1
|
Gianneschi G, Scolpino A, Oleske J. Risk of autoimmunity, cancer seeding, and adverse events in human trials of whole-tissue autologous therapeutic vaccines. CANCER PATHOGENESIS AND THERAPY 2025; 3:129-134. [PMID: 40182122 PMCID: PMC11963168 DOI: 10.1016/j.cpt.2024.05.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/24/2024] [Revised: 05/20/2024] [Accepted: 05/26/2024] [Indexed: 04/05/2025]
Abstract
Background Whole-tissue autologous therapeutic vaccines (WATVs) are a form of cancer immunotherapy that use a patient's own pathological tissue. Concerns exist regarding the potential of WATVs to induce autoimmunity or the spread of cancer; however, their adverse events (AEs) have not been adequately studied. This literature review primarily aimed to evaluate the risks of autoimmunity and cancer seeding associated with using WATVs in human clinical trials. Its secondary objectives included assessing the incidence of AEs graded 1-5 using the Common Terminology Criteria for Adverse Events v5.0. Methods The inclusion criteria were any clinical trial using human subjects in which at least part of the cancer vaccine was derived from the patient's own tumor tissue, which likely preserved the unique tumor-associated antigens (TAAs) present in the patient's tumor (i.e., whole-tissue). Tumor vaccine trials that used limited TAAs or highly processed tumor antigens were excluded. Published clinical trials were searched using Google Scholar until March 2024. The authors elaborated on the risk of bias in such cases, as indicated. All reviewed publications were searched for evidence of autoimmunity, cancer seeding, and other AEs. The Preferred Reporting Items for Systematic Reviews and Meta-Analyses 2020 statement guided the review. Results Data from 55 human clinical trials, abstracts, case reports, and unpublished data were analyzed, including 3323 patients treated with WATVs for various cancers. The primary outcomes were: (1) no documented cases of WATV-induced autoimmunity, (2) no documented cases of WATV-induced spreading or seeding of noninfectious cancers, and (3) the observed 0.24% (2/838) risk of spreading or seeding infectious cancers was attributed to inadequate sterilization. The secondary outcomes were: (1) no deaths were attributed to WATV therapy, (2) 0.18% (6/3323) incidence of grade 4 AEs, (3) 0.42% (14/3323) incidence of grade 3 AEs, (4) the incidence of grades 1-2 AEs was 52.21% (478/916). Conclusions WATVs carry no risk of inducing autoimmunity and essentially no risk of cancer seeding if properly sterilized. WATVs also exhibit a side effect profile comparable to that of routine vaccinations, with common, mild, and transient adverse effects. The combined risk of grade 3 and 4 AEs was 0.60% (20/3323). No deaths were causally associated with WATV treatment.
Collapse
Affiliation(s)
- Garrett Gianneschi
- Department of Neurology, Rutgers University - New Jersey Medical School, Newark, NJ 07103, USA
| | - Anthony Scolpino
- Division of Immunology, Department of Pediatrics, Rutgers University - New Jersey Medical School, Newark, NJ 07103, USA
| | - James Oleske
- Division of Immunology, Department of Pediatrics, Rutgers University - New Jersey Medical School, Newark, NJ 07103, USA
| |
Collapse
|
2
|
Delgado-Almenta V, Blaya-Cánovas JL, Calahorra J, López-Tejada A, Griñán-Lisón C, Granados-Principal S. Cancer Vaccines and Beyond: The Transformative Role of Nanotechnology in Immunotherapy. Pharmaceutics 2025; 17:216. [PMID: 40006583 PMCID: PMC11859086 DOI: 10.3390/pharmaceutics17020216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Revised: 01/20/2025] [Accepted: 02/05/2025] [Indexed: 02/27/2025] Open
Abstract
Cancer is one of the leading causes of morbidity and mortality globally, responsible for approximately 10 million deaths in 2022 and an estimated 21 million new cases in 2024. Traditional cancer treatments such as surgery, radiation therapy, and chemotherapy often present limitations in efficacy and side effects. However, immunotherapeutic vaccines have emerged as a promising approach, leveraging the body's immune system to target and eliminate cancer cells. This review examines the evolving landscape of cancer vaccines, differentiating between preventive and therapeutic strategies and highlighting the significance of tumor-specific antigens, including tumor-associated antigens (TAAs) and neoantigens. Recent advancements in vaccine technology, particularly through nanotechnology, have resulted in the development of nanovaccines, which enhance antigen stability, optimize delivery to immune cells, and promote robust immune responses. Notably, clinical data indicate that patients receiving immune checkpoint inhibitors can achieve overall survival rates of approximately 34.8 months compared to just 15.7 months for traditional therapies. Despite these advancements, challenges remain, such as the immunosuppressive tumor microenvironment and tumor heterogeneity. Emerging evidence suggests that combining nanovaccines with immunomodulators may enhance therapeutic efficacy by overcoming these obstacles. Continued research and interdisciplinary collaboration will be essential to fully exploit the promise of nanovaccines, ultimately leading to more effective and accessible treatments for cancer patients. The future of cancer immunotherapy appears increasingly hopeful as these innovative strategies pave the way for enhanced patient outcomes and an improved quality of life in oncology.
Collapse
Affiliation(s)
- Violeta Delgado-Almenta
- GENYO, Centre for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government, 18016 Granada, Spain; (V.D.-A.); (J.L.B.-C.); (J.C.); (A.L.-T.)
| | - Jose L. Blaya-Cánovas
- GENYO, Centre for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government, 18016 Granada, Spain; (V.D.-A.); (J.L.B.-C.); (J.C.); (A.L.-T.)
- Instituto de Investigación Biosanitaria ibs.GRANADA, 18012 Granada, Spain
| | - Jesús Calahorra
- GENYO, Centre for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government, 18016 Granada, Spain; (V.D.-A.); (J.L.B.-C.); (J.C.); (A.L.-T.)
- Instituto de Investigación Biosanitaria ibs.GRANADA, 18012 Granada, Spain
| | - Araceli López-Tejada
- GENYO, Centre for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government, 18016 Granada, Spain; (V.D.-A.); (J.L.B.-C.); (J.C.); (A.L.-T.)
- Instituto de Investigación Biosanitaria ibs.GRANADA, 18012 Granada, Spain
- Department of Biochemistry and Molecular Biology II, Faculty of Pharmacy, University of Granada, Campus de Cartuja s/n, 18011 Granada, Spain
| | - Carmen Griñán-Lisón
- GENYO, Centre for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government, 18016 Granada, Spain; (V.D.-A.); (J.L.B.-C.); (J.C.); (A.L.-T.)
- Instituto de Investigación Biosanitaria ibs.GRANADA, 18012 Granada, Spain
- Department of Biochemistry and Molecular Biology II, Faculty of Pharmacy, University of Granada, Campus de Cartuja s/n, 18011 Granada, Spain
- Excellence Research Unit “Modeling Nature” (MNat), Centro de Investigación Biomédica (CIBM), University of Granada, 18016 Granada, Spain
| | - Sergio Granados-Principal
- GENYO, Centre for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government, 18016 Granada, Spain; (V.D.-A.); (J.L.B.-C.); (J.C.); (A.L.-T.)
- Instituto de Investigación Biosanitaria ibs.GRANADA, 18012 Granada, Spain
- Department of Biochemistry and Molecular Biology II, Faculty of Pharmacy, University of Granada, Campus de Cartuja s/n, 18011 Granada, Spain
| |
Collapse
|
3
|
Chen S, Cheng S, Cai J, Liu Z, Li H, Wang P, Li Y, Yang F, Chen K, Qiu M. The current therapeutic cancer vaccines landscape in non-small cell lung cancer. Int J Cancer 2024; 155:1909-1927. [PMID: 39109825 DOI: 10.1002/ijc.35088] [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: 11/30/2023] [Revised: 05/12/2024] [Accepted: 05/29/2024] [Indexed: 10/04/2024]
Abstract
Currently, conventional immunotherapies for the treatment of non-small cell lung cancer (NSCLC) have low response rates and benefit only a minority of patients, particularly those with advanced disease, so novel therapeutic strategies are urgent deeded. Therapeutic cancer vaccines, a form of active immunotherapy, harness potential to activate the adaptive immune system against tumor cells via antigen cross-presentation. Cancer vaccines can establish enduring immune memory and guard against recurrences. Vaccine-induced tumor cell death prompts antigen epitope spreading, activating functional T cells and thereby sustaining a cancer-immunity cycle. The success of vaccines against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has rendered cancer vaccines a promising avenue, especially when combined with immunotherapy or chemoradiotherapy for NSCLC. This review delves into the intricate antitumor immune mechanisms underlying therapeutic cancer vaccines, enumerates the tumor antigen spectrum of NSCLC, discusses different cancer vaccines progress and summarizes relevant clinical trials. Additionally, we analyze the combination strategies, current limitations, and future prospects of cancer vaccines in NSCLC treatment, aiming to offer fresh insights for their clinical application in managing NSCLC. Overall, cancer vaccines offer promising potential for NSCLC treatment, particularly combining with chemoradiotherapy or immunotherapy could further improve survival in advanced patients. Exploring inhaled vaccines or prophylactic vaccines represents a crucial research avenue.
Collapse
Affiliation(s)
- Shaoyi Chen
- Department of Thoracic Surgery, Peking University People's Hospital, Beijing, China
- Thoracic Oncology Institute, Peking University People's Hospital Thoracic Oncology Institute & Research Unit of Intelligence Diagnosis and Treatment in Early Non-small Cell Lung Cancer, Beijing, China
- Institute of Advanced Clinical Medicine, Peking University, Beijing, China
| | - Sida Cheng
- Department of Thoracic Surgery, Peking University People's Hospital, Beijing, China
- Thoracic Oncology Institute, Peking University People's Hospital Thoracic Oncology Institute & Research Unit of Intelligence Diagnosis and Treatment in Early Non-small Cell Lung Cancer, Beijing, China
- Institute of Advanced Clinical Medicine, Peking University, Beijing, China
| | - Jingsheng Cai
- Department of Thoracic Surgery, Peking University People's Hospital, Beijing, China
- Thoracic Oncology Institute, Peking University People's Hospital Thoracic Oncology Institute & Research Unit of Intelligence Diagnosis and Treatment in Early Non-small Cell Lung Cancer, Beijing, China
- Institute of Advanced Clinical Medicine, Peking University, Beijing, China
| | - Zheng Liu
- Department of Thoracic Surgery, Peking University People's Hospital, Beijing, China
- Thoracic Oncology Institute, Peking University People's Hospital Thoracic Oncology Institute & Research Unit of Intelligence Diagnosis and Treatment in Early Non-small Cell Lung Cancer, Beijing, China
- Institute of Advanced Clinical Medicine, Peking University, Beijing, China
| | - Haoran Li
- Department of Thoracic Surgery, Peking University People's Hospital, Beijing, China
- Thoracic Oncology Institute, Peking University People's Hospital Thoracic Oncology Institute & Research Unit of Intelligence Diagnosis and Treatment in Early Non-small Cell Lung Cancer, Beijing, China
- Institute of Advanced Clinical Medicine, Peking University, Beijing, China
| | - Peiyu Wang
- Department of Thoracic Surgery, Peking University People's Hospital, Beijing, China
- Thoracic Oncology Institute, Peking University People's Hospital Thoracic Oncology Institute & Research Unit of Intelligence Diagnosis and Treatment in Early Non-small Cell Lung Cancer, Beijing, China
- Institute of Advanced Clinical Medicine, Peking University, Beijing, China
| | - Yun Li
- Department of Thoracic Surgery, Peking University People's Hospital, Beijing, China
- Thoracic Oncology Institute, Peking University People's Hospital Thoracic Oncology Institute & Research Unit of Intelligence Diagnosis and Treatment in Early Non-small Cell Lung Cancer, Beijing, China
- Institute of Advanced Clinical Medicine, Peking University, Beijing, China
| | - Fan Yang
- Department of Thoracic Surgery, Peking University People's Hospital, Beijing, China
- Thoracic Oncology Institute, Peking University People's Hospital Thoracic Oncology Institute & Research Unit of Intelligence Diagnosis and Treatment in Early Non-small Cell Lung Cancer, Beijing, China
- Institute of Advanced Clinical Medicine, Peking University, Beijing, China
| | - Kezhong Chen
- Department of Thoracic Surgery, Peking University People's Hospital, Beijing, China
- Thoracic Oncology Institute, Peking University People's Hospital Thoracic Oncology Institute & Research Unit of Intelligence Diagnosis and Treatment in Early Non-small Cell Lung Cancer, Beijing, China
- Institute of Advanced Clinical Medicine, Peking University, Beijing, China
| | - Mantang Qiu
- Department of Thoracic Surgery, Peking University People's Hospital, Beijing, China
- Thoracic Oncology Institute, Peking University People's Hospital Thoracic Oncology Institute & Research Unit of Intelligence Diagnosis and Treatment in Early Non-small Cell Lung Cancer, Beijing, China
- Institute of Advanced Clinical Medicine, Peking University, Beijing, China
| |
Collapse
|
4
|
Mundhara N, Sadhukhan P. Cracking the Codes behind Cancer Cells' Immune Evasion. Int J Mol Sci 2024; 25:8899. [PMID: 39201585 PMCID: PMC11354234 DOI: 10.3390/ijms25168899] [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: 07/12/2024] [Revised: 08/03/2024] [Accepted: 08/09/2024] [Indexed: 09/02/2024] Open
Abstract
Immune evasion is a key phenomenon in understanding tumor recurrence, metastasis, and other critical steps in tumor progression. The tumor microenvironment (TME) is in constant flux due to the tumor's ability to release signals that affect it, while immune cells within it can impact cancer cell behavior. Cancer cells undergo several changes, which can change the enrichment of different immune cells and modulate the activity of existing immune cells in the tumor microenvironment. Cancer cells can evade immune surveillance by downregulating antigen presentation or expressing immune checkpoint molecules. High levels of tumor-infiltrating lymphocytes (TILs) correlate with better outcomes, and robust immune responses can control tumor growth. On the contrary, increased enrichment of Tregs, myeloid-derived suppressor cells, and M2-like anti-inflammatory macrophages can hinder effective immune surveillance and predict poor prognosis. Overall, understanding these immune evasion mechanisms guides therapeutic strategies. Researchers aim to modulate the TME to enhance immune surveillance and improve patient outcomes. In this review article, we strive to summarize the composition of the tumor immune microenvironment, factors affecting the tumor immune microenvironment (TIME), and different therapeutic modalities targeting the immune cells. This review is a first-hand reference to understand the basics of immune surveillance and immune evasion.
Collapse
Affiliation(s)
| | - Pritam Sadhukhan
- Department of Oncology, Johns Hopkins University, Baltimore, MD 21287, USA
| |
Collapse
|
5
|
Sanaei MJ, Pourbagheri-Sigaroodi A, Rezvani A, Zaboli E, Salari S, Masjedi MR, Bashash D. Lung cancer vaccination from concept to reality: A critical review of clinical trials and latest advances. Life Sci 2024; 346:122652. [PMID: 38641048 DOI: 10.1016/j.lfs.2024.122652] [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: 12/24/2023] [Revised: 04/14/2024] [Accepted: 04/16/2024] [Indexed: 04/21/2024]
Abstract
Lung cancer is a highly lethal malignancy that poses a significant burden on public health worldwide. There have been numerous therapeutic approaches, among which cancer vaccines have emerged as a promising approach to harnessing the patient's immune system to induce long-lasting anti-tumor immunity. The current study aims to provide an overview of cancer vaccination in the context of lung cancer to establish a clearer landscape for lung cancer treatment. To provide a comprehensive review, we not only gathered the published studies of lung cancer vaccination and discussed their effectiveness and safety profile but also analyzed all the relevant clinical trials registered on www.clinicaltrials.gov until March 2024. We demonstrated all utilized vaccine platforms along with having a glance at novel technologies such as mRNA vaccines. The present review discussed the challenges and shortcomings of lung cancer vaccination, as well as the way they could be managed to pave the way for reaching the most optimized vaccine formulation.
Collapse
Affiliation(s)
- Mohammad-Javad Sanaei
- Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Atieh Pourbagheri-Sigaroodi
- Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Alireza Rezvani
- Department of Internal Medicine, Hematology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Ehsan Zaboli
- Gastrointestinal Cancer Research Center, Mazandaran University of Medical Sciences, Sari, Iran
| | - Sina Salari
- Department of Medical Oncology-Hematology, Taleghani Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Reza Masjedi
- Cancer Control Research Center, Cancer Control Foundation, Iran University of Medical Sciences, Tehran, Iran; Department of Pulmonary Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Davood Bashash
- Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
6
|
Ingels J, De Cock L, Stevens D, Mayer RL, Théry F, Sanchez GS, Vermijlen D, Weening K, De Smet S, Lootens N, Brusseel M, Verstraete T, Buyle J, Van Houtte E, Devreker P, Heyns K, De Munter S, Van Lint S, Goetgeluk G, Bonte S, Billiet L, Pille M, Jansen H, Pascal E, Deseins L, Vantomme L, Verdonckt M, Roelandt R, Eekhout T, Vandamme N, Leclercq G, Taghon T, Kerre T, Vanommeslaeghe F, Dhondt A, Ferdinande L, Van Dorpe J, Desender L, De Ryck F, Vermassen F, Surmont V, Impens F, Menten B, Vermaelen K, Vandekerckhove B. Neoantigen-targeted dendritic cell vaccination in lung cancer patients induces long-lived T cells exhibiting the full differentiation spectrum. Cell Rep Med 2024; 5:101516. [PMID: 38626769 PMCID: PMC11148567 DOI: 10.1016/j.xcrm.2024.101516] [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: 09/19/2023] [Revised: 02/09/2024] [Accepted: 03/25/2024] [Indexed: 05/24/2024]
Abstract
Non-small cell lung cancer (NSCLC) is known for high relapse rates despite resection in early stages. Here, we present the results of a phase I clinical trial in which a dendritic cell (DC) vaccine targeting patient-individual neoantigens is evaluated in patients with resected NSCLC. Vaccine manufacturing is feasible in six of 10 enrolled patients. Toxicity is limited to grade 1-2 adverse events. Systemic T cell responses are observed in five out of six vaccinated patients, with T cell responses remaining detectable up to 19 months post vaccination. Single-cell analysis indicates that the responsive T cell population is polyclonal and exhibits the near-entire spectrum of T cell differentiation states, including a naive-like state, but excluding exhausted cell states. Three of six vaccinated patients experience disease recurrence during the follow-up period of 2 years. Collectively, these data support the feasibility, safety, and immunogenicity of this treatment in resected NSCLC.
Collapse
Affiliation(s)
- Joline Ingels
- Department of Diagnostic Sciences, Ghent University, 9000 Ghent, East-Flanders, Belgium; Cancer Research Institute Ghent (CRIG), 9000 Ghent, Easy-Flanders, Belgium
| | - Laurenz De Cock
- Cancer Research Institute Ghent (CRIG), 9000 Ghent, Easy-Flanders, Belgium; Department of Biomolecular Medicine, Ghent University, 9000 Ghent, East-Flanders, Belgium
| | - Dieter Stevens
- Cancer Research Institute Ghent (CRIG), 9000 Ghent, Easy-Flanders, Belgium; Respiratory Medicine, Ghent University Hospital, 9000 Ghent, East-Flanders, Belgium
| | - Rupert L Mayer
- Cancer Research Institute Ghent (CRIG), 9000 Ghent, Easy-Flanders, Belgium; Department of Biomolecular Medicine, Ghent University, 9000 Ghent, East-Flanders, Belgium; VIB-UGent Center for Medical Biotechnology, VIB, 9000 Ghent, East-Flanders, Belgium
| | - Fabien Théry
- Department of Biomolecular Medicine, Ghent University, 9000 Ghent, East-Flanders, Belgium; VIB-UGent Center for Medical Biotechnology, VIB, 9000 Ghent, East-Flanders, Belgium
| | - Guillem Sanchez Sanchez
- Department of Pharmacotherapy and Pharmaceutics, Université Libre de Bruxelles, 1050 Brussels, Brussels, Belgium; Institute for Medical Immunology, Université Libre de Bruxelles, 1050 Brussels, Brussels, Belgium; Université Libre de Bruxelles Center for Research in Immunology, Université Libre de Bruxelles, 1050 Brussels, Brussels, Belgium; WELBIO Department, WEL Research Institute, 1300 Wavre, Walloon Brabant, Belgium
| | - David Vermijlen
- Department of Pharmacotherapy and Pharmaceutics, Université Libre de Bruxelles, 1050 Brussels, Brussels, Belgium; Institute for Medical Immunology, Université Libre de Bruxelles, 1050 Brussels, Brussels, Belgium; Université Libre de Bruxelles Center for Research in Immunology, Université Libre de Bruxelles, 1050 Brussels, Brussels, Belgium; WELBIO Department, WEL Research Institute, 1300 Wavre, Walloon Brabant, Belgium
| | - Karin Weening
- Department of Diagnostic Sciences, Ghent University, 9000 Ghent, East-Flanders, Belgium
| | - Saskia De Smet
- GMP Unit Cell Therapy, Ghent University Hospital, 9000 Ghent, East-Flanders, Belgium
| | - Nele Lootens
- GMP Unit Cell Therapy, Ghent University Hospital, 9000 Ghent, East-Flanders, Belgium
| | - Marieke Brusseel
- GMP Unit Cell Therapy, Ghent University Hospital, 9000 Ghent, East-Flanders, Belgium
| | - Tasja Verstraete
- Respiratory Medicine, Ghent University Hospital, 9000 Ghent, East-Flanders, Belgium
| | - Jolien Buyle
- Respiratory Medicine, Ghent University Hospital, 9000 Ghent, East-Flanders, Belgium
| | - Eva Van Houtte
- GMP Unit Cell Therapy, Ghent University Hospital, 9000 Ghent, East-Flanders, Belgium
| | - Pam Devreker
- GMP Unit Cell Therapy, Ghent University Hospital, 9000 Ghent, East-Flanders, Belgium
| | - Kelly Heyns
- GMP Unit Cell Therapy, Ghent University Hospital, 9000 Ghent, East-Flanders, Belgium
| | - Stijn De Munter
- Department of Diagnostic Sciences, Ghent University, 9000 Ghent, East-Flanders, Belgium; Cancer Research Institute Ghent (CRIG), 9000 Ghent, Easy-Flanders, Belgium
| | - Sandra Van Lint
- Cancer Research Institute Ghent (CRIG), 9000 Ghent, Easy-Flanders, Belgium; Respiratory Medicine, Ghent University Hospital, 9000 Ghent, East-Flanders, Belgium
| | - Glenn Goetgeluk
- Department of Diagnostic Sciences, Ghent University, 9000 Ghent, East-Flanders, Belgium
| | - Sarah Bonte
- Cancer Research Institute Ghent (CRIG), 9000 Ghent, Easy-Flanders, Belgium; VIB-UGent Center for Medical Biotechnology, VIB, 9000 Ghent, East-Flanders, Belgium
| | - Lore Billiet
- Department of Diagnostic Sciences, Ghent University, 9000 Ghent, East-Flanders, Belgium; Cancer Research Institute Ghent (CRIG), 9000 Ghent, Easy-Flanders, Belgium
| | - Melissa Pille
- Department of Diagnostic Sciences, Ghent University, 9000 Ghent, East-Flanders, Belgium
| | - Hanne Jansen
- Department of Diagnostic Sciences, Ghent University, 9000 Ghent, East-Flanders, Belgium
| | - Eva Pascal
- Department of Diagnostic Sciences, Ghent University, 9000 Ghent, East-Flanders, Belgium; Cancer Research Institute Ghent (CRIG), 9000 Ghent, Easy-Flanders, Belgium
| | - Lucas Deseins
- Department of Diagnostic Sciences, Ghent University, 9000 Ghent, East-Flanders, Belgium; Cancer Research Institute Ghent (CRIG), 9000 Ghent, Easy-Flanders, Belgium
| | - Lies Vantomme
- Department of Biomolecular Medicine, Ghent University, 9000 Ghent, East-Flanders, Belgium
| | - Maarten Verdonckt
- Department of Diagnostic Sciences, Ghent University, 9000 Ghent, East-Flanders, Belgium
| | - Ria Roelandt
- VIB Single Cell Core, VIB, 9000/3000 Ghent/Leuven, East-Flanders/Flemish Brabant, Belgium
| | - Thomas Eekhout
- VIB Single Cell Core, VIB, 9000/3000 Ghent/Leuven, East-Flanders/Flemish Brabant, Belgium
| | - Niels Vandamme
- VIB Single Cell Core, VIB, 9000/3000 Ghent/Leuven, East-Flanders/Flemish Brabant, Belgium
| | - Georges Leclercq
- Department of Diagnostic Sciences, Ghent University, 9000 Ghent, East-Flanders, Belgium
| | - Tom Taghon
- Department of Diagnostic Sciences, Ghent University, 9000 Ghent, East-Flanders, Belgium
| | - Tessa Kerre
- Cancer Research Institute Ghent (CRIG), 9000 Ghent, Easy-Flanders, Belgium; VIB-UGent Center for Medical Biotechnology, VIB, 9000 Ghent, East-Flanders, Belgium; Hematology, Ghent University Hospital, 9000 Ghent, East-Flanders, Belgium
| | - Floris Vanommeslaeghe
- Nephrology, Ghent University Hospital, Ghent University, 9000 Ghent, East-Flanders, Belgium
| | - Annemieke Dhondt
- Nephrology, Ghent University Hospital, Ghent University, 9000 Ghent, East-Flanders, Belgium
| | - Liesbeth Ferdinande
- Cancer Research Institute Ghent (CRIG), 9000 Ghent, Easy-Flanders, Belgium; Pathology, Ghent University Hospital, 9000 Ghent, East-Flanders, Belgium
| | - Jo Van Dorpe
- Cancer Research Institute Ghent (CRIG), 9000 Ghent, Easy-Flanders, Belgium; Pathology, Ghent University Hospital, 9000 Ghent, East-Flanders, Belgium
| | - Liesbeth Desender
- Thoracic and Vascular Surgery, Ghent University Hospital, 9000 Ghent, East-Flanders, Belgium
| | - Frederic De Ryck
- Thoracic and Vascular Surgery, Ghent University Hospital, 9000 Ghent, East-Flanders, Belgium
| | - Frank Vermassen
- Thoracic and Vascular Surgery, Ghent University Hospital, 9000 Ghent, East-Flanders, Belgium
| | - Veerle Surmont
- Respiratory Medicine, Ghent University Hospital, 9000 Ghent, East-Flanders, Belgium
| | - Francis Impens
- Department of Biomolecular Medicine, Ghent University, 9000 Ghent, East-Flanders, Belgium; VIB-UGent Center for Medical Biotechnology, VIB, 9000 Ghent, East-Flanders, Belgium
| | - Björn Menten
- Cancer Research Institute Ghent (CRIG), 9000 Ghent, Easy-Flanders, Belgium; Department of Biomolecular Medicine, Ghent University, 9000 Ghent, East-Flanders, Belgium
| | - Karim Vermaelen
- Cancer Research Institute Ghent (CRIG), 9000 Ghent, Easy-Flanders, Belgium; Respiratory Medicine, Ghent University Hospital, 9000 Ghent, East-Flanders, Belgium.
| | - Bart Vandekerckhove
- Department of Diagnostic Sciences, Ghent University, 9000 Ghent, East-Flanders, Belgium; Cancer Research Institute Ghent (CRIG), 9000 Ghent, Easy-Flanders, Belgium; GMP Unit Cell Therapy, Ghent University Hospital, 9000 Ghent, East-Flanders, Belgium.
| |
Collapse
|
7
|
Kumari K, Singh A, Chaudhary A, Singh RK, Shanker A, Kumar V, Haque R. Neoantigen Identification and Dendritic Cell-Based Vaccines for Lung Cancer Immunotherapy. Vaccines (Basel) 2024; 12:498. [PMID: 38793749 PMCID: PMC11125796 DOI: 10.3390/vaccines12050498] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2024] [Revised: 04/29/2024] [Accepted: 05/01/2024] [Indexed: 05/26/2024] Open
Abstract
Immunotherapies can treat many cancers, including difficult-to-treat cases such as lung cancer. Due to its tolerability, long-lasting therapeutic responses, and efficacy in a wide spectrum of patients, immunotherapy can also help to treat lung cancer, which has few treatment choices. Tumor-specific antigens (TSAs) for cancer vaccinations and T-cell therapies are difficult to discover. Neoantigens (NeoAgs) from genetic mutations, irregular RNA splicing, protein changes, or viral genetic sequences in tumor cells provide a solution. NeoAgs, unlike TSAs, are non-self and can cause an immunological response. Next-generation sequencing (NGS) and bioinformatics can swiftly detect and forecast tumor-specific NeoAgs. Highly immunogenic NeoAgs provide personalized or generalized cancer immunotherapies. Dendritic cells (DCs), which originate and regulate T-cell responses, are widely studied potential immunotherapeutic therapies for lung cancer and other cancers. DC vaccines are stable, reliable, and safe in clinical trials. The purpose of this article is to evaluate the current status, limitations, and prospective clinical applications of DC vaccines, as well as the identification and selection of major histocompatibility complex (MHC) class I and II genes for NeoAgs. Our goal is to explain DC biology and activate DC manipulation to help researchers create extremely potent cancer vaccines for patients.
Collapse
Affiliation(s)
- Komal Kumari
- Department of Biotechnology, Central University of South Bihar, Gaya 824236, Bihar, India; (K.K.); (A.C.)
| | - Amarnath Singh
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA;
| | - Archana Chaudhary
- Department of Biotechnology, Central University of South Bihar, Gaya 824236, Bihar, India; (K.K.); (A.C.)
| | - Rakesh Kumar Singh
- Department of Biochemistry, Institute of Science, Banaras Hindu University, Varanasi 221005, Uttar Pradesh, India;
| | - Asheesh Shanker
- Department of Bioinformatics, Central University of South Bihar, Gaya 824236, Bihar, India
| | - Vinay Kumar
- Heart and Vascular Institute, Pennsylvania State University, Hershey Medical Center, Hershey, PA 17033, USA;
| | - Rizwanul Haque
- Department of Biotechnology, Central University of South Bihar, Gaya 824236, Bihar, India; (K.K.); (A.C.)
| |
Collapse
|
8
|
Fantini M, Tsang KY, Arlen PM. Generation of the therapeutic monoclonal antibody NEO-201, derived from a cancer vaccine, which targets human malignancies and immune suppressor cells. Expert Rev Vaccines 2024; 23:812-829. [PMID: 39186325 DOI: 10.1080/14760584.2024.2397011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 08/13/2024] [Accepted: 08/22/2024] [Indexed: 08/27/2024]
Abstract
INTRODUCTION Cancer vaccines stimulate the activation of specific humoral and cellular adaptive responses against cancer cells.Antibodies generated post vaccination can be isolated and further selected to develop highly specific and potent monoclonal antibodies (mAbs) against tumor-associated antigens. AREAS COVERED This review describes different types of cancer vaccines, the process of the generation of the mAb NEO-201 from the Hollinshead cancer vaccine platform, the characterization of the antigen recognized by NEO-201, the ability of NEO-201 to bind and mediate the killing of cancer cells and immunosuppressive cells (gMDSCs and Tregs) through ADCC and CDC, NEO-201 preclinical and clinical toxicity and efficacy. EXPERT OPINION To overcome the problem of poor clinical efficacy of cancer vaccines, due to the activity of immunosuppressive cells, cancer vaccines could be combined with other immunotherapeutics able to deplete immunosuppressive cells. Results from clinical trials, employing NEO-201 alone or in combination with pembrolizumab, showed that durable stabilization of disease after treatment was due to the ability of NEO-201 to target and reduce the percentage of circulating Tregs and gMDSCs.These findings provide compelling support to combine NEO-201 with cancer vaccines to reintegrate their ability to elicit a robust and durable immune adaptive response against cancer.
Collapse
|
9
|
Grogg J, Vernet R, Charrier E, Urwyler M, Von Rohr O, Saingier V, Courtout F, Lathuiliere A, Gaudenzio N, Engel A, Mach N. Engineering a versatile and retrievable cell macroencapsulation device for the delivery of therapeutic proteins. iScience 2023; 26:107372. [PMID: 37539029 PMCID: PMC10393802 DOI: 10.1016/j.isci.2023.107372] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 06/12/2023] [Accepted: 07/10/2023] [Indexed: 08/05/2023] Open
Abstract
Encapsulated cell therapy holds a great potential to deliver sustained levels of highly potent therapeutic proteins to patients and improve chronic disease management. A versatile encapsulation device that is biocompatible, scalable, and easy to administer, retrieve, or replace has yet to be validated for clinical applications. Here, we report on a cargo-agnostic, macroencapsulation device with optimized features for protein delivery. It is compatible with adherent and suspension cells, and can be administered and retrieved without burdensome surgical procedures. We characterized its biocompatibility and showed that different cell lines producing different therapeutic proteins can be combined in the device. We demonstrated the ability of cytokine-secreting cells encapsulated in our device and implanted in human skin to mobilize and activate antigen-presenting cells, which could potentially serve as an effective adjuvant strategy in cancer immunization therapies. We believe that our device may contribute to cell therapies for cancer, metabolic disorders, and protein-deficient diseases.
Collapse
Affiliation(s)
- Julien Grogg
- Department of Oncology, Geneva University Hospitals and Medical School, 1211 Geneva, Switzerland
- Centre for Translational Research in Onco-Hematology, Oncology Division, Geneva University Hospital and University of Geneva, Geneva, Switzerland
- MaxiVAX SA, Geneva, Switzerland
| | - Remi Vernet
- Department of Oncology, Geneva University Hospitals and Medical School, 1211 Geneva, Switzerland
- Centre for Translational Research in Onco-Hematology, Oncology Division, Geneva University Hospital and University of Geneva, Geneva, Switzerland
| | - Emily Charrier
- Department of Oncology, Geneva University Hospitals and Medical School, 1211 Geneva, Switzerland
- Centre for Translational Research in Onco-Hematology, Oncology Division, Geneva University Hospital and University of Geneva, Geneva, Switzerland
- MaxiVAX SA, Geneva, Switzerland
| | - Muriel Urwyler
- Department of Oncology, Geneva University Hospitals and Medical School, 1211 Geneva, Switzerland
- Centre for Translational Research in Onco-Hematology, Oncology Division, Geneva University Hospital and University of Geneva, Geneva, Switzerland
| | - Olivier Von Rohr
- Department of Oncology, Geneva University Hospitals and Medical School, 1211 Geneva, Switzerland
- Centre for Translational Research in Onco-Hematology, Oncology Division, Geneva University Hospital and University of Geneva, Geneva, Switzerland
| | - Valentin Saingier
- Department of Oncology, Geneva University Hospitals and Medical School, 1211 Geneva, Switzerland
- Centre for Translational Research in Onco-Hematology, Oncology Division, Geneva University Hospital and University of Geneva, Geneva, Switzerland
| | - Fabien Courtout
- Department of Oncology, Geneva University Hospitals and Medical School, 1211 Geneva, Switzerland
- Centre for Translational Research in Onco-Hematology, Oncology Division, Geneva University Hospital and University of Geneva, Geneva, Switzerland
| | - Aurelien Lathuiliere
- Department of Rehabilitation and Geriatrics, University of Geneva, 1211 Geneva, Switzerland
| | - Nicolas Gaudenzio
- Toulouse Institute for Infectious and Inflammatory Diseases (Infinity) INSERM UMR1291 - CNRS UMR5051 - University Toulouse III, Toulouse, France
- Genoskin SAS, Toulouse, France
| | - Adrien Engel
- Department of Oncology, Geneva University Hospitals and Medical School, 1211 Geneva, Switzerland
- Centre for Translational Research in Onco-Hematology, Oncology Division, Geneva University Hospital and University of Geneva, Geneva, Switzerland
- MaxiVAX SA, Geneva, Switzerland
| | - Nicolas Mach
- Department of Oncology, Geneva University Hospitals and Medical School, 1211 Geneva, Switzerland
- Centre for Translational Research in Onco-Hematology, Oncology Division, Geneva University Hospital and University of Geneva, Geneva, Switzerland
| |
Collapse
|
10
|
Chang R, Gulley JL, Fong L. Vaccinating against cancer: getting to prime time. J Immunother Cancer 2023; 11:jitc-2022-006628. [PMID: 37286302 DOI: 10.1136/jitc-2022-006628] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/15/2023] [Indexed: 06/09/2023] Open
Abstract
Immunotherapies, such as immune checkpoint inhibitors, cellular therapies, and T-cell engagers, have fundamentally changed our approach to treating cancer. However, successes with cancer vaccines have been more difficult to realize. While vaccines against specific viruses have been widely adopted to prevent the development of cancer, only two vaccines can improve survival in advanced disease: sipuleucel-T and talimogene laherparepvec. These represent the two approaches that have the most traction: vaccinating against cognate antigen and priming responses using tumors in situ. Here, we review the challenges and opportunities researchers face in developing therapeutic vaccines for cancer.
Collapse
Affiliation(s)
- Ryan Chang
- Hematology/Oncology, University of California, San Francisco, California, USA
| | - James L Gulley
- NCI, National Institutes of Health, Bethesda, Maryland, USA
| | - Lawrence Fong
- Hematology/Oncology, University of California, San Francisco, California, USA
| |
Collapse
|
11
|
Oladejo M, Paulishak W, Wood L. Synergistic potential of immune checkpoint inhibitors and therapeutic cancer vaccines. Semin Cancer Biol 2023; 88:81-95. [PMID: 36526110 DOI: 10.1016/j.semcancer.2022.12.003] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2022] [Revised: 12/06/2022] [Accepted: 12/09/2022] [Indexed: 12/15/2022]
Abstract
Cancer vaccines and immune checkpoint inhibitors (ICIs) function at different stages of the cancer immune cycle due to their distinct mechanisms of action. Therapeutic cancer vaccines enhance the activation and infiltration of cytotoxic immune cells into the tumor microenvironment (TME), while ICIs, prevent and/or reverse the dysfunction of these immune cells. The efficacy of both classes of immunotherapy has been evaluated in monotherapy, but they have been met with several challenges. Although therapeutic cancer vaccines can activate anti-tumor immune responses, these responses are susceptible to attenuation by immunoregulatory molecules. Similarly, ICIs are ineffective in the absence of tumor-infiltrating lymphocytes (TILs). Further, ICIs are often associated with immune-related adverse effects that may limit quality of life and compliance. However, the combination of the improved immunogenicity afforded by cancer vaccines and restrained immunosuppression provided by immune checkpoint inhibitors may provide a suitable platform for therapeutic synergism. In this review, we revisit the history and various classifications of therapeutic cancer vaccines. We also provide a summary of the currently approved ICIs. Finally, we provide mechanistic insights into the synergism between ICIs and cancer vaccines.
Collapse
Affiliation(s)
- Mariam Oladejo
- Department of Immunotherapeutics and Biotechnology, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Abilene, TX 79601, USA
| | - Wyatt Paulishak
- Department of Immunotherapeutics and Biotechnology, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Abilene, TX 79601, USA
| | - Laurence Wood
- Department of Immunotherapeutics and Biotechnology, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Abilene, TX 79601, USA.
| |
Collapse
|
12
|
Potluri HK, Ng TL, Newton MA, McNeel DG. GM-CSF elicits antibodies to tumor-associated proteins when used as a prostate cancer vaccine adjuvant. Cancer Immunol Immunother 2022; 71:2267-2275. [PMID: 35133464 PMCID: PMC9744072 DOI: 10.1007/s00262-022-03150-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Accepted: 01/07/2022] [Indexed: 12/14/2022]
Abstract
Antibody responses to off-target cancer-associated proteins have been detected following immunotherapies for cancer, suggesting these may be the result of antigen spread. We have previously reported that serum antibodies to prostate cancer-associated proteins were detectable using a high-throughput peptide array. We hypothesized that the breadth of antibody responses elicited by a vaccine could serve as a measure of the magnitude of its induced antigen spread. Consequently, sera from patients with prostate cancer, treated prior to or after vaccination in one of four separate clinical trials, were evaluated for antibody responses to an array of 177,604 peptides derived from over 1600 prostate cancer-associated gene products. Antibody responses to the same group of 5680 peptides previously reported were identified following vaccinations in which patients were administered GM-CSF as an adjuvant, but not with vaccine in the absence of GM-CSF. Hence, antibody responses to off-target proteins following vaccination may not necessarily serve as evidence of antigen spread and must be interpreted with particular caution following vaccine strategies that use GM-CSF, as GM-CSF appears to have direct effects on the production of antibodies. The evaluation of T cell responses to non-target antigens is likely a preferred approach for detection of immune-mediated antigen spread.
Collapse
Affiliation(s)
- Hemanth K Potluri
- Wisconsin Institutes for Medical Research, University of Wisconsin Carbone Cancer Center, 1111 Highland Avenue, Madison, WI, 53705, USA
| | - Tun L Ng
- Department of Biostatistics and Medical Informatics, University of Wisconsin, 1111 Highland Avenue, Madison, WI, 53705, USA
| | - Michael A Newton
- Department of Biostatistics and Medical Informatics, University of Wisconsin, 1111 Highland Avenue, Madison, WI, 53705, USA
| | - Douglas G McNeel
- Wisconsin Institutes for Medical Research, University of Wisconsin Carbone Cancer Center, 1111 Highland Avenue, Madison, WI, 53705, USA.
| |
Collapse
|
13
|
Development of Cancer Immunotherapies. Cancer Treat Res 2022; 183:1-48. [PMID: 35551655 DOI: 10.1007/978-3-030-96376-7_1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Cancer immunotherapy, or the utilization of components of the immune system to target and eliminate cancer, has become a highly active area of research in the past several decades and a common treatment strategy for several cancer types. The concept of harnessing the immune system for this purpose originated over 100 years ago when a physician by the name of William Coley successfully treated several of his cancer patients with a combination of live and attenuated bacteria, later known as "Coley's Toxins", after observing a subset of prior patients enter remission following their diagnosis with the common bacterial infection, erysipelas. However, it was not until late in the twentieth century that cancer immunotherapies were developed for widespread use, thereby transforming the treatment landscape of numerous cancer types. Pivotal studies elucidating molecular and cellular functions of immune cells, such as the discovery of IL-2 and production of monoclonal antibodies, fostered the development of novel techniques for studying the immune system and ultimately the development and approval of several cancer immunotherapies by the United States Food and Drug Association in the 1980s and 1990s, including the tuberculosis vaccine-Bacillus Calmette-Guérin, IL-2, and the CD20-targeting monoclonal antibody. Approval of the first therapeutic cancer vaccine, Sipuleucel-T, for the treatment of metastatic castration-resistant prostate cancer and the groundbreaking success and approval of immune checkpoint inhibitors and chimeric antigen receptor T cell therapy in the last decade, have driven an explosion of interest in and pursuit of novel cancer immunotherapy strategies. A broad range of modalities ranging from antibodies to adoptive T cell therapies is under investigation for the generalized treatment of a broad spectrum of cancers as well as personalized medicine. This chapter will focus on the recent advances, current strategies, and future outlook of immunotherapy development for the treatment of cancer.
Collapse
|
14
|
Bastin DJ, Quizi J, Kennedy MA, Kekre N, Auer RC. Current challenges in the manufacture of clinical-grade autologous whole cell vaccines for hematological malignancies. Cytotherapy 2022; 24:979-989. [PMID: 35562303 DOI: 10.1016/j.jcyt.2022.03.010] [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: 01/17/2022] [Revised: 03/21/2022] [Accepted: 03/21/2022] [Indexed: 11/03/2022]
Abstract
Autologous whole cell vaccines use a patient's own tumor cells as a source of antigen to elicit an anti-tumor immune response in vivo. Recently, the authors conducted a systematic review of clinical trials employing these products in hematological cancers that showed a favorable safety profile and trend toward efficacy. However, it was noted that manufacturing challenges limit both the efficacy and clinical implementation of these vaccine products. In the current literature review, the authors sought to define the issues surrounding the manufacture of autologous whole cell products for hematological cancers. The authors describe key factors, including the acquisition, culture, cryopreservation and transduction of malignant cells, that require optimization for further advancement of the field. Furthermore, the authors provide a summary of pre-clinical work that informs how the identified challenges may be overcome. The authors also highlight areas in which future basic research would be of benefit to the field. The goal of this review is to provide a roadmap for investigators seeking to advance the field of autologous cell vaccines as it applies to hematological malignancies.
Collapse
Affiliation(s)
- Donald J Bastin
- Cancer Therapeutics Program, Ottawa Hospital Research Institute, Ottawa, Canada; Schulich School of Medicine, Western University, London, Canada
| | - Jennifer Quizi
- Cancer Therapeutics Program, Ottawa Hospital Research Institute, Ottawa, Canada
| | - Michael A Kennedy
- Cancer Therapeutics Program, Ottawa Hospital Research Institute, Ottawa, Canada
| | - Natasha Kekre
- Cancer Therapeutics Program, Ottawa Hospital Research Institute, Ottawa, Canada; Faculty of Medicine, University of Ottawa, Ottawa, Canada
| | - Rebecca C Auer
- Cancer Therapeutics Program, Ottawa Hospital Research Institute, Ottawa, Canada; Faculty of Medicine, University of Ottawa, Ottawa, Canada; Department of Surgery, University of Ottawa, Ottawa, Canada; Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, Canada.
| |
Collapse
|
15
|
García-Pardo M, Gorria T, Malenica I, Corgnac S, Teixidó C, Mezquita L. Vaccine Therapy in Non-Small Cell Lung Cancer. Vaccines (Basel) 2022; 10:vaccines10050740. [PMID: 35632496 PMCID: PMC9146850 DOI: 10.3390/vaccines10050740] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 05/06/2022] [Accepted: 05/07/2022] [Indexed: 02/04/2023] Open
Abstract
Immunotherapy using immune checkpoint modulators has revolutionized the oncology field, emerging as a new standard of care for multiple indications, including non-small cell lung cancer (NSCLC). However, prognosis for patients with lung cancer is still poor. Although immunotherapy is highly effective in some cases, not all patients experience significant or durable responses, and further strategies are needed to improve outcomes. Therapeutic cancer vaccines are designed to exploit the body’s immune system to activate long-lasting memory against tumor cells that ensure tumor regression, with minimal toxicity. A unique feature of cancer vaccines lies in their complementary approach to boost antitumor immunity that could potentially act synergistically with immune checkpoint inhibitors (ICIs). However, single-line immunization against tumor epitopes with vaccine-based therapeutics has been disappointingly unsuccessful, to date, in lung cancer. The high level of success of several recent vaccines against SARS-CoV-2 has highlighted the evolving advances in science and technology in the vaccines field, raising hope that this strategy can be successfully applied to cancer treatments. In this review, we describe the biology behind the cancer vaccines, and discuss current evidence for the different types of therapeutic cancer vaccines in NSCLC, including their mechanisms of action, current clinical development, and future strategies.
Collapse
Affiliation(s)
| | - Teresa Gorria
- Medical Oncology Department, Hospital Clínic de Barcelona, 08036 Barcelona, Spain;
| | - Ines Malenica
- Laboratory of Hepatobiliary Immunopathology, IRCCS Humanitas Research Hospital, Via Manzoni 56, 20089 Milan, Italy;
| | - Stéphanie Corgnac
- INSERM UMR 1186, Integrative Tumor Immunology and Immunotherapy, Gustave Roussy, Faculté de Médecine, Université Paris-Saclay, 94805 Villejuif, France;
| | - Cristina Teixidó
- Department of Pathology, Hospital Clínic of Barcelona, University of Barcelona, Villarroel 170, 08036 Barcelona, Spain;
- Laboratory of Translational Genomics and Targeted Therapies in Solid Tumors, IDIBAPS, 08036 Barcelona, Spain
| | - Laura Mezquita
- Medical Oncology Department, Hospital Clínic de Barcelona, 08036 Barcelona, Spain;
- Laboratory of Translational Genomics and Targeted Therapies in Solid Tumors, IDIBAPS, 08036 Barcelona, Spain
- Department of Medicine, University of Barcelona, 08036 Barcelona, Spain
- Correspondence:
| |
Collapse
|
16
|
Lokhov PG, Lichtenberg S, Balashova EE. Changing Landscape of Cancer Vaccines-Novel Proteomics Platform for New Antigen Compositions. Int J Mol Sci 2022; 23:4401. [PMID: 35457221 PMCID: PMC9029553 DOI: 10.3390/ijms23084401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Revised: 04/12/2022] [Accepted: 04/12/2022] [Indexed: 11/16/2022] Open
Abstract
The creation of cancer vaccines is a constant priority for research and biotechnology. Therefore, the emergence of any new technology in this field is a significant event, especially because previous technologies have not yielded results. Recently, the development of a cancer vaccine has been complemented by a new proteomics technology platform that allows the creation of antigen compositions known as antigenic essences. Antigenic essence comprises a target fraction of cellular antigens, the composition of which is precisely controlled by peptide mass spectrometry and compared to the proteomic footprint of the target cells to ensure similarity. This proteomics platform offers potential for a massive upgrade of conventional cellular cancer vaccines. Antigenic essences have the same mechanism of action, but without the disadvantages, and with notable advantages such as precise targeting of the immune response, safety, controlled composition, improved immunogenicity, addressed MHC restriction, and extended range of vaccination doses. The present paper calls attention to this novel platform, stimulates discussion of the role of antigenic essence in vaccine development, and consolidates academic science with biotech capabilities. A brief description of the platform, list of cellular cancer vaccines suitable for the upgrade, main recommendations, limitations, and legal and ethical aspects of vaccine upgrade are reported here.
Collapse
Affiliation(s)
- Petr G. Lokhov
- Biobohemia, Inc., 1 Broadway, 14th Floor, Cambridge, MA 02142, USA; (S.L.); (E.E.B.)
- Institute of Biomedical Chemistry, 10 Building 8, Pogodinskaya Street, 119121 Moscow, Russia
| | - Steven Lichtenberg
- Biobohemia, Inc., 1 Broadway, 14th Floor, Cambridge, MA 02142, USA; (S.L.); (E.E.B.)
- Institute of Biomedical Chemistry, 10 Building 8, Pogodinskaya Street, 119121 Moscow, Russia
| | - Elena E. Balashova
- Biobohemia, Inc., 1 Broadway, 14th Floor, Cambridge, MA 02142, USA; (S.L.); (E.E.B.)
- Institute of Biomedical Chemistry, 10 Building 8, Pogodinskaya Street, 119121 Moscow, Russia
| |
Collapse
|
17
|
Hu X, Zhou W, Pi R, Zhao X, Wang W. Genetically modified cancer vaccines: Current status and future prospects. Med Res Rev 2022; 42:1492-1517. [PMID: 35235212 DOI: 10.1002/med.21882] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2020] [Revised: 12/13/2021] [Accepted: 01/23/2022] [Indexed: 02/05/2023]
Abstract
Vaccines can stimulate the immune system to protect individuals from infectious diseases. Moreover, vaccines have also been applied to the prevention and treatment of cancers. Due to advances in genetic engineering technology, cancer vaccines could be genetically modified to increase antitumor efficacy. Various genes could be inserted into cells to boost the immune response, such as cytokines, T cell costimulatory molecules, tumor-associated antigens, and tumor-specific antigens. Genetically modified cancer vaccines utilize innate and adaptive immune responses to induce durable antineoplastic capacity and prevent the recurrence. This review will discuss the major approaches used to develop genetically modified cancer vaccines and explore recent advances to increase the understanding of engineered cancer vaccines.
Collapse
Affiliation(s)
- Xiaoyi Hu
- Department of Gynecology and Obstetrics, Development and Related Disease of Women and Children Key Laboratory of Sichuan Province, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second Hospital, Sichuan University, Chengdu, P. R. China.,State Key Laboratory of Biotherapy and Cancer Center, Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, P. R. China
| | - Weilin Zhou
- State Key Laboratory of Biotherapy and Cancer Center, Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, P. R. China
| | - Ruyu Pi
- Department of Gynecology and Obstetrics, Development and Related Disease of Women and Children Key Laboratory of Sichuan Province, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second Hospital, Sichuan University, Chengdu, P. R. China.,State Key Laboratory of Biotherapy and Cancer Center, Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, P. R. China
| | - Xia Zhao
- Department of Gynecology and Obstetrics, Development and Related Disease of Women and Children Key Laboratory of Sichuan Province, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second Hospital, Sichuan University, Chengdu, P. R. China.,State Key Laboratory of Biotherapy and Cancer Center, Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, P. R. China
| | - Wei Wang
- State Key Laboratory of Biotherapy and Cancer Center, Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, P. R. China
| |
Collapse
|
18
|
Wiseman CL, Kharazi A, Sunkari VG, Galeas JL, Dozio V, Hashwah H, Macúchová E, Williams WV, Lacher MD. Regression of Breast Cancer Metastases Following Treatment with Irradiated SV-BR-1-GM, a GM-CSF Overexpressing Breast Cancer Cell Line: Intellectual Property and Immune Markers of Response. Recent Pat Anticancer Drug Discov 2022; 18:224-240. [PMID: 35593340 PMCID: PMC10009895 DOI: 10.2174/1574892817666220518123331] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 02/28/2022] [Accepted: 03/06/2022] [Indexed: 01/28/2023]
Abstract
BACKGROUND SV-BR-1-GM, derived from a patient with grade 2 (moderately differentiated) breast cancer, is a GM-CSF-secreting breast cancer cell line with properties of antigen-presenting cells. SV-BR-1-GM and next-generation versions are covered by several pending and granted patents. METHODS We report findings from an open-label phase I, single-arm pilot study with irradiated SV-BR-1-GM cells in 3 breast and 1 ovarian cancer subjects. Inoculations were preceded by lowdose intravenous cyclophosphamide and followed by interferon-alpha2b injections into the SVBR- 1-GM inoculation sites. We assessed both cellular and humoral immune responses, and measured expression levels of SV-BR-1-GM HLA alleles. RESULTS Treatment was generally safe and well tolerated. Immune responses were elicited universally. Overall survival was more than 33 months for three of the four patients. As previously reported, one patient had prompt regression of metastases in lung, breast, and soft tissue. Following cessation of treatment, the patient relapsed widely, including in the brain. Upon retreatment, rapid tumor response was again seen, including complete regression of brain metastases. Consistent with a role of Class II HLA in contributing to SV-BR-1-GM's mechanism of action, this patient allele-matched SV-BR-1-GM at the HLA-DRB1 and HLA-DRB3 loci. We are in the process of developing next-generation SV-BR-1-GM, expressing patient-specific HLAs. Patent applications were filed in various jurisdictions. Thus far, one is granted, in Japan. CONCLUSION A whole-cell immunotherapy regimen with SV-BR-1-GM cells induced regression of metastatic breast cancer. We develop intellectual property based on SV-BR-1-GM's predicted mechanism of action to develop additional whole-cell immunotherapies for cancer patients.
Collapse
Affiliation(s)
- Charles L. Wiseman
- BriaCell Therapeutics Corporation, 2929 Arch Street, 3 Floor, Philadelphia, PA, 19104, USA
| | - Alexander Kharazi
- Immunotherapy Laboratory, St. Vincent Medical Center, Los Angeles, CA, USA
| | - Vivekananda G. Sunkari
- BriaCell Therapeutics Corporation, 2929 Arch Street, 3 Floor, Philadelphia, PA, 19104, USA
| | - Jacqueline L. Galeas
- BriaCell Therapeutics Corporation, 2929 Arch Street, 3 Floor, Philadelphia, PA, 19104, USA
| | - Vito Dozio
- Operations Department, Biognosys AG, Wagistrasse 21, 8952, Schlieren, Switzerland
| | - Hind Hashwah
- Sales and Marketing Nebion AG, Hohlstrasse 515, 8048, Zurich, Switzerland
| | - Eva Macúchová
- Sales and Marketing Nebion AG, Hohlstrasse 515, 8048, Zurich, Switzerland
| | - William V. Williams
- BriaCell Therapeutics Corporation, 2929 Arch Street, 3 Floor, Philadelphia, PA, 19104, USA
| | - Markus D. Lacher
- BriaCell Therapeutics Corporation, 2929 Arch Street, 3 Floor, Philadelphia, PA, 19104, USA
| |
Collapse
|
19
|
James CA, Ronning P, Cullinan D, Cotto KC, Barnell EK, Campbell KM, Skidmore ZL, Sanford DE, Goedegebuure SP, Gillanders WE, Griffith OL, Hawkins WG, Griffith M. In silico epitope prediction analyses highlight the potential for distracting antigen immunodominance with allogeneic cancer vaccines. CANCER RESEARCH COMMUNICATIONS 2021; 1:115-126. [PMID: 35611186 PMCID: PMC9126504 DOI: 10.1158/2767-9764.crc-21-0029] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Allogeneic cancer vaccines are designed to induce antitumor immune responses with the goal of impacting tumor growth. Typical allogeneic cancer vaccines are produced by expansion of established cancer cell lines, transfection with vectors encoding immunostimulatory cytokines, and lethal irradiation. More than 100 clinical trials have investigated the clinical benefit of allogeneic cancer vaccines in various cancer types. Results show limited therapeutic benefit in clinical trials and currently there are no FDA approved allogeneic cancer vaccines. We used recently developed bioinformatics tools including the pVAC-seq suite of software tools to analyze DNA/RNA sequencing data from the TCGA to examine the repertoire of antigens presented by a typical allogeneic cancer vaccine, and to simulate allogeneic cancer vaccine clinical trials. Specifically, for each simulated clinical trial we modeled the repertoire of antigens presented by allogeneic cancer vaccines consisting of three hypothetical cancer cell lines to 30 patients with the same cancer type. Simulations were repeated ten times for each cancer type. Each tumor sample in the vaccine and the vaccine recipient was subjected to HLA typing, differential expression analyses for tumor associated antigens (TAAs), germline variant calling, and neoantigen prediction. These analyses provided a robust, quantitative comparison between potentially beneficial TAAs and neoantigens versus distracting antigens present in the allogeneic cancer vaccines. We observe that distracting antigens greatly outnumber shared TAAs and neoantigens, providing one potential explanation for the lack of observed responses to allogeneic cancer vaccines. This analysis provides additional rationale for the redirection of efforts towards a personalized cancer vaccine approach.
Collapse
Affiliation(s)
- C. Alston James
- Department of Surgery, Washington University School of Medicine, St. Louis, Missouri
| | - Peter Ronning
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri.,McDonnell Genome Institute, Washington University School of Medicine, St. Louis, Missouri
| | - Darren Cullinan
- Department of Surgery, Washington University School of Medicine, St. Louis, Missouri
| | - Kelsy C. Cotto
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Erica K. Barnell
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri.,McDonnell Genome Institute, Washington University School of Medicine, St. Louis, Missouri
| | - Katie M. Campbell
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Zachary L. Skidmore
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Dominic E. Sanford
- Department of Surgery, Washington University School of Medicine, St. Louis, Missouri.,Siteman Cancer Center, Washington University School of Medicine, St. Louis, Missouri
| | - S. Peter Goedegebuure
- Department of Surgery, Washington University School of Medicine, St. Louis, Missouri
| | - William E. Gillanders
- Department of Surgery, Washington University School of Medicine, St. Louis, Missouri.,Siteman Cancer Center, Washington University School of Medicine, St. Louis, Missouri
| | - Obi L. Griffith
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri.,McDonnell Genome Institute, Washington University School of Medicine, St. Louis, Missouri.,Siteman Cancer Center, Washington University School of Medicine, St. Louis, Missouri.,Department of Genetics, Washington University School of Medicine, St. Louis, Missouri.,CorrespondingAuthor: Malachi Griffith, McDonnell Genome Institute, 4444 Forest Park Avenue, Campus Box 8501, St. Louis, MO 63108. Phone: 314-286-1274; E-mail: ; Obi L. Griffith, McDonnell Genome Institute, 4444 Forest Park Avenue, Campus Box 8501, St. Louis, MO 63108. E-mail: ; and William G. Hawkins, McDonnell Genome Institute, 4444 Forest Park Avenue, Campus Box 8501, St. Louis, MO 63108. E-mail:
| | - William G. Hawkins
- Department of Surgery, Washington University School of Medicine, St. Louis, Missouri.,Siteman Cancer Center, Washington University School of Medicine, St. Louis, Missouri.,CorrespondingAuthor: Malachi Griffith, McDonnell Genome Institute, 4444 Forest Park Avenue, Campus Box 8501, St. Louis, MO 63108. Phone: 314-286-1274; E-mail: ; Obi L. Griffith, McDonnell Genome Institute, 4444 Forest Park Avenue, Campus Box 8501, St. Louis, MO 63108. E-mail: ; and William G. Hawkins, McDonnell Genome Institute, 4444 Forest Park Avenue, Campus Box 8501, St. Louis, MO 63108. E-mail:
| | - Malachi Griffith
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri.,McDonnell Genome Institute, Washington University School of Medicine, St. Louis, Missouri.,Siteman Cancer Center, Washington University School of Medicine, St. Louis, Missouri.,Department of Genetics, Washington University School of Medicine, St. Louis, Missouri.,CorrespondingAuthor: Malachi Griffith, McDonnell Genome Institute, 4444 Forest Park Avenue, Campus Box 8501, St. Louis, MO 63108. Phone: 314-286-1274; E-mail: ; Obi L. Griffith, McDonnell Genome Institute, 4444 Forest Park Avenue, Campus Box 8501, St. Louis, MO 63108. E-mail: ; and William G. Hawkins, McDonnell Genome Institute, 4444 Forest Park Avenue, Campus Box 8501, St. Louis, MO 63108. E-mail:
| |
Collapse
|
20
|
Sankarasubramanian S, Pfohl U, Regenbrecht CRA, Reinhard C, Wedeken L. Context Matters-Why We Need to Change From a One Size Fits all Approach to Made-to-Measure Therapies for Individual Patients With Pancreatic Cancer. Front Cell Dev Biol 2021; 9:760705. [PMID: 34805167 PMCID: PMC8599957 DOI: 10.3389/fcell.2021.760705] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Accepted: 10/18/2021] [Indexed: 12/12/2022] Open
Abstract
Pancreatic cancer is one of the deadliest cancers and remains a major unsolved health problem. While pancreatic ductal adenocarcinoma (PDAC) is associated with driver mutations in only four major genes (KRAS, TP53, SMAD4, and CDKN2A), every tumor differs in its molecular landscape, histology, and prognosis. It is crucial to understand and consider these differences to be able to tailor treatment regimens specific to the vulnerabilities of the individual tumor to enhance patient outcome. This review focuses on the heterogeneity of pancreatic tumor cells and how in addition to genetic alterations, the subsequent dysregulation of multiple signaling cascades at various levels, epigenetic and metabolic factors contribute to the oncogenesis of PDAC and compensate for each other in driving cancer progression if one is tackled by a therapeutic approach. This implicates that besides the need for new combinatorial therapies for PDAC, a personalized approach for treating this highly complex cancer is required. A strategy that combines both a target-based and phenotypic approach to identify an effective treatment, like Reverse Clinical Engineering® using patient-derived organoids, is discussed as a promising way forward in the field of personalized medicine to tackle this deadly disease.
Collapse
Affiliation(s)
| | - Ulrike Pfohl
- CELLphenomics GmbH, Berlin, Germany
- ASC Oncology GmbH, Berlin, Germany
- Institute for Molecular Bio Science, Goethe University Frankfurt Am Main, Frankfurt, Germany
| | - Christian R. A. Regenbrecht
- CELLphenomics GmbH, Berlin, Germany
- ASC Oncology GmbH, Berlin, Germany
- Institute for Pathology, Universitätsklinikum Göttingen, Göttingen, Germany
| | | | - Lena Wedeken
- CELLphenomics GmbH, Berlin, Germany
- ASC Oncology GmbH, Berlin, Germany
| |
Collapse
|
21
|
ROS Cocktails as an Adjuvant for Personalized Antitumor Vaccination? Vaccines (Basel) 2021; 9:vaccines9050527. [PMID: 34069708 PMCID: PMC8161309 DOI: 10.3390/vaccines9050527] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 05/15/2021] [Accepted: 05/17/2021] [Indexed: 12/22/2022] Open
Abstract
Cancer is the second leading cause of death worldwide. Today, the critical role of the immune system in tumor control is undisputed. Checkpoint antibody immunotherapy augments existing antitumor T cell activity with durable clinical responses in many tumor entities. Despite the presence of tumor-associated antigens and neoantigens, many patients have an insufficient repertoires of antitumor T cells. Autologous tumor vaccinations aim at alleviating this defect, but clinical success is modest. Loading tumor material into autologous dendritic cells followed by their laboratory expansion and therapeutic vaccination is promising, both conceptually and clinically. However, this process is laborious, time-consuming, costly, and hence less likely to solve the global cancer crisis. Therefore, it is proposed to re-focus on personalized anticancer vaccinations to enhance the immunogenicity of autologous therapeutic tumor vaccines. Recent work re-established the idea of using the alarming agents of the immune system, oxidative modifications, as an intrinsic adjuvant to broaden the antitumor T cell receptor repertoire in cancer patients. The key novelty is the use of gas plasma, a multi-reactive oxygen and nitrogen species-generating technology, for diversifying oxidative protein modifications in a, so far, unparalleled manner. This significant innovation has been successfully used in proof-of-concept studies and awaits broader recognition and implementation to explore its chances and limitations of providing affordable personalized anticancer vaccines in the future. Such multidisciplinary advance is timely, as the current COVID-19 crisis is inexorably reflecting the utmost importance of innovative and effective vaccinations in modern times.
Collapse
|
22
|
Stevens D, Ingels J, Van Lint S, Vandekerckhove B, Vermaelen K. Dendritic Cell-Based Immunotherapy in Lung Cancer. Front Immunol 2021; 11:620374. [PMID: 33679709 PMCID: PMC7928408 DOI: 10.3389/fimmu.2020.620374] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Accepted: 12/29/2020] [Indexed: 12/24/2022] Open
Abstract
Lung cancer remains the leading cause of cancer-related death worldwide. The advent of immune checkpoint inhibitors has led to a paradigm shift in the treatment of metastatic non-small cell and small cell lung cancer. However, despite prolonged overall survival, only a minority of the patients derive clinical benefit from these treatments suggesting that the full anti-tumoral potential of the immune system is not being harnessed yet. One way to overcome this problem is to combine immune checkpoint blockade with different strategies aimed at inducing or restoring cellular immunity in a tumor-specific, robust, and durable way. Owing to their unique capacity to initiate and regulate T cell responses, dendritic cells have been extensively explored as tools for immunotherapy in many tumors, including lung cancer. In this review, we provide an update on the nearly twenty years of experience with dendritic cell-based immunotherapy in lung cancer. We summarize the main results from the early phase trials and give an overview of the future perspectives within this field.
Collapse
Affiliation(s)
- Dieter Stevens
- Respiratory Medicine - Thoracic Oncology Cluster, Ghent University Hospital, Ghent, Belgium.,Respiratory Medicine - Tumor Immunology Laboratory, Ghent University, Ghent, Belgium
| | - Joline Ingels
- Department of Diagnostic Sciences, Ghent University Hospital, Ghent, Belgium
| | - Sandra Van Lint
- Respiratory Medicine - Tumor Immunology Laboratory, Ghent University, Ghent, Belgium.,Cancer Research Institute Ghent, Ghent University, Ghent, Belgium
| | - Bart Vandekerckhove
- Department of Diagnostic Sciences, Ghent University Hospital, Ghent, Belgium.,GMP Cell Therapy Unit, Department of Regenerative Medicine, Ghent University Hospital, Ghent, Belgium.,Cancer Research Institute Ghent, Ghent University, Ghent, Belgium
| | - Karim Vermaelen
- Respiratory Medicine - Thoracic Oncology Cluster, Ghent University Hospital, Ghent, Belgium.,Respiratory Medicine - Tumor Immunology Laboratory, Ghent University, Ghent, Belgium.,Cancer Research Institute Ghent, Ghent University, Ghent, Belgium
| |
Collapse
|
23
|
Al-Share B, Hammad N, Diab M. Pancreatic adenocarcinoma: molecular drivers and the role of targeted therapy. Cancer Metastasis Rev 2021; 40:355-371. [PMID: 33398620 DOI: 10.1007/s10555-020-09948-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Accepted: 12/15/2020] [Indexed: 01/05/2023]
Abstract
Prognosis from pancreatic ductal adenocarcinoma (PDAC) continues to be poor despite the many efforts channeled to improve its management. Although the mainstay treatment is still traditional chemotherapy, recent advances highlighted a promising potential for targeted therapy in the management of this disease. Those advances emphasize the significance of timely genomic profiling of tumor tissue as well as germline testing of patients to identify potential markers of targeted therapy. While targeted therapy is reserved for a relatively small subset of patients with PDAC, ongoing research is uncovering additional markers, and targeted agents, that will hopefully translate to better outcomes for patients.
Collapse
Affiliation(s)
- Bayan Al-Share
- Department of Oncology, Wayne State University, Karmanos Cancer Institute, Detroit, MI, USA
| | - Nour Hammad
- Department of Oncology, Ascension Providence Hospital and Medical Center/Michigan State University/Collage of Human Medicine, Southfield, MI, USA
| | - Maria Diab
- Department of Oncology, Emory University, Atlanta, GA, USA.
| |
Collapse
|
24
|
Igarashi Y, Sasada T. Cancer Vaccines: Toward the Next Breakthrough in Cancer Immunotherapy. J Immunol Res 2020; 2020:5825401. [PMID: 33282961 PMCID: PMC7685825 DOI: 10.1155/2020/5825401] [Citation(s) in RCA: 106] [Impact Index Per Article: 21.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 09/26/2020] [Accepted: 09/30/2020] [Indexed: 12/13/2022] Open
Abstract
Until now, three types of well-recognized cancer treatments have been developed, i.e., surgery, chemotherapy, and radiotherapy; these either remove or directly attack the cancer cells. These treatments can cure cancer at earlier stages but are frequently ineffective for treating cancer in the advanced or recurrent stages. Basic and clinical research on the tumor microenvironment, which consists of cancerous, stromal, and immune cells, demonstrates the critical role of antitumor immunity in cancer development and progression. Cancer immunotherapies have been proposed as the fourth cancer treatment. In particular, clinical application of immune checkpoint inhibitors, such as anti-CTLA-4 and anti-PD-1/PD-L1 antibodies, in various cancer types represents a major breakthrough in cancer therapy. Nevertheless, accumulating data regarding immune checkpoint inhibitors demonstrate that these are not always effective but are instead only effective in limited cancer populations. Indeed, several issues remain to be solved to improve their clinical efficacy; these include low cancer cell antigenicity and poor infiltration and/or accumulation of immune cells in the cancer microenvironment. Therefore, to accelerate the further development of cancer immunotherapies, more studies are necessary. In this review, we will summarize the current status of cancer immunotherapies, especially cancer vaccines, and discuss the potential problems and solutions for the next breakthrough in cancer immunotherapy.
Collapse
Affiliation(s)
- Yuka Igarashi
- Kanagawa Cancer Center, Research Institute, Division of Cancer Immunotherapy, Japan
| | - Tetsuro Sasada
- Kanagawa Cancer Center, Research Institute, Division of Cancer Immunotherapy, Japan
- Kanagawa Cancer Center, Cancer Vaccine and Immunotherapy Center, Japan
| |
Collapse
|
25
|
Abstract
Cancer gene therapy emerged as a promising treatment modality 3 decades ago. However, the failure of the first gene therapy trials in cancer treatment has decreased its popularity. Likewise, immunotherapy has followed a similar course. While it was a popular and promising treatment with IL-2 and interferon and cancer vaccines in the 1980s, it later lost its popularity. Immunotherapy became one of the main options for cancer treatment with the successful use of immune checkpoint inhibitors in clinics approximately 10 years ago. The success of immunotherapy has increased even more with the introduction of cancer gene therapy methods in this area. With the identification of the oncolytic herpes simplex virus and Chimeric antigen receptor (CAR) T-cells, immune gene therapy has become an essential modality in cancer treatments such as surgery, radiotherapy, chemotherapy, and targeted therapies.
Collapse
Affiliation(s)
- Hakan Akbulut
- Department of Basic Oncology, Ankara University Cancer Research Institute, Ankara, Turkey,Department of Medical Oncology, Ankara University School of Medicine, Ankara, Turkey
| |
Collapse
|
26
|
Palata O, Podzimkova Hradilova N, Mysiková D, Kutna B, Mrazkova H, Lischke R, Spisek R, Adkins I. Detection of tumor antigens and tumor-antigen specific T cells in NSCLC patients: Correlation of the quality of T cell responses with NSCLC subtype. Immunol Lett 2020; 219:46-53. [PMID: 31931024 DOI: 10.1016/j.imlet.2020.01.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Revised: 11/29/2019] [Accepted: 01/07/2020] [Indexed: 11/15/2022]
Abstract
Allogeneic cancer cell lines serve as universal source of tumor-associated antigens in cancer vaccines. Immunogenic high hydrostatic pressure-killed cancer cells derived from cell lines can be used for the generation of dendritic cell (DC)-based active cellular immunotherapy of non-small cell lung cancer (NSCLC). We investigated the expression of 12 known NSCLC tumor-associated antigens (TAA) (CEA, MAGE-A1, MAGE-A3, MAGE-A4, PRAME, hTERT, HER2, MUC1, Survivin, STEAP1, SOX2 and NY-ESO-1) in 6 NSCLC cell lines as candidates for the generation of DC-based lung cancer vaccine. We showed that the selected antigenic profile of these cell lines overlaps to various degrees with that of primary NSCLC tumors (n = 52), indicating that 4 out of 6 NSCLC cell lines would be suitable for DC-based vaccine generation. We further investigated the presence of TAA-specific T cells in blood of NSCLC patients (n = 32) using commercially available peptide mixes in an in vitro stimulation assay. IFN-γ+CD8+ and IFN-γ+CD4+ T cell responses to all antigens were detected in NSCLC patients. Interestingly, despite higher TAA expression in squamous cell carcinoma (SCC) the responsiveness of patients' T cells to stimulation was significantly lower in SCC patients than in adenocarcinoma (AC) patients. This suggests qualitative differences in T cell functionality between NSCLC subtypes. Based on this study, and in order to maximize the amount of treatable patients, we selected a mix of H520 and H522 NSCLC cell lines for DC-based vaccine preparation. We also established a minimal panel of antigenic peptide mixes (CEA, hTERT, PRAME, HER2) for immunomonitoring of T cell responses during the DC-based lung cancer immunotherapy in Phase I lung cancer clinical trial (NCT02470468).
Collapse
Affiliation(s)
- Ondrej Palata
- Sotio, Prague, Czech Republic; Department of Immunology, 2nd Faculty of Medicine and University Hospital Motol, Charles University, Czech Republic
| | - Nada Podzimkova Hradilova
- Sotio, Prague, Czech Republic; Department of Immunology, 2nd Faculty of Medicine and University Hospital Motol, Charles University, Czech Republic
| | - Dagmar Mysiková
- Thoracic and Lung Transplantation Division, 3rd Department of Surgery, First Faculty of Medicine, Charles University in Prague and University Hospital Motol, Czech Republic
| | - Beata Kutna
- Thoracic and Lung Transplantation Division, 3rd Department of Surgery, First Faculty of Medicine, Charles University in Prague and University Hospital Motol, Czech Republic
| | - Hana Mrazkova
- Thoracic and Lung Transplantation Division, 3rd Department of Surgery, First Faculty of Medicine, Charles University in Prague and University Hospital Motol, Czech Republic
| | - Robert Lischke
- Thoracic and Lung Transplantation Division, 3rd Department of Surgery, First Faculty of Medicine, Charles University in Prague and University Hospital Motol, Czech Republic
| | - Radek Spisek
- Sotio, Prague, Czech Republic; Department of Immunology, 2nd Faculty of Medicine and University Hospital Motol, Charles University, Czech Republic
| | - Irena Adkins
- Sotio, Prague, Czech Republic; Department of Immunology, 2nd Faculty of Medicine and University Hospital Motol, Charles University, Czech Republic.
| |
Collapse
|
27
|
Holt GE, Daftarian P. Non-small-cell lung cancer homing peptide-labeled dendrimers selectively transfect lung cancer cells. Immunotherapy 2019; 10:1349-1360. [PMID: 30474481 DOI: 10.2217/imt-2018-0078] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
AIM Lung cancer gene therapies require reagents to selectively transfect lung tumors after systemic administration. MATERIALS & METHODS We created a reagent called NSCLC-NP by attaching a peptide with binding affinity for lung cancer to polyamidoamine dendrimers. The positively charged dendrimers electrostatically bind negatively charged nucleic acids, inhibit endogenous nucleases and transfect cells targeted by the attached peptide. RESULTS In vitro, NSCLC-NP complexed to DNA plasmids bound and transfected three human lung cancer cell lines producing protein expression of the plasmid's gene. In vivo, systemically administered NSCLC-NP selectively transfected lung cancer cells growing in RAG1KO mice. CONCLUSION The capability of NSCLC-NP to selectively transfect lung cancer allows its future use as a vehicle to implement human lung cancer gene therapy strategies.
Collapse
Affiliation(s)
- Gregory E Holt
- Department of Medicine, Division of Pulmonary, Allergy, Critical Care & Sleep Medicine, University of Miami, Miami, FL, USA.,Department of Medicine, Division of Pulmonology, Miami VA Medical Center, Miami, FL, USA
| | - Pirouz Daftarian
- Department of Ophthalmology, University of Miami, FL, USA.,JSR Micro Life Sciences, Sunnyvale, CA 94089, USA
| |
Collapse
|
28
|
Alonso-Guallart P, Zitsman JS, Stern J, Kofman SB, Woodland D, Ho SH, Sondermeijer HP, Bühler L, Griesemer A, Sykes M, Duran-Struuck R. Characterization, biology, and expansion of regulatory T cells in the Cynomolgus macaque for preclinical studies. Am J Transplant 2019; 19:2186-2198. [PMID: 30768842 PMCID: PMC6658340 DOI: 10.1111/ajt.15313] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Revised: 01/16/2019] [Accepted: 02/04/2019] [Indexed: 01/25/2023]
Abstract
Reliable in vitro expansion protocols of regulatory T cells (Tregs) are needed for clinical use. We studied the biology of Mauritian Cynomolgus macaque (MCM) Tregs and developed four in vitro Treg expansion protocols for translational studies. Tregs expanded 3000-fold when artificial antigen presenting cells (aAPCs) expressing human CD80, CD58 and CD32 were used throughout the culture. When donor peripheral blood mononuclear cells (PBMCs) were used as the single source of APCs followed by aAPCs, Tregs expanded 2000-fold. Tregs from all protocols suppressed the proliferation of anti-CD2CD3CD28 bead-stimulated autologous PBMCs albeit with different potencies, varying from 1:2-1:4 Treg:PBMC ratios, up to >1:32. Reculture of cryopreserved Tregs permitted reexpansion with improved suppressive activity. Occasionally, CD8 contamination was observed and resolved by resorting. Specificity studies showed greater suppression of stimulation by anti-CD2CD3CD28 beads of PBMCs from the same donor used for stimulation during the Treg cultures and of autologous cells than of third-party PBMC responders. Similar to humans, the Treg-specific demethylated region (TSDR) within the Foxp3 locus correlated with suppressive activity and expression of Foxp3. Contrary to humans, FoxP3 expression did not correlate with CD45RA or CD127 expression. In summary, we have characterized MCM Tregs and developed four Treg expansion protocols that can be used for preclinical applications.
Collapse
Affiliation(s)
- Paula Alonso-Guallart
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University Medical Center, New York, NY, United States
| | - Jonah S. Zitsman
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University Medical Center, New York, NY, United States
| | - Jeffrey Stern
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University Medical Center, New York, NY, United States
| | - Sigal B. Kofman
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University Medical Center, New York, NY, United States
| | - David Woodland
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University Medical Center, New York, NY, United States
| | - Siu-Hong Ho
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University Medical Center, New York, NY, United States
| | - Hugo P. Sondermeijer
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University Medical Center, New York, NY, United States.,Current address; Department of Physiology, Maastricht University, Maastricht, The Netherlands
| | - Leo Bühler
- Current address; Department of Surgery, University Hospital of Geneva, Switzerland
| | - Adam Griesemer
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University Medical Center, New York, NY, United States.,Department of Surgery, Columbia University Medical Center, New York, NY, United States
| | - Megan Sykes
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University Medical Center, New York, NY, United States.,Department of Surgery, Columbia University Medical Center, New York, NY, United States.,Department of Microbiology and Immunology, Columbia University Medical Center, New York, NY, United States
| | - Raimon Duran-Struuck
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University Medical Center, New York, NY, United States.,Current address; Department of Pathobiology, University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
29
|
Leduc C, Quoix E. [Vaccines for the treatment of non-small cell lung cancer]. Rev Mal Respir 2019; 36:415-425. [PMID: 30902445 DOI: 10.1016/j.rmr.2018.12.003] [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: 07/20/2017] [Accepted: 05/31/2018] [Indexed: 11/26/2022]
Abstract
Antigen-specific immunotherapy also known as cancer vaccination offers a novel approach for the treatment of non-small cell lung cancer patients. It relies on specific priming of the immune system in order to provoke or increase adaptive antitumor immune response against the vaccine component. Several molecules have been developed in lung cancer, based on whole-tumor cells, dendritic cells, peptides, recombinant proteins, or viral vectors. The aim of this review is to describe the mechanism of action of these vaccines and the results of the main clinical studies.
Collapse
Affiliation(s)
- C Leduc
- Service de pneumologie, CHRU de Strasbourg, Strasbourg, France
| | - E Quoix
- Service de pneumologie, CHRU de Strasbourg, Strasbourg, France.
| |
Collapse
|
30
|
Abstract
The promise of immune-based therapies to treat cancer has been realized over the last several years with several breakthrough therapies, including T-cell checkpoint inhibitors and chimeric antigen receptor (CAR)-T cell therapies. While cancer vaccines have been investigated for many decades, to date only one has been approved in the USA as a treatment for existing cancer. The failure of several anti-tumor vaccines in large phase III trials has led many to question their future role in cancer treatment. Trials to date have demonstrated that many cancer vaccines can elicit tumor-specific T cells, but these T cells may be insufficient to mediate substantial anti-tumor effects without concurrent blockade of tumor-resistance mechanisms. Emerging data from preclinical models and clinical trials demonstrate that cancer vaccines have greater activity in low-volume disease and in combination with other immune-modulating therapies, including T-cell checkpoint blockade, targeting these resistance mechanisms. Because T-cell checkpoint therapies likely require the presence or activity of tumor-specific T cells, cancer vaccines may be optimal agents to use in combination to enable these therapies to work for greater numbers of patients. Future trials will explore optimal vaccine approaches and antigens that work best in combination treatment approaches and in earlier stages of disease.
Collapse
|
31
|
Boss C, Bouche N, De Marchi U. Encapsulated Optically Responsive Cell Systems: Toward Smart Implants in Biomedicine. Adv Healthc Mater 2018; 7:e1701148. [PMID: 29283209 DOI: 10.1002/adhm.201701148] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Revised: 11/06/2017] [Indexed: 01/09/2023]
Abstract
Managing increasingly prevalent chronic diseases will require close continuous monitoring of patients. Cell-based biosensors may be used for implantable diagnostic systems to monitor health status. Cells are indeed natural sensors in the body. Functional cellular systems can be maintained in the body for long-term implantation using cell encapsulation technology. By taking advantage of recent progress in miniaturized optoelectronic systems, the genetic engineering of optically responsive cells may be combined with cell encapsulation to generate smart implantable cell-based sensing systems. In biomedical research, cell-based biosensors may be used to study cell signaling, therapeutic effects, and dosing of bioactive molecules in preclinical models. Today, a wide variety of genetically encoded fluorescent sensors have been developed for real-time imaging of living cells. Here, recent developments in genetically encoded sensors, cell encapsulation, and ultrasmall optical systems are highlighted. The integration of these components in a new generation of biosensors is creating innovative smart in vivo cell-based systems, bringing novel perspectives for biomedical research and ultimately allowing unique health monitoring applications.
Collapse
Affiliation(s)
- Christophe Boss
- Device EngineeringNestlé Institute of Health Sciences EPFL Innovation Park Lausanne CH‐1015 Switzerland
| | - Nicolas Bouche
- Device EngineeringNestlé Institute of Health Sciences EPFL Innovation Park Lausanne CH‐1015 Switzerland
| | - Umberto De Marchi
- Mitochondrial FunctionNestlé Institute of Health Sciences EPFL Innovation Park Lausanne CH‐1015 Switzerland
| |
Collapse
|
32
|
Rojas-Sepúlveda D, Tittarelli A, Gleisner MA, Ávalos I, Pereda C, Gallegos I, González FE, López MN, Butte JM, Roa JC, Fluxá P, Salazar-Onfray F. Tumor lysate-based vaccines: on the road to immunotherapy for gallbladder cancer. Cancer Immunol Immunother 2018; 67:1897-1910. [PMID: 29600445 PMCID: PMC6244977 DOI: 10.1007/s00262-018-2157-5] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Accepted: 03/26/2018] [Indexed: 12/14/2022]
Abstract
Immunotherapy based on checkpoint blockers has proven survival benefits in patients with melanoma and other malignancies. Nevertheless, a significant proportion of treated patients remains refractory, suggesting that in combination with active immunizations, such as cancer vaccines, they could be helpful to improve response rates. During the last decade, we have used dendritic cell (DC) based vaccines where DCs loaded with an allogeneic heat-conditioned melanoma cell lysate were tested in a series of clinical trials. In these studies, 60% of stage IV melanoma DC-treated patients showed immunological responses correlating with improved survival. Further studies showed that an essential part of the clinical efficacy was associated with the use of conditioned lysates. Gallbladder cancer (GBC) is a high-incidence malignancy in South America. Here, we evaluated the feasibility of producing effective DCs using heat-conditioned cell lysates derived from gallbladder cancer cell lines (GBCCL). By characterizing nine different GBCCLs and several fresh tumor tissues, we found that they expressed some tumor-associated antigens such as CEA, MUC-1, CA19-9, Erb2, Survivin, and several carcinoembryonic antigens. Moreover, heat-shock treatment of GBCCLs induced calreticulin translocation and release of HMGB1 and ATP, both known to act as danger signals. Monocytes stimulated with combinations of conditioned lysates exhibited a potent increase of DC-maturation markers. Furthermore, conditioned lysate-matured DCs were capable of strongly inducing CD4+ and CD8+ T cell activation, in both allogeneic and autologous cell co-cultures. Finally, in vitro stimulated CD8+ T cells recognize HLA-matched GBCCLs. In summary, GBC cell lysate-loaded DCs may be considered for future immunotherapy approaches.
Collapse
Affiliation(s)
- Daniel Rojas-Sepúlveda
- Disciplinary Program of Immunology, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Independencia 1027, building H, Third floor, 8380453, Santiago, Chile.,Millennium Institute on Immunology and Immunotherapy, Universidad de Chile, 8380453, Santiago, Chile.,Faculty of Science, Universidad San Sebastián, Lota 2465, 7510157, Santiago, Chile
| | - Andrés Tittarelli
- Disciplinary Program of Immunology, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Independencia 1027, building H, Third floor, 8380453, Santiago, Chile.,Millennium Institute on Immunology and Immunotherapy, Universidad de Chile, 8380453, Santiago, Chile
| | - María Alejandra Gleisner
- Disciplinary Program of Immunology, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Independencia 1027, building H, Third floor, 8380453, Santiago, Chile.,Millennium Institute on Immunology and Immunotherapy, Universidad de Chile, 8380453, Santiago, Chile
| | - Ignacio Ávalos
- Disciplinary Program of Immunology, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Independencia 1027, building H, Third floor, 8380453, Santiago, Chile.,Millennium Institute on Immunology and Immunotherapy, Universidad de Chile, 8380453, Santiago, Chile
| | - Cristián Pereda
- Disciplinary Program of Immunology, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Independencia 1027, building H, Third floor, 8380453, Santiago, Chile.,Millennium Institute on Immunology and Immunotherapy, Universidad de Chile, 8380453, Santiago, Chile
| | - Iván Gallegos
- Pathological Anatomy Service, Clinic Hospital, Universidad de Chile, 8380456, Santiago, Chile
| | - Fermín Eduardo González
- Millennium Institute on Immunology and Immunotherapy, Universidad de Chile, 8380453, Santiago, Chile.,Department of Conservative Dentistry, Faculty of Dentistry, Universidad de Chile, 8380492, Santiago, Chile
| | - Mercedes Natalia López
- Disciplinary Program of Immunology, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Independencia 1027, building H, Third floor, 8380453, Santiago, Chile.,Millennium Institute on Immunology and Immunotherapy, Universidad de Chile, 8380453, Santiago, Chile
| | - Jean Michel Butte
- Department of Surgery, Fundación Arturo López Pérez, Institute of Oncology, 7500921, Santiago, Chile
| | - Juan Carlos Roa
- Department of Pathology, School of Medicine, Pontificia Universidad Católica de Chile, 8330023, Santiago, Chile.,Center for Investigation in Translational Oncology (CITO), Advanced Center for Chronic Diseases (ACCDiS), School of Medicine, Pontificia Universidad Católica de Chile, 8330023, Santiago, Chile
| | - Paula Fluxá
- Disciplinary Program of Immunology, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Independencia 1027, building H, Third floor, 8380453, Santiago, Chile.,Millennium Institute on Immunology and Immunotherapy, Universidad de Chile, 8380453, Santiago, Chile
| | - Flavio Salazar-Onfray
- Disciplinary Program of Immunology, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Independencia 1027, building H, Third floor, 8380453, Santiago, Chile. .,Millennium Institute on Immunology and Immunotherapy, Universidad de Chile, 8380453, Santiago, Chile.
| |
Collapse
|
33
|
Sanborn RE, Ross HJ, Aung S, Acheson A, Moudgil T, Puri S, Hilton T, Fisher B, Coffey T, Paustian C, Neuberger M, Walker E, Hu HM, Urba WJ, Fox BA. A pilot study of an autologous tumor-derived autophagosome vaccine with docetaxel in patients with stage IV non-small cell lung cancer. J Immunother Cancer 2017; 5:103. [PMID: 29258618 PMCID: PMC5735525 DOI: 10.1186/s40425-017-0306-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2017] [Accepted: 11/29/2017] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Tumor-derived autophagosome vaccines (DRibbles) have the potential to broaden immune response to poorly immunogenic tumors. METHODS Autologous vaccine generated from tumor cells harvested from pleural effusions was administered to patients with advanced NSCLC with the objectives of assessing safety and immune response. Four patients were vaccinated and evaluable for immune response; each received two to four doses of vaccine. Study therapy included two cycles of docetaxel 75 mg/m2 on days 1 and 29 to treat the tumor, release hidden antigens and produce lymphopenia. DRibbles were to be administered intradermally on days 14, 43, 57, 71, and 85, together with GM-CSF (50 μg/d x 6d, administered via SQ mini pump). Peripheral blood was tested for immune parameters at baseline and at each vaccination. RESULTS Three of four patients had tumor cells available for testing. Autologous tumor-specific immune response was seen in two of the three, manifested by IL-5 (1 patient after 3 doses), and IFN-γ, TNF-α, IL-5, IL-10 (after 4 doses in one patient). All 4 patients had evidence of specific antibody responses against potential tumor antigens. All patients came off study after 4 or fewer vaccine treatments due to progression of disease. No significant immune toxicities were seen during the course of the study. CONCLUSIONS DRibble vaccine given with GM-CSF appeared safe and capable of inducing an immune response against tumor cells in this small, pilot study. There was no evidence of efficacy in this small poor-prognosis patient population, with treatment not feasible. Trial registration NCT00850785, initial registration date February 23, 2009.
Collapse
Affiliation(s)
- Rachel E. Sanborn
- Robert W. Franz Cancer Research Center, Earle A. Chiles Research Institute, Providence Portland Medical Center, Portland, OR USA
- Earle A. Chiles Research Institute, N.E. Glisan Street, 2N35, Portland, OR 97213 USA
| | | | - Sandra Aung
- UbiVac, Portland, OR USA
- Present address: Nektar Therapeutics, San Francisco, USA
| | - Anupama Acheson
- Robert W. Franz Cancer Research Center, Earle A. Chiles Research Institute, Providence Portland Medical Center, Portland, OR USA
| | - Tarsem Moudgil
- Laboratory of Molecular and Tumor Immunology, Robert W. Franz Cancer Research Center, Earle A. Chiles Research Institute, Providence Cancer Center, Portland, OR USA
| | - Sachin Puri
- Laboratory of Molecular and Tumor Immunology, Robert W. Franz Cancer Research Center, Earle A. Chiles Research Institute, Providence Cancer Center, Portland, OR USA
| | | | - Brenda Fisher
- Robert W. Franz Cancer Research Center, Earle A. Chiles Research Institute, Providence Portland Medical Center, Portland, OR USA
| | - Todd Coffey
- Robert W. Franz Cancer Research Center, Earle A. Chiles Research Institute, Providence Portland Medical Center, Portland, OR USA
| | - Christopher Paustian
- Laboratory of Molecular and Tumor Immunology, Robert W. Franz Cancer Research Center, Earle A. Chiles Research Institute, Providence Cancer Center, Portland, OR USA
| | - Michael Neuberger
- Laboratory of Molecular and Tumor Immunology, Robert W. Franz Cancer Research Center, Earle A. Chiles Research Institute, Providence Cancer Center, Portland, OR USA
- Present address: Department of General, Visceral and Transplantation Surgery, University of Munich, Campus Grosshadern, Munich, Germany
| | - Edwin Walker
- Immunological Monitoring Laboratory, Robert W. Franz Cancer Research Center, Earle A. Chiles Research Institute, Providence Cancer Center, Portland, OR USA
| | - Hong-Ming Hu
- UbiVac, Portland, OR USA
- Laboratory of Cancer Immunobiology, Robert W. Franz Cancer Research Center, Earle A. Chiles Research Institute, Providence Cancer Center, Portland, OR USA
| | - Walter J. Urba
- Robert W. Franz Cancer Research Center, Earle A. Chiles Research Institute, Providence Portland Medical Center, Portland, OR USA
| | - Bernard A. Fox
- UbiVac, Portland, OR USA
- Laboratory of Molecular and Tumor Immunology, Robert W. Franz Cancer Research Center, Earle A. Chiles Research Institute, Providence Cancer Center, Portland, OR USA
- Department of Molecular Microbiology and Immunology; and Knight Cancer Institute, Oregon Health and Science University, Portland, OR USA
| |
Collapse
|
34
|
Saliba H, Heurtault B, Bouharoun-Tayoun H, Flacher V, Frisch B, Fournel S, Chamat S. Enhancing tumor specific immune responses by transcutaneous vaccination. Expert Rev Vaccines 2017; 16:1079-1094. [DOI: 10.1080/14760584.2017.1382357] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Affiliation(s)
- Hanadi Saliba
- Laboratory of Design and Application of Bioactive Molecules, University of Strasbourg, Illkirch Cedex, France
- Laboratory of Immunology, Lebanese University, Fanar, Lebanon
| | - Béatrice Heurtault
- Laboratory of Design and Application of Bioactive Molecules, University of Strasbourg, Illkirch Cedex, France
| | | | - Vincent Flacher
- Laboratory of Immunopathology and Therapeutic Chemistry, Institut de Biologie Moléculaire et Cellulaire, Strasbourg, France
| | - Benoît Frisch
- Laboratory of Design and Application of Bioactive Molecules, University of Strasbourg, Illkirch Cedex, France
| | - Sylvie Fournel
- Laboratory of Design and Application of Bioactive Molecules, University of Strasbourg, Illkirch Cedex, France
| | - Soulaima Chamat
- Laboratory of Immunology, Lebanese University, Fanar, Lebanon
- Faculty of Medicine, Lebanese University, Hadath, Lebanon
| |
Collapse
|
35
|
Sondhi D, Stiles KM, De BP, Crystal RG. Genetic Modification of the Lung Directed Toward Treatment of Human Disease. Hum Gene Ther 2017; 28:3-84. [PMID: 27927014 DOI: 10.1089/hum.2016.152] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Genetic modification therapy is a promising therapeutic strategy for many diseases of the lung intractable to other treatments. Lung gene therapy has been the subject of numerous preclinical animal experiments and human clinical trials, for targets including genetic diseases such as cystic fibrosis and α1-antitrypsin deficiency, complex disorders such as asthma, allergy, and lung cancer, infections such as respiratory syncytial virus (RSV) and Pseudomonas, as well as pulmonary arterial hypertension, transplant rejection, and lung injury. A variety of viral and non-viral vectors have been employed to overcome the many physical barriers to gene transfer imposed by lung anatomy and natural defenses. Beyond the treatment of lung diseases, the lung has the potential to be used as a metabolic factory for generating proteins for delivery to the circulation for treatment of systemic diseases. Although much has been learned through a myriad of experiments about the development of genetic modification of the lung, more work is still needed to improve the delivery vehicles and to overcome challenges such as entry barriers, persistent expression, specific cell targeting, and circumventing host anti-vector responses.
Collapse
Affiliation(s)
- Dolan Sondhi
- Department of Genetic Medicine, Weill Cornell Medical College , New York, New York
| | - Katie M Stiles
- Department of Genetic Medicine, Weill Cornell Medical College , New York, New York
| | - Bishnu P De
- Department of Genetic Medicine, Weill Cornell Medical College , New York, New York
| | - Ronald G Crystal
- Department of Genetic Medicine, Weill Cornell Medical College , New York, New York
| |
Collapse
|
36
|
Aguiar PN, De Mello RA, Barreto CMN, Perry LA, Penny-Dimri J, Tadokoro H, Lopes GDL. Immune checkpoint inhibitors for advanced non-small cell lung cancer: emerging sequencing for new treatment targets. ESMO Open 2017; 2:e000200. [PMID: 29209522 PMCID: PMC5703392 DOI: 10.1136/esmoopen-2017-000200] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2017] [Revised: 06/08/2017] [Accepted: 06/09/2017] [Indexed: 12/20/2022] Open
Abstract
Lung cancer is the leading cause of cancer-related deaths in the world. Immune checkpoint inhibitors (ICI) stimulate cytotoxic lymphocyte activity against tumour cells. These agents are available for the treatment of non-small cell lung cancer (NSCLC) after failure of platinum-based therapy. One recent study has demonstrated that ICI monotherapy was superior to platinum-based chemotherapy for first-line treatment. Nevertheless, this benefit was only for a minority of the population (30%) whose tumour programmed death receptor ligand-1 (PD-L1) expression was above 50%. Therefore, several strategies are under investigation. One option for patients with PD-L1 expression lower than 50% may be the combination of ICI with platinum-based chemotherapy or with ICIs against different targets. However, all of these combinations are at an early stage of investigation and may be very expensive or toxic, producing several harmful adverse events.
Collapse
Affiliation(s)
| | - Ramon Andrade De Mello
- Department of Biomedical Sciences and Medicine, Division of Medical Oncology, University of Algarve, Faro, Portugal
| | | | | | | | - Hakaru Tadokoro
- Division of Medical Oncology, Federal University of São Paulo, São Paulo, Brazil
| | | |
Collapse
|
37
|
Yan WL, Shen KY, Tien CY, Chen YA, Liu SJ. Recent progress in GM-CSF-based cancer immunotherapy. Immunotherapy 2017; 9:347-360. [PMID: 28303764 DOI: 10.2217/imt-2016-0141] [Citation(s) in RCA: 144] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Cancer immunotherapy is a growing field. GM-CSF, a potent cytokine promoting the differentiation of myeloid cells, can also be used as an immunostimulatory adjuvant to elicit antitumor immunity. Additionally, GM-CSF is essential for the differentiation of dendritic cells, which are responsible for processing and presenting tumor antigens for the priming of antitumor cytotoxic T lymphocytes. Some strategies have been developed for GM-CSF-based cancer immunotherapy in clinical practice: GM-CSF monotherapy, GM-CSF-secreting cancer cell vaccines, GM-CSF-fused tumor-associated antigen protein-based vaccines, GM-CSF-based DNA vaccines and GM-CSF combination therapy. GM-CSF also contributes to the regulation of immunosuppression in the tumor microenvironment. This review provides recommendations regarding GM-CSF-based cancer immunotherapy.
Collapse
Affiliation(s)
- Wan-Lun Yan
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei, Taiwan.,National Institute of Infectious Diseases & Vaccinology, National Health Research Institutes, No. 35 Keyan Road, Zhunan Town, Miaoli County 350, Taiwan
| | - Kuan-Yin Shen
- National Institute of Infectious Diseases & Vaccinology, National Health Research Institutes, No. 35 Keyan Road, Zhunan Town, Miaoli County 350, Taiwan.,Graduate Instituteof Biomedical Sciences, China Medical University, Taichung, Taiwan
| | - Chun-Yuan Tien
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei, Taiwan.,National Institute of Infectious Diseases & Vaccinology, National Health Research Institutes, No. 35 Keyan Road, Zhunan Town, Miaoli County 350, Taiwan
| | - Yu-An Chen
- Graduate Instituteof Biomedical Sciences, China Medical University, Taichung, Taiwan
| | - Shih-Jen Liu
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei, Taiwan.,National Institute of Infectious Diseases & Vaccinology, National Health Research Institutes, No. 35 Keyan Road, Zhunan Town, Miaoli County 350, Taiwan
| |
Collapse
|
38
|
Kazemi MH, Raoofi Mohseni S, Hojjat-Farsangi M, Anvari E, Ghalamfarsa G, Mohammadi H, Jadidi-Niaragh F. Adenosine and adenosine receptors in the immunopathogenesis and treatment of cancer. J Cell Physiol 2017; 233:2032-2057. [DOI: 10.1002/jcp.25873] [Citation(s) in RCA: 97] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2017] [Accepted: 02/21/2017] [Indexed: 12/18/2022]
Affiliation(s)
- Mohammad H. Kazemi
- Student Research Committee, Department of Immunology, School of Medicine; Iran University of Medical Sciences (IUMS); Tehran Iran
- Immunology Research Center; Tabriz University of Medical Sciences; Tabriz Iran
| | - Sahar Raoofi Mohseni
- Department of Immunology, School of Public Health; Tehran University of Medical Sciences; Tehran Iran
| | - Mohammad Hojjat-Farsangi
- Department of Oncology-Pathology, Immune and Gene Therapy Lab, Cancer Center Karolinska (CCK); Karolinska University Hospital Solna and Karolinska Institute; Stockholm Sweden
- Department of Immunology, School of Medicine; Bushehr University of Medical Sciences; Bushehr Iran
| | - Enayat Anvari
- Faculty of Medicine, Department of Physiology; Ilam University of Medical Sciences; Ilam Iran
| | - Ghasem Ghalamfarsa
- Medicinal Plants Research Center; Yasuj University of Medical Sciences; Yasuj Iran
| | - Hamed Mohammadi
- Immunology Research Center; Tabriz University of Medical Sciences; Tabriz Iran
- Faculty of Medicine, Department of Immunology; Tabriz University of Medical Sciences; Tabriz Iran
| | - Farhad Jadidi-Niaragh
- Immunology Research Center; Tabriz University of Medical Sciences; Tabriz Iran
- Department of Immunology, School of Public Health; Tehran University of Medical Sciences; Tehran Iran
- Faculty of Medicine, Department of Immunology; Tabriz University of Medical Sciences; Tabriz Iran
- Drug Applied Research Center; Tabriz University of Medical Sciences; Tabriz Iran
| |
Collapse
|
39
|
|
40
|
Hosen N, Maeda T, Hashii Y, Tsuboi A, Nishida S, Nakata J, Oji Y, Oka Y, Sugiyama H. Wilms tumor 1 peptide vaccination after hematopoietic stem cell transplant in leukemia patients. Stem Cell Investig 2016; 3:90. [PMID: 28078270 DOI: 10.21037/sci.2016.11.08] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2016] [Accepted: 11/07/2016] [Indexed: 01/03/2023]
Abstract
Although the prognosis of leukemia patients after allogeneic hematopoietic stem cell transplantation (HSCT) has greatly improved, relapse is still a major cause of death after HSCT. Cancer vaccines may have the potential to enhance the graft-versus-leukemia (GVL) effect. The post-allogeneic HSCT period provides a unique platform for vaccination, because (I) tumor burden is minimal, (II) lymphopenia allows for rapid expansion of cytotoxic T cells (CTLs), (III) donor-derived CTLs are not exhausted, (IV) inflammation is caused by alloreactions, and (V) the abundance of regulatory T cells is low due to their late recovery. Tumor cell lysates, dendritic cells (DCs), and peptides derived from leukemia-associated antigens (LAAs) have been used as vaccines. Clinical trials with several types of vaccines for post-HSCT patients revealed that the vaccination induced an immunological response and might benefit patients with minimal residual disease; however, the efficacy of this approach must be examined in randomized studies. In addition, it is important to consider the combination of cancer vaccine with checkpoint antibodies, recently shown to be useful in treating leukemia relapse after HSCT.
Collapse
Affiliation(s)
- Naoki Hosen
- Department of Cancer stem cell Biology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Tetsuo Maeda
- Department of Hematology and Oncology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Yoshiko Hashii
- Department of Pediatrics, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Akihiro Tsuboi
- Department of Cancer Immunotherapy, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Sumiyuki Nishida
- Department of Respiratory Medicine, Allergy and Rheumatic Diseases, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Jun Nakata
- Department of Cancer Immunotherapy, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Yusuke Oji
- Department of Cancer stem cell Biology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Yoshihiro Oka
- Department of Cancer Immunology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Haruo Sugiyama
- Department of Cancer Immunology, Osaka University Graduate School of Medicine, Osaka, Japan
| |
Collapse
|
41
|
Mountzios G, Linardou H, Kosmidis P. Immunotherapy in non-small cell lung cancer: the clinical impact of immune response and targeting. ANNALS OF TRANSLATIONAL MEDICINE 2016; 4:268. [PMID: 27563655 PMCID: PMC4971380 DOI: 10.21037/atm.2016.06.24] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Accepted: 06/20/2016] [Indexed: 12/21/2022]
Abstract
Non-small cell lung cancer (NSCLC) remains the leading cause of cancer-related death worldwide. In recent years, through a better understanding of the interactions between the immune system and tumor cells (TC), immunotherapy has emerged as a promising therapeutic strategy. Chemotherapy has long been reported to interfere with the immune response to the tumor and conversely, anti-tumor immunity may add to those effects. Anti-tumor vaccines, such as MAGE-A3, Tecetomide, TG4010, CIMAvax, tumor cell vaccines and dendritic cell (DC) vaccines emerged as potent inducers of the immune response against the tumor. More recently the approval of the anti-programmed cell death 1 (anti-PD-1) monoclonal antibodies nivolumab and pembrolizumab for previously treated advanced squamous and non-squamous NSCLC, as well as other immune checkpoint inhibitors delivering promising results, has radically transformed the therapeutic landscape of NSCLC. Combination strategies now appear as the next step. Notwithstanding these successes, immunotherapy still holds significant drawbacks and currently several improvements are needed before routine use in clinical practice, including identification of robust biomarkers for optimal patient selection, as well as defining the best way to evaluate response.
Collapse
Affiliation(s)
- Giannis Mountzios
- Department of Medical Oncology, University of Athens School of Medicine, Athens, Greece
| | - Helena Linardou
- 1 Oncology Department, Metropolitan Hospital, Piraeus, Greece
| | | |
Collapse
|
42
|
Mayor M, Yang N, Sterman D, Jones DR, Adusumilli PS. Immunotherapy for non-small cell lung cancer: current concepts and clinical trials. Eur J Cardiothorac Surg 2016; 49:1324-33. [PMID: 26516195 PMCID: PMC4851162 DOI: 10.1093/ejcts/ezv371] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Revised: 09/04/2015] [Accepted: 09/21/2015] [Indexed: 01/05/2023] Open
Abstract
Recent successes in immunotherapeutic strategies are being investigated to combat cancers that have less than ideal responses to standard of care treatment, such as non-small-cell lung cancer. In this paper, we summarize concepts and the current status of immunotherapy for non-small cell lung cancer, including salient features of the major categories of immunotherapy-monoclonal antibody therapy, immune checkpoint blockade, immunotoxins, anticancer vaccines, and adoptive cell therapy.
Collapse
Affiliation(s)
- Marissa Mayor
- Thoracic Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Neng Yang
- Thoracic Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Daniel Sterman
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, New York University, New York, NY, USA
| | - David R Jones
- Thoracic Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Prasad S Adusumilli
- Thoracic Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA Center for Cell Engineering, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| |
Collapse
|
43
|
Curry WT, Gorrepati R, Piesche M, Sasada T, Agarwalla P, Jones PS, Gerstner ER, Golby AJ, Batchelor TT, Wen PY, Mihm MC, Dranoff G. Vaccination with Irradiated Autologous Tumor Cells Mixed with Irradiated GM-K562 Cells Stimulates Antitumor Immunity and T Lymphocyte Activation in Patients with Recurrent Malignant Glioma. Clin Cancer Res 2016; 22:2885-96. [PMID: 26873960 DOI: 10.1158/1078-0432.ccr-15-2163] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2015] [Accepted: 02/04/2016] [Indexed: 12/13/2022]
Abstract
PURPOSE Recurrent malignant glioma carries a dismal prognosis, and novel therapies are needed. We examined the feasibility and safety of vaccination with irradiated autologous glioma cells mixed with irradiated GM-K562 cells in patients undergoing craniotomy for recurrent malignant glioma. EXPERIMENTAL DESIGN We initiated a phase I study examining the safety of 2 doses of GM-K562 cells mixed with autologous cells. Primary endpoints were feasibility and safety. Feasibility was defined as the ability for 60% of enrolled subjects to initiate vaccination. Dose-limiting toxicity was assessed via a 3+3 dose-escalation format, examining irradiated tumor cells mixed with 5 × 10(6) GM-K562 cells or 1 × 10(7) GM-K562 cells. Eligibility required a priori indication for resection of a recurrent high-grade glioma. We measured biological activity by measuring delayed type hypersensitivity (DTH) responses, humoral immunity against tumor-associated antigens, and T-lymphocyte activation. RESULTS Eleven patients were enrolled. Sufficient numbers of autologous tumor cells were harvested in 10 patients, all of whom went on to receive vaccine. There were no dose-limiting toxicities. Vaccination strengthened DTH responses to irradiated autologous tumor cells in most patients, and vigorous humoral responses to tumor-associated angiogenic cytokines were seen as well. T-lymphocyte activation was seen with significantly increased expression of CTLA-4, PD-1, 4-1BB, and OX40 by CD4(+) cells and PD-1 and 4-1BB by CD8(+) cells. Activation was coupled with vaccine-associated increase in the frequency of regulatory CD4(+) T lymphocytes. CONCLUSIONS Vaccination with irradiated autologous tumor cells mixed with GM-K562 cells is feasible, well tolerated, and active in patients with recurrent malignant glioma. Clin Cancer Res; 22(12); 2885-96. ©2016 AACR.
Collapse
Affiliation(s)
- William T Curry
- Department of Neurosurgery, Massachusetts General Hospital, Boston, Massachusetts. Cancer Center, Massachusetts General Hospital, Boston, Massachusetts. Harvard Medical School, Boston, Massachusetts.
| | - Ramana Gorrepati
- Department of Neurosurgery, Massachusetts General Hospital, Boston, Massachusetts
| | - Matthias Piesche
- Department of Medicine, Dana Farber Cancer Institute, Boston, Massachusetts. Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Tetsuro Sasada
- Cancer Vaccine Center, Dana Farber Cancer Institute, Boston, Massachusetts
| | - Pankaj Agarwalla
- Department of Neurosurgery, Massachusetts General Hospital, Boston, Massachusetts
| | - Pamela S Jones
- Department of Neurosurgery, Massachusetts General Hospital, Boston, Massachusetts
| | - Elizabeth R Gerstner
- Cancer Center, Massachusetts General Hospital, Boston, Massachusetts. Harvard Medical School, Boston, Massachusetts
| | - Alexandra J Golby
- Harvard Medical School, Boston, Massachusetts. Department of Neurosurgery, Brigham and Women's Hospital, Boston, Massachusetts
| | - Tracy T Batchelor
- Cancer Center, Massachusetts General Hospital, Boston, Massachusetts. Harvard Medical School, Boston, Massachusetts
| | - Patrick Y Wen
- Harvard Medical School, Boston, Massachusetts. Division of Neuro-oncology, Dana Farber Cancer Institute, Boston, Massachusetts
| | - Martin C Mihm
- Harvard Medical School, Boston, Massachusetts. Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts
| | - Glenn Dranoff
- Harvard Medical School, Boston, Massachusetts. Department of Medicine, Dana Farber Cancer Institute, Boston, Massachusetts. Cancer Vaccine Center, Dana Farber Cancer Institute, Boston, Massachusetts
| |
Collapse
|
44
|
Lievense L, Aerts J, Hegmans J. Immune Therapy. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2016; 893:59-90. [PMID: 26667339 DOI: 10.1007/978-3-319-24223-1_4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Lung cancer has long been considered an unsuitable target for immunotherapy due to its proposed immunoresistant properties. However, recent evidence has shown that anti-tumor immune responses can occur in lung cancer patients, paving the way for lung cancer as a novel target for immunotherapy. In order to take full advantage of the potential of immunotherapy, research is focusing on the presence and function of various immunological cell types in the tumor microenvironment. Immune cells which facilitate or inhibit antitumor responses have been identified and their prognostic value in lung cancer has been established. Knowledge regarding these pro- and anti-tumor immune cells and their mechanisms of action has facilitated the identification of numerous potential immunotherapeutic strategies and opportunities for intervention. A plethora of immunotherapeutic approaches is currently being developed and studied in lung cancer patients and phase 3 clinical trials are ongoing. Many different immunotherapies have shown promising clinical effects in patients with limited and advanced stage lung cancer, however, future years will have to tell whether immunotherapy will earn its place in the standard treatment of lung cancer.
Collapse
Affiliation(s)
- Lysanne Lievense
- Department of Pulmonary Medicine, Erasmus MC Cancer Institute, Dr. Molewaterplein 50, Rotterdam, 3015 GD, The Netherlands
| | - Joachim Aerts
- Department of Pulmonary Medicine, Erasmus MC Cancer Institute, Dr. Molewaterplein 50, Rotterdam, 3015 GD, The Netherlands
| | - Joost Hegmans
- Department of Pulmonary Medicine, Erasmus MC Cancer Institute, Dr. Molewaterplein 50, Rotterdam, 3015 GD, The Netherlands.
| |
Collapse
|
45
|
|
46
|
Domingues D, Turner A, Silva MD, Marques DS, Mellidez JC, Wannesson L, Mountzios G, de Mello RA. Immunotherapy and lung cancer: current developments and novel targeted therapies. Immunotherapy 2015; 6:1221-35. [PMID: 25496336 DOI: 10.2217/imt.14.82] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Non-small-cell lung cancer (NSCLC) is a highly prevalent and aggressive disease. In the metastatic setting, major advances include the incorporation of immunotherapy and targeted therapies into the clinician's therapeutic armamentarium. Standard chemotherapeutic regimens have long been reported to interfere with the immune response to the tumor; conversely, antitumor immunity may add to the effects of those therapies. The aim of immunotherapy is to specifically enhance the immune response directed to the tumor. Recently, many trials addressed the role of such therapies for metastatic NSCLC treatment: ipilimumab, tremelimumab, nivolumab and lambrolizumab are immunotherapeutic agents of main interest in this field. In addition, anti-tumor vaccines, such as MAGE-A3, Tecetomide, TG4010, CIMAvax, ganglioside vaccines, tumor cell vaccines and dendritic cell vaccines, emerged as potent inducers of immune response against the tumor. The current work aims to address the most recent developments regarding these innovative immunotherapies and their implementation in the treatment of metastatic NSCLC.
Collapse
Affiliation(s)
- Duarte Domingues
- Department of Medical Oncology, Portuguese Oncology Institute (IPO PORTO), Rua Dr. António Bernardino de Almeida, 4200-072, Porto, Portugal
| | | | | | | | | | | | | | | |
Collapse
|
47
|
Encapsulated cellular implants for recombinant protein delivery and therapeutic modulation of the immune system. Int J Mol Sci 2015; 16:10578-600. [PMID: 26006227 PMCID: PMC4463663 DOI: 10.3390/ijms160510578] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2015] [Revised: 04/28/2015] [Accepted: 04/30/2015] [Indexed: 12/15/2022] Open
Abstract
Ex vivo gene therapy using retrievable encapsulated cellular implants is an effective strategy for the local and/or chronic delivery of therapeutic proteins. In particular, it is considered an innovative approach to modulate the activity of the immune system. Two recently proposed therapeutic schemes using genetically engineered encapsulated cells are discussed here: the chronic administration of monoclonal antibodies for passive immunization against neurodegenerative diseases and the local delivery of a cytokine as an adjuvant for anti-cancer vaccines.
Collapse
|
48
|
Whole Tumor Antigen Vaccines: Where Are We? Vaccines (Basel) 2015; 3:344-72. [PMID: 26343191 PMCID: PMC4494356 DOI: 10.3390/vaccines3020344] [Citation(s) in RCA: 197] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2015] [Revised: 04/13/2015] [Accepted: 04/16/2015] [Indexed: 12/19/2022] Open
Abstract
With its vast amount of uncharacterized and characterized T cell epitopes available for activating CD4+ T helper and CD8+ cytotoxic lymphocytes simultaneously, whole tumor antigen represents an attractive alternative source of antigens as compared to tumor-derived peptides and full-length recombinant tumor proteins for dendritic cell (DC)-based immunotherapy. Unlike defined tumor-derived peptides and proteins, whole tumor lysate therapy is applicable to all patients regardless of their HLA type. DCs are essentially the master regulators of immune response, and are the most potent antigen-presenting cell population for priming and activating naïve T cells to target tumors. Because of these unique properties, numerous DC-based immunotherapies have been initiated in the clinics. In this review, we describe the different types of whole tumor antigens that we could use to pulse DCs ex vivo and in vivo. We also discuss the different routes of delivering whole tumor antigens to DCs in vivo and activating them with toll-like receptor agonists.
Collapse
|
49
|
Wang M, Cao JX, Liu YS, Xu BL, Li D, Zhang XY, Li JL, Liu JL, Wang HB, Wang ZX. Evaluation of tumour vaccine immunotherapy for the treatment of advanced non-small cell lung cancer: a systematic meta-analysis. BMJ Open 2015; 5:e006321. [PMID: 25872936 PMCID: PMC4401843 DOI: 10.1136/bmjopen-2014-006321] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
OBJECTIVES Our meta-analysis performed a systematic evaluation on the therapeutic efficacy and safety of tumour vaccines for the treatment of advanced non-small cell lung cancer (NSCLC). DESIGN Systematic review and meta-analysis of randomised controlled trials (RCT). DATA SOURCES PubMed, the Cochrane Center Register of Controlled Trials, Science Direct and EMBASE were searched from January 1980 until January 2015. ELIGIBILITY CRITERIA FOR SELECTING STUDIES RCT were included; the control arm had to receive either placebo or chemotherapy or no treatment. MAIN OUTCOME MEASURES The quality of the data from individual papers was assessed for overall survival (OS), clinical response rate and side effects. RESULTS Overall, 11 RCT of advanced NSCLC with a total of 3986 patients were conducted for meta-analysis. The results showed that the vaccine arm significantly extended primary endpoint median overall survival compared with control group (p<0.00001) (HR 0.760; 95% CI 0.644 to 0.896; p=0.001). Three subgroup patients with tumour vaccine at 1-year, 2-year and 3-year survival rates also gained significant benefits compared with their corresponding control group (p=0.0004, 0.03 and 0.19, respectively). Besides, a significant improvement in median time to progression (TTP), median progression-free survival (PFS) and a trend of improvement in objective response rate were observed after tumour vaccine treatment (p=0.001, 0.005 and 0.05, respectively; median PFS HR 0.842; 95% CI 0.744 to 0.954; p=0.007). A few severe adverse effects occurred in the tumour vaccine group, but fewer side effects were observed in the vaccine group compared with the control group (p<0.00001). CONCLUSIONS Taken together, NSCLC tumour vaccines markedly prolong median OS (p<0.00001), median TTP (p=0.001) and median PFS (p=0.005), improve clinical response rate (p=0.05) and lessen adverse side effects (p<0.00001). Our meta-analysis suggests tumour vaccines improve the efficacy of the treatment, and also provide superiority in treatment of patients with advanced NSCLC among a variety of immunotherapy strategies.
Collapse
Affiliation(s)
- Min Wang
- Biotherapy Center, General Hospital of Beijing Military Command, Beijing, People's Republic of China
| | - Jun-Xia Cao
- Biotherapy Center, General Hospital of Beijing Military Command, Beijing, People's Republic of China
| | - Yi-Shan Liu
- Biotherapy Center, General Hospital of Beijing Military Command, Beijing, People's Republic of China
| | - Bei-Lei Xu
- Biotherapy Center, General Hospital of Beijing Military Command, Beijing, People's Republic of China
| | - Duo Li
- Biotherapy Center, General Hospital of Beijing Military Command, Beijing, People's Republic of China
| | - Xiao-Yan Zhang
- Biotherapy Center, General Hospital of Beijing Military Command, Beijing, People's Republic of China
| | - Jun-Li Li
- Biotherapy Center, General Hospital of Beijing Military Command, Beijing, People's Republic of China
| | - Jin-Long Liu
- Biotherapy Center, General Hospital of Beijing Military Command, Beijing, People's Republic of China
| | - Hai-Bo Wang
- Biotherapy Center, General Hospital of Beijing Military Command, Beijing, People's Republic of China
| | - Zheng-Xu Wang
- Biotherapy Center, General Hospital of Beijing Military Command, Beijing, People's Republic of China
| |
Collapse
|
50
|
Aris M, Bravo AI, Barrio MM, Mordoh J. Inoculation site from a cutaneous melanoma patient treated with an allogeneic therapeutic vaccine: a case report. Front Immunol 2015; 6:144. [PMID: 25870600 PMCID: PMC4378302 DOI: 10.3389/fimmu.2015.00144] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2014] [Accepted: 03/16/2015] [Indexed: 01/09/2023] Open
Abstract
We have developed a therapeutic vaccine consisting of a mixture of lethally-irradiated allogeneic cutaneous melanoma cell lines with BCG and GM-CSF as adjuvants. The CSF-470 vaccine is currently being assayed in a Phase II-III trial against medium-dose IFN-α2b. All vaccinated patients immunized intradermally developed large edematous erythema reactions, which then transformed into subcutaneous nodules active for several months. However, vaccine injection sites were not routinely biopsied. We describe the case of a female patient, previously classified as stage III, but who, due to the simultaneous discovery of bone metastases only received one vaccination was withdrawn from the study, and continued her treatment elsewhere. This patient developed a post-vaccination nodule which was surgically removed 7 weeks later, and allowed to analyze the reactivity and immune profiling of the inoculation site. An inflammatory reaction with zones of fibrosis, high irrigation, and brisk lymphoid infiltration, primarily composed of CD8(+) and CD20(+) lymphocytes, was observed. There were no remaining BCG bacilli, and scarce CD4(+) and Foxp3(+) T cells were determined. MART-1 Ag was found throughout the vaccination site. CD11c(+) Ag presenting cells were either dispersed or forming dense nests. Some CD11c(+) cells proliferated; most of them contained intracellular MART-1 Ag, and some interacted with CD8(+) lymphocytes. These observations suggest a potent, long-lasting local inflammatory response with recruitment of Ag-presenting cells that incorporate melanoma Ags, probably leading to Ag presentation to naïve T cells.
Collapse
Affiliation(s)
- Mariana Aris
- Centro de Investigaciones Oncológicas-Fundación Cáncer (CIO-FUCA) , Ciudad Autónoma de Buenos Aires , Argentina
| | - Alicia Inés Bravo
- Unidad de Inmunopatología, Hospital Interzonal General de Agudos Eva Perón , San Martín, Provincia de Buenos Aires , Argentina
| | - María Marcela Barrio
- Centro de Investigaciones Oncológicas-Fundación Cáncer (CIO-FUCA) , Ciudad Autónoma de Buenos Aires , Argentina
| | - José Mordoh
- Centro de Investigaciones Oncológicas-Fundación Cáncer (CIO-FUCA) , Ciudad Autónoma de Buenos Aires , Argentina ; Laboratorio de Cancerología, Fundación Instituto Leloir, IIBBA-CONICET , Ciudad Autónoma de Buenos Aires , Argentina ; Instituto Médico Especializado Alexander Fleming , Ciudad Autónoma de Buenos Aires , Argentina
| |
Collapse
|