunosuppressive entities, including the combined use of vaccines with immune checkpoint inhibitors, certain chemotherapeutics, small-molecule targeted therapies, and radiation. The potential influence of the appropriate patient population and clinical trial endpoint in vaccine therapy studies is discussed, as well as the potential importance of biomarkers in future directions of this field.
Collapse
Affiliation(s)
- Jeffrey Schlom
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 10 Center Dr, Rm 8B09, Bethesda, MD 20892, USA.
| |
Collapse
|
44
|
Ménétrier-Caux C, Curiel T, Faget J, Manuel M, Caux C, Zou W. Targeting regulatory T cells. Target Oncol 2012; 7:15-28. [PMID: 22327882 DOI: 10.1007/s11523-012-0208-y] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2011] [Accepted: 01/13/2012] [Indexed: 01/08/2023]
Abstract
Cancers express tumor-associated antigens that should elicit immune response to antagonize the tumor growth, but spontaneous immune rejection of established cancer is rare, suggesting an immunosuppressive environment hindering host antitumor immunity. Among the specific and active tumor-mediated mechanisms, CD4(+)CD25(high) T regulatory cells (Treg) are important mediators of active immune evasion in cancer. In this review, we will discuss Treg subpopulations and the mechanisms of their suppressive functions. Treg depletion improves endogenous antitumor immunity and the efficacy of active immunotherapy in animal models for cancer, suggesting that inhibiting Treg function could also improve the limited successes of human cancer immunotherapy. We will also discuss specific strategies for devising effective cancer immunotherapy targeting Treg.
Collapse
|
45
|
Beyer M, Schumak B, Weihrauch MR, Andres B, Giese T, Endl E, Knolle PA, Classen S, Limmer A, Schultze JL. In vivo expansion of naïve CD4+ CD25(high) FOXP3+ regulatory T cells in patients with colorectal carcinoma after IL-2 administration. PLoS One 2012; 7:e30422. [PMID: 22276195 PMCID: PMC3262821 DOI: 10.1371/journal.pone.0030422] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2011] [Accepted: 12/15/2011] [Indexed: 11/18/2022] Open
Abstract
Regulatory T cells (T(reg) cells) are increased in context of malignancies and their expansion can be correlated with higher disease burden and decreased survival. Initially, interleukin 2 (IL-2) has been used as T-cell growth factor in clinical vaccination trials. In murine models, however, a role of IL-2 in development, differentiation, homeostasis, and function of T(reg) cells was established. In IL-2 treated cancer patients a further T(reg)-cell expansion was described, yet, the mechanism of expansion is still elusive. Here we report that functional T(reg) cells of a naïve phenotype--as determined by CCR7 and CD45RA expression--are significantly expanded in colorectal cancer patients. Treatment of 15 UICC stage IV colorectal cancer patients with IL-2 in a phase I/II peptide vaccination trial further enlarges the already increased naïve T(reg)-cell pool. Higher frequencies of T-cell receptor excision circles in naïve T(reg) cells indicate IL-2 dependent thymic generation of naïve T(reg) cells as a mechanism leading to increased frequencies of T(reg) cells post IL-2 treatment in cancer patients. This finding could be confirmed in naïve murine T(reg) cells after IL-2 administration. These results point to a more complex regulation of T(reg) cells in context of IL-2 administration. Future strategies therefore might aim at combining IL-2 therapy with novel strategies to circumvent expansion and differentiation of naïve T(reg) cells.
Collapse
Affiliation(s)
- Marc Beyer
- Life and Medical Sciences Institute, Genomics and Immunoregulation, University of Bonn, Bonn, Germany
- * E-mail: (MB); (JLS)
| | - Beatrix Schumak
- Institute of Medical Microbiology, Immunology and Parasitology (IMMIP), University of Bonn, Bonn, Germany
| | | | - Bettina Andres
- Clinic I for Internal Medicine, University of Cologne, Cologne, Germany
| | - Thomas Giese
- Institute of Immunology, University of Heidelberg, Heidelberg, Germany
| | - Elmar Endl
- Institute for Molecular Medicine and Experimental Immunology, University of Bonn, Bonn, Germany
| | - Percy A. Knolle
- Institute for Molecular Medicine and Experimental Immunology, University of Bonn, Bonn, Germany
| | - Sabine Classen
- Life and Medical Sciences Institute, Genomics and Immunoregulation, University of Bonn, Bonn, Germany
| | - Andreas Limmer
- Institute for Molecular Medicine and Experimental Immunology, University of Bonn, Bonn, Germany
| | - Joachim L. Schultze
- Life and Medical Sciences Institute, Genomics and Immunoregulation, University of Bonn, Bonn, Germany
- * E-mail: (MB); (JLS)
| |
Collapse
|
46
|
Lee S, Margolin K. Cytokines in cancer immunotherapy. Cancers (Basel) 2011; 3:3856-93. [PMID: 24213115 PMCID: PMC3763400 DOI: 10.3390/cancers3043856] [Citation(s) in RCA: 458] [Impact Index Per Article: 35.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2011] [Revised: 09/24/2011] [Accepted: 09/27/2011] [Indexed: 02/06/2023] Open
Abstract
Cytokines are molecular messengers that allow the cells of the immune system to communicate with one another to generate a coordinated, robust, but self-limited response to a target antigen. The growing interest over the past two decades in harnessing the immune system to eradicate cancer has been accompanied by heightened efforts to characterize cytokines and exploit their vast signaling networks to develop cancer treatments. The goal of this paper is to review the major cytokines involved in cancer immunotherapy and discuss their basic biology and clinical applications. The paper will also describe new cytokines in pre-clinical development, combinations of biological agents, novel delivery mechanisms, and potential directions for future investigation using cytokines.
Collapse
Affiliation(s)
- Sylvia Lee
- Division of Medical Oncology, Department of Medicine, University of Washington, Seattle, WA 98195, USA; E-Mail:
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Kim Margolin
- Division of Medical Oncology, Department of Medicine, University of Washington, Seattle, WA 98195, USA; E-Mail:
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| |
Collapse
|
47
|
Klein O, Schmidt C, Knights A, Davis ID, Chen W, Cebon J. Melanoma vaccines: developments over the past 10 years. Expert Rev Vaccines 2011; 10:853-73. [PMID: 21692705 DOI: 10.1586/erv.11.74] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Decades of preclinical evaluation and clinical trials into melanoma vaccines have yielded spectacular progress in our understanding of melanoma antigens and the immune mechanisms of tumor rejection. Key insights and the results of their clinical evaluation are reviewed in this article. Unfortunately, durable clinical benefit following vaccination remains uncommon. Two recent clinical advances that will impact on melanoma vaccine development are trials with inhibitors of CTLA-4 and oncogenic BRAF. Long-term therapeutic control of melanoma will require integration of specific active immunotherapy with these emerging successful therapies from the disparate fields of immune regulation and signal transduction.
Collapse
Affiliation(s)
- Oliver Klein
- Ludwig Institute for Cancer Research, Austin Branch, Austin Hospital, Studley Road, Heidelberg, Victoria, 3084, Australia
| | | | | | | | | | | |
Collapse
|
48
|
Abstract
Few immunotherapists would accept the concept of a single vaccination inducing a therapeutic anticancer immune response in a patient with advanced cancer. But what is the evidence to support the "more-is-better" approach of multiple vaccinations? Because we are unaware of trials comparing the effect of a single vaccine versus multiple vaccinations on patient outcome, we considered that an anticancer immune response might provide a surrogate measure of the effectiveness of vaccination strategies. Because few large trials include immunologic monitoring, the majority of information is gleaned from smaller trials in which an evaluation of immune responses to vaccine or tumor, before and at 1 or more times following the first vaccine, was performed. In some studies, there is convincing evidence that repeated administration of a specific vaccine can augment the immune response to antigens contained in the vaccine. In other settings, multiple vaccinations can significantly reduce the immune response to 1 or more targets. Results from 3 large adjuvant vaccine studies support the potential detrimental effect of multiple vaccinations as clinical outcomes in the control arms were significantly better than that for treatment groups. Recent research has provided insights into mechanisms that are likely responsible for the reduced responses in the studies noted above, but supporting evidence from clinical specimens is generally lacking. Interpretation of these results is further complicated by the possibility that the dominant immune response may evolve to recognize epitopes not present in the vaccine. Nonetheless, the Food and Drug Administration approval of the first therapeutic cancer vaccine and recent developments from preclinical models and clinical trials provide a substantial basis for optimism and a critical evaluation of cancer vaccine strategies.
Collapse
Affiliation(s)
- Sarah E Church
- Robert W. Franz Cancer Research Center, Earle A. Chiles Research Institute, Providence Cancer Center, Portland Medical Center, USA
| | | | | | | | | | | | | |
Collapse
|
49
|
|
50
|
Abstract
Cancer immunotherapy consists of approaches that modify the host immune system, and/or the utilization of components of the immune system, as cancer treatment. During the past 25 years, 17 immunologic products have received regulatory approval based on anticancer activity as single agents and/or in combination with chemotherapy. These include the nonspecific immune stimulants BCG and levamisole; the cytokines interferon-α and interleukin-2; the monoclonal antibodies rituximab, ofatumumab, alemtuzumab, trastuzumab, bevacizumab, cetuximab, and panitumumab; the radiolabeled antibodies Y-90 ibritumomab tiuxetan and I-131 tositumomab; the immunotoxins denileukin diftitox and gemtuzumab ozogamicin; nonmyeloablative allogeneic transplants with donor lymphocyte infusions; and the anti-prostate cancer cell-based therapy sipuleucel-T. All but two of these products are still regularly used to treat various B- and T-cell malignancies, and numerous solid tumors, including breast, lung, colorectal, prostate, melanoma, kidney, glioblastoma, bladder, and head and neck. Positive randomized trials have recently been reported for idiotype vaccines in lymphoma and a peptide vaccine in melanoma. The anti-CTLA-4 monoclonal antibody ipilumumab, which blocks regulatory T-cells, is expected to receive regulatory approval in the near future, based on a randomized trial in melanoma. As the fourth modality of cancer treatment, biotherapy/immunotherapy is an increasingly important component of the anticancer armamentarium.
Collapse
Affiliation(s)
- Robert O Dillman
- Hoag Cancer Institute of Hoag Hospital , Newport Beach, California 92658, USA.
| |
Collapse
|