1
|
Kumar KP, Madhusoodanan M, Pangath M, Menon D. Innovative landscapes in intraperitoneal therapy of ovarian cancer. Drug Deliv Transl Res 2025:10.1007/s13346-024-01765-w. [PMID: 39888579 DOI: 10.1007/s13346-024-01765-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/09/2024] [Indexed: 02/01/2025]
Abstract
Epithelial ovarian cancer is the most prevalent gynecological malignancy, characterized by high mortality rates due to its late-stage diagnosis and frequent recurrence. The current standard of care for ovarian cancer is a combination of debulking surgery followed by the conventional mode of chemotherapy. Despite significant advances in therapeutic modalities, the overall survival rate of EOC continues to be poor, mainly because low concentrations of the chemotherapeutics reach the peritoneum, which is the primary site of ovarian cancer, leading to disease relapse. Here, intraperitoneal chemotherapy gains advantage due to its ability to deliver the drug molecules directly to the peritoneal cavity and provide localized and sustained effects. This is facilitated by the use of diverse kinds of nano or micron sized delivery systems, which help in transporting drugs, vaccines, antibodies and genes appropriately to the peritoneum for its desired function. This review article delves on how intraperitoneal delivery impacts the therapy of epithelial ovarian cancer spanning the conventional therapeutic modes to the recent nanoinnovations in chemotherapy, immunotherapy and gene therapy.
Collapse
Affiliation(s)
- Krishna Pradeep Kumar
- Amrita School of Nanosciences & Molecular Medicine, Amrita Vishwa Vidyapeetham, Kochi, Kerala, India
| | - Maneesha Madhusoodanan
- Amrita School of Nanosciences & Molecular Medicine, Amrita Vishwa Vidyapeetham, Kochi, Kerala, India
| | - Meghna Pangath
- Amrita School of Nanosciences & Molecular Medicine, Amrita Vishwa Vidyapeetham, Kochi, Kerala, India
| | - Deepthy Menon
- Amrita School of Nanosciences & Molecular Medicine, Amrita Vishwa Vidyapeetham, Kochi, Kerala, India.
| |
Collapse
|
2
|
Balan D, Kampan NC, Plebanski M, Abd Aziz NH. Unlocking ovarian cancer heterogeneity: advancing immunotherapy through single-cell transcriptomics. Front Oncol 2024; 14:1388663. [PMID: 38873253 PMCID: PMC11169633 DOI: 10.3389/fonc.2024.1388663] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Accepted: 05/15/2024] [Indexed: 06/15/2024] Open
Abstract
Ovarian cancer, a highly fatal gynecological cancer, warrants the need for understanding its heterogeneity. The disease's prevalence and impact are underscored with statistics on mortality rates. Ovarian cancer is categorized into distinct morphological groups, each with its characteristics and prognosis. Despite standard treatments, survival rates remain low due to relapses and chemoresistance. Immune system involvement is evident in ovarian cancer's progression, although the tumor employs immune evasion mechanisms. Immunotherapy, particularly immune checkpoint blockade therapy, is promising, but ovarian cancer's heterogeneity limits its efficacy. Single-cell sequencing technology could be explored as a solution to dissect the heterogeneity within tumor-associated immune cell populations and tumor microenvironments. This cutting-edge technology has the potential to enhance diagnosis, prognosis, and personalized immunotherapy in ovarian cancer, reflecting its broader application in cancer research. The present review focuses on recent advancements and the challenges in applying single-cell transcriptomics to ovarian cancer.
Collapse
Affiliation(s)
- Dharvind Balan
- Department of Obstetrics and Gynaecology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Nirmala Chandralega Kampan
- Department of Obstetrics and Gynaecology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Magdalena Plebanski
- School of Health and Biomedical Sciences, RMIT University, Bundoora, VIC, Australia
| | - Nor Haslinda Abd Aziz
- Department of Obstetrics and Gynaecology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| |
Collapse
|
3
|
Wallis B, Bowman KR, Lu P, Lim CS. The Challenges and Prospects of p53-Based Therapies in Ovarian Cancer. Biomolecules 2023; 13:159. [PMID: 36671544 PMCID: PMC9855757 DOI: 10.3390/biom13010159] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 01/07/2023] [Accepted: 01/09/2023] [Indexed: 01/13/2023] Open
Abstract
It has been well established that mutations in the tumor suppressor gene, p53, occur readily in a vast majority of cancer tumors, including ovarian cancer. Typically diagnosed in stages three or four, ovarian cancer is the fifth leading cause of death in women, despite accounting for only 2.5% of all female malignancies. The overall 5-year survival rate for ovarian cancer is around 47%; however, this drops to an abysmal 29% for the most common type of ovarian cancer, high-grade serous ovarian carcinoma (HGSOC). HGSOC has upwards of 96% of cases expressing mutations in p53. Therefore, wild-type (WT) p53 and p53-based therapies have been explored as treatment options via a plethora of drug delivery vehicles including nanoparticles, viruses, polymers, and liposomes. However, previous p53 therapeutics have faced many challenges, which have resulted in their limited translational success to date. This review highlights a selection of these historical p53-targeted therapeutics for ovarian cancer, why they failed, and what the future could hold for a new generation of this class of therapies.
Collapse
Affiliation(s)
| | | | | | - Carol S. Lim
- Department of Molecular Pharmaceutics, College of Pharmacy, University of Utah, Salt Lake City, UT 84112, USA
| |
Collapse
|
4
|
Nanotechnology-Based Nucleic Acid Vaccines for Treatment of Ovarian Cancer. Pharm Res 2023; 40:123-144. [PMID: 36376606 PMCID: PMC9663189 DOI: 10.1007/s11095-022-03434-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Accepted: 11/03/2022] [Indexed: 11/16/2022]
Abstract
Anticancer vaccines represent a promising approach for effective treatment of cancer and along with recent advantages of nucleic acid-based vaccines for other diseases form a prospective and potentially efficacious direction of the research, development and clinical applications. Despite the ongoing several clinical trials of mRNA vaccines for the treatment of various types of cancer, to-date no cancer vaccines were approved by the US Food and Drug Administration. The present review analyzes and summarizes major approaches for treating of different forms of ovarian cancer including mRNA-based vaccines as well as nanotechnology-based approaches for their delivery.
Collapse
|
5
|
Chavda VP, Patel AB, Vora LK, Apostolopoulos V, Uhal BD. Dendritic cell-based vaccine: the state-of-the-art vaccine platform for COVID-19 management. Expert Rev Vaccines 2022; 21:1395-1403. [PMID: 35929957 DOI: 10.1080/14760584.2022.2110076] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION A correlation between new coronaviruses and host immunity, as well as the role of defective immune function in host response, would be extremely helpful in understanding coronavirus disease (COVID-19) pathogenicity, and a coherent structure of treatments and vaccines. As existing vaccines may be inadequate for new viral variants emerging in various regions of the world, it is a vital requirement for fresh and effective therapeutic alternatives. AREA COVERED Immunotherapy may give a viable protective option for COVID-19, a disease that is currently a big burden on global health and economic systems. Herein, we have outlined three dendritic cell (DC)-based vaccines for COVID-19 which are in human clinical trials and have shown encouraging outcomes. EXPERT OPINION With existing knowledge of the virus, and the nature of DC, DC-based vaccines may be proven to be effective in inducing long-lasting protective immunity, especially T cell responses.
Collapse
Affiliation(s)
- Vivek P Chavda
- Department of Pharmaceutics and Pharmaceutical Technology, L M College of Pharmacy, Ahmedabad - 380009, Gujarat, India
| | - Aayushi B Patel
- Pharmacy Section, LM College of Pharmacy, Ahmedabad - 380058, Gujarat, India
| | - Lalitkumar K Vora
- School of Pharmacy, Queen's University Belfast, 97 Lisburn Road, BT9 7BL, UK
| | - Vasso Apostolopoulos
- Institute for Health and Sport, Victoria University, Melbourne, VIC, 3030, Australia
| | - Bruce D Uhal
- Department of Physiology, Michigan State University, East Lansing, MI 48824, USA
| |
Collapse
|
6
|
Revythis A, Limbu A, Mikropoulos C, Ghose A, Sanchez E, Sheriff M, Boussios S. Recent Insights into PARP and Immuno-Checkpoint Inhibitors in Epithelial Ovarian Cancer. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:8577. [PMID: 35886427 PMCID: PMC9317199 DOI: 10.3390/ijerph19148577] [Citation(s) in RCA: 67] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 07/06/2022] [Accepted: 07/12/2022] [Indexed: 02/06/2023]
Abstract
Ovarian cancer is one of the most common gynecologic cancers and has the highest mortality rate of any other cancer of the female reproductive system. Epithelial ovarian cancer (EOC) accounts for approximately 90% of all ovarian malignancies. The standard therapeutic strategy includes cytoreductive surgery accompanied by pre- or postoperative platinum-based chemotherapy. Nevertheless, up to 80% of the patients relapse within the following 12-18 months from the completion of the treatment and then receive first-line chemotherapy depending on platinum sensitivity. Mutations in BRCA1/2 genes are the most significant molecular aberrations in EOC and serve as prognostic and predictive biomarkers. Poly ADP-ribose polymerase (PARP) inhibitors exploit defects in the DNA repair pathway through synthetic lethality. They have also been shown to trap PARP1 and PARP2 on DNA, leading to PARP-DNA complexes. Olaparib, rucaparib, and niraparib have all obtained Food and Drug Administration (FDA) and/or the European Medicine Agency (EMA) approval for the treatment of EOC in different settings. Immune checkpoint inhibitors (ICI) have improved the survival of several cancers and are under evaluation in EOC. However, despite the success of immunotherapy in other malignancies, the use of antibodies inhibiting the immune checkpoint programmed cell death (PD-1) or its ligand (PD-L1) obtained modest results in EOC so far, with median response rates of up to 10%. As such, ICI have not yet been approved for the treatment of EOC. We herein provided a comprehensive insight into the most recent progress in synthetic lethality PARP inhibitors, along with the mechanisms of resistance. We also summarised data regarding the role of immune checkpoint inhibitors, the use of vaccination therapy, and adoptive immunotherapy in treating epithelial ovarian cancer.
Collapse
Affiliation(s)
- Antonios Revythis
- Department of Medical Oncology, Medway NHS Foundation Trust, Windmill Road, Gillingham ME7 5NY, Kent, UK; (A.R.); (A.L.); (A.G.); (E.S.)
| | - Anu Limbu
- Department of Medical Oncology, Medway NHS Foundation Trust, Windmill Road, Gillingham ME7 5NY, Kent, UK; (A.R.); (A.L.); (A.G.); (E.S.)
| | - Christos Mikropoulos
- St. Lukes Cancer Centre, Royal Surrey County Hospital, Egerton Rd., Guildford GU2 7XX, Surrey, UK;
| | - Aruni Ghose
- Department of Medical Oncology, Medway NHS Foundation Trust, Windmill Road, Gillingham ME7 5NY, Kent, UK; (A.R.); (A.L.); (A.G.); (E.S.)
- Department of Medical Oncology, Barts Cancer Centre, St. Bartholomew’s Hospital, Barts Health NHS Trust, London KT1 2EE, UK
- Department of Medical Oncology, Mount Vernon Cancer Centre, East and North Hertfordshire NHS Trust, London KT1 2EE, UK
- Centre for Education, Faculty of Life Sciences and Medicine, King’s College London, London SE5 9NU, UK
| | - Elisabet Sanchez
- Department of Medical Oncology, Medway NHS Foundation Trust, Windmill Road, Gillingham ME7 5NY, Kent, UK; (A.R.); (A.L.); (A.G.); (E.S.)
| | - Matin Sheriff
- Department of Urology, Medway NHS Foundation Trust, Windmill Road, Gillingham ME7 5NY, Kent, UK;
| | - Stergios Boussios
- Department of Medical Oncology, Medway NHS Foundation Trust, Windmill Road, Gillingham ME7 5NY, Kent, UK; (A.R.); (A.L.); (A.G.); (E.S.)
- Faculty of Life Sciences & Medicine, School of Cancer & Pharmaceutical Sciences, King’s College London, London SE1 9RT, UK
- AELIA Organization, 9th Km Thessaloniki—Thermi, 57001 Thessaloniki, Greece
| |
Collapse
|
7
|
Chilimoniuk Z, Rocka A, Stefaniak M, Tomczyk Ż, Jasielska F, Madras D, Filip A. Molecular methods for increasing the effectiveness of ovarian cancer treatment: a systematic review. Future Oncol 2022; 18:1627-1650. [PMID: 35129396 DOI: 10.2217/fon-2021-0565] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Background: The aim of the current study is to analyze and summarize the latest research on improving therapy in ovarian cancer. Materials & methods: Data analysis was based on a review of publications from 2011 to 2021 in the PubMed database with use of the search terms including 'EGFR ovarian cancer', 'folate receptor inhibitors ovarian cancer', 'VEGF ovarian cancer', 'PDGF ovarian cancer' and 'CTLA-4 ovarian cancer'. Results: 6643 articles were found; 238 clinical trials and randomized control trials were analyzed; 122 studies were rejected due to inconsistency with the topic of the work. Conclusion: Extensive research on the treatment of ovarian cancer increases the chance of developing the most effective therapy suited to the individual needs of the patient.
Collapse
Affiliation(s)
- Zuzanna Chilimoniuk
- Students' Scientific Association at the Department of Cancer Genetics with Cytogenetics Laboratory, Medical University of Lublin, ul. Radziwiłłowska 11, Lublin, 20-080, Poland
| | - Agata Rocka
- Students' Scientific Association at the Department of Cancer Genetics with Cytogenetics Laboratory, Medical University of Lublin, ul. Radziwiłłowska 11, Lublin, 20-080, Poland
| | - Martyna Stefaniak
- Students' Scientific Association at the Department of Cancer Genetics with Cytogenetics Laboratory, Medical University of Lublin, ul. Radziwiłłowska 11, Lublin, 20-080, Poland
| | - Żaklina Tomczyk
- Students' Scientific Association at the Department of Cancer Genetics with Cytogenetics Laboratory, Medical University of Lublin, ul. Radziwiłłowska 11, Lublin, 20-080, Poland
| | - Faustyna Jasielska
- Students' Scientific Association at the Department of Cancer Genetics with Cytogenetics Laboratory, Medical University of Lublin, ul. Radziwiłłowska 11, Lublin, 20-080, Poland
| | - Dominika Madras
- Students' Scientific Association at the Department of Cancer Genetics with Cytogenetics Laboratory, Medical University of Lublin, ul. Radziwiłłowska 11, Lublin, 20-080, Poland
| | - Agata Filip
- Department of Cancer Genetics with Cytogenetics Laboratory, Medical University of Lublin, ul. Radziwiłłowska 11, Lublin, 20-080, Poland
| |
Collapse
|
8
|
Bund V, Azaïs H, Bibi-Triki S, Lecointre L, Betrian SB, Angeles MA, Eberst L, Faller E, Boisramé T, Bendifallah S, Akladios C, Deluche É. Basics of immunotherapy for epithelial ovarian cancer. J Gynecol Obstet Hum Reprod 2021; 51:102283. [PMID: 34875397 DOI: 10.1016/j.jogoh.2021.102283] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Accepted: 12/02/2021] [Indexed: 10/19/2022]
Abstract
Epithelial ovarian cancer (EOC) is the most lethal of all gynecological cancers. Despite excellent responses to standard treatment in approximately 70% of patients, most of them will relapse within 5 years of initial treatment and many of them will develop chemotherapy-resistant disease. It is then important to find other means of treatment for these patients such as immunotherapy or targeted therapy. To understand immunotherapy, it is important to explain the dynamic interplay between cancer and the immune system. Compared to traditional tumor therapies, immunotherapy acts primarily on the immune system or the tumor microenvironment but not directly on the tumor cells, and it may also promote synergistic anti-tumor actions as part of a combined treatment. The aim of this narrative review is to provide a basic understanding of immunotherapy the interest of this treatment in EOC, and to present the main ongoing studies that could change patient management in the future.
Collapse
Affiliation(s)
- Virginie Bund
- Department of Gynecologic Surgery, Hôpitaux Universitaires de Strasbourg, 67000 Strasbourg, France; Laboratoire d'ImmunoRhumatologie Moléculaire, Institut national de la santé et de la recherche médicale (INSERM) UMR_S 1109, Institut thématique interdisciplinaire (ITI) de Médecine de Précision de Strasbourg, Transplantex NG, Faculté de Médecine, Fédération Hospitalo-Universitaire OMICARE, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Université de Strasbourg, Strasbourg, France.
| | - Henri Azaïs
- Department of Gynecologic and Breast Oncological Surgery, Georges-Pompidou European Hospital, APHP. Centre, France.
| | - Sabrina Bibi-Triki
- Laboratoire d'ImmunoRhumatologie Moléculaire, Institut national de la santé et de la recherche médicale (INSERM) UMR_S 1109, Institut thématique interdisciplinaire (ITI) de Médecine de Précision de Strasbourg, Transplantex NG, Faculté de Médecine, Fédération Hospitalo-Universitaire OMICARE, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Université de Strasbourg, Strasbourg, France.
| | - Lise Lecointre
- Department of Gynecologic Surgery, Hôpitaux Universitaires de Strasbourg, 67000 Strasbourg, France; IHU-Strasbourg (Institut Hospitalo-Universitaire), Strasbourg, France.
| | - Sarah Bétrian Betrian
- Medical oncology Department, Institut Claudius Regaud, Institut Universitaire du Cancer, Toulouse, France.
| | - Martina Aida Angeles
- Department of Gynecologic and Breast Oncological Surgery, European Georges-Pompidou Hospital, APHP. Centre, France.
| | - Lauriane Eberst
- Department of Oncology, Institut de Cancérologie de Strasbourg (ICANS), Strasbourg, France.
| | - Emilie Faller
- Department of Gynecologic Surgery, Hôpitaux Universitaires de Strasbourg, 67000 Strasbourg, France
| | - Thomas Boisramé
- Department of Gynecologic Surgery, Hôpitaux Universitaires de Strasbourg, 67000 Strasbourg, France
| | | | - Chérif Akladios
- Department of Gynecologic Surgery, Hôpitaux Universitaires de Strasbourg, 67000 Strasbourg, France; I.R.C.A.D - Institut de Recherche contre les Cancers de l'Appareil Digestif. 67000 Strasbourg, France.
| | - Élise Deluche
- Medical oncology Department, Limoges University Hospital, France.
| | | |
Collapse
|
9
|
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
|
10
|
Sarivalasis A, Morotti M, Mulvey A, Imbimbo M, Coukos G. Cell therapies in ovarian cancer. Ther Adv Med Oncol 2021; 13:17588359211008399. [PMID: 33995591 PMCID: PMC8072818 DOI: 10.1177/17588359211008399] [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: 12/03/2020] [Accepted: 03/17/2021] [Indexed: 12/15/2022] Open
Abstract
Epithelial ovarian cancer (EOC) is the most important cause of gynecological cancer-related mortality. Despite improvements in medical therapies, particularly with the incorporation of drugs targeting homologous recombination deficiency, EOC survival rates remain low. Adoptive cell therapy (ACT) is a personalized form of immunotherapy in which autologous lymphocytes are expanded, manipulated ex vivo, and re-infused into patients to mediate cancer rejection. This highly promising novel approach with curative potential encompasses multiple strategies, including the adoptive transfer of tumor-infiltrating lymphocytes, natural killer cells, or engineered immune components such as chimeric antigen receptor (CAR) constructs and engineered T-cell receptors. Technical advances in genomics and immuno-engineering have made possible neoantigen-based ACT strategies, as well as CAR-T cells with increased cell persistence and intratumoral trafficking, which have the potential to broaden the opportunity for patients with EOC. Furthermore, dendritic cell-based immunotherapies have been tested in patients with EOC with modest but encouraging results, while the combination of DC-based vaccination as a priming modality for other cancer therapies has shown encouraging results. In this manuscript, we provide a clinically oriented historical overview of various forms of cell therapies for the treatment of EOC, with an emphasis on T-cell therapy.
Collapse
Affiliation(s)
- Apostolos Sarivalasis
- Department of Oncology, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland
| | - Matteo Morotti
- Department of Oncology, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland
| | - Arthur Mulvey
- Department of Oncology, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland
| | - Martina Imbimbo
- Department of Oncology, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland
| | - George Coukos
- CHUV, Rue du Bugnon 46, Lausanne BH09-701, Switzerland
| |
Collapse
|
11
|
McGray AJR, Eppolito C, Miliotto A, Singel KL, Stephenson K, Lugade A, Segal BH, Keler T, Webster G, Lichty B, Kozbor D, Odunsi K. A prime/boost vaccine platform efficiently identifies CD27 agonism and depletion of myeloid-derived suppressor cells as therapies that rationally combine with checkpoint blockade in ovarian cancer. Cancer Immunol Immunother 2021; 70:3451-3460. [PMID: 33880648 PMCID: PMC8057655 DOI: 10.1007/s00262-021-02936-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Accepted: 03/31/2021] [Indexed: 12/30/2022]
Abstract
Cancer immunotherapies have generated remarkable clinical responses for some patients with advanced/metastatic disease, prompting exploration of rational combination therapies to bolster anti-tumor immunity in patients with limited response or those who experience tumor progression following an initial response to immunotherapy. In contrast to other tumor indications, objective response rates to single-agent PD-1/PD-L1 blockade in ovarian cancer are limited, suggesting a need to identify combinatorial approaches that lead to tumor regression in a setting where checkpoint blockade alone is ineffective. Using a pre-clinical model of aggressive intraperitoneal ovarian cancer, we have previously reported on a heterologous prime/boost cancer vaccine that elicits robust anti-tumor immunity, prolongs survival of tumor-bearing mice, and which is further improved when combined with checkpoint blockade. As tumor control in this model is CD8 + T cell dependent, we reasoned that the prime/boost vaccine platform could be used to explore additional treatment combinations intended to bolster the effects of CD8 + T cells. Using whole tumor transcriptomic data, we identified candidate therapeutic targets anticipated to rationally combine with prime/boost vaccination. In the context of a highly effective cancer vaccine, CD27 agonism or antibody-mediated depletion of granulocytic cells each modestly increased tumor control following vaccination, with anti-PD-1 therapy further improving treatment efficacy. These findings support the use of immunotherapies with well-defined mechanisms(s) of action as a valuable platform for identifying candidate combination approaches for further therapeutic testing in ovarian cancer.
Collapse
Affiliation(s)
- A J R McGray
- Center for Immunotherapy, Roswell Park Comprehensive Cancer Center, Roswell Park Cancer Institute, Elm and Carlton Sts, Buffalo, NY, 14263, USA.
| | - C Eppolito
- Center for Immunotherapy, Roswell Park Comprehensive Cancer Center, Roswell Park Cancer Institute, Elm and Carlton Sts, Buffalo, NY, 14263, USA
| | - A Miliotto
- Center for Immunotherapy, Roswell Park Comprehensive Cancer Center, Roswell Park Cancer Institute, Elm and Carlton Sts, Buffalo, NY, 14263, USA
| | - K L Singel
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
- Office of Evaluation, Performance, and Reporting, National Institutes of Health, Bethesda, MD, USA
| | - K Stephenson
- McMaster Immunology Research Centre, McMaster University, Hamilton, ON, Canada
- Turnstone Biologics, Ottawa, ON, Canada
| | - A Lugade
- Center for Immunotherapy, Roswell Park Comprehensive Cancer Center, Roswell Park Cancer Institute, Elm and Carlton Sts, Buffalo, NY, 14263, USA
| | - B H Segal
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
- Department of Internal Medicine, Roswell Park Comprehensive Cancer Center and Jacobs School of Medicine and Biomedical Sciences, University At Buffalo, Buffalo, NY, USA
| | - T Keler
- Celldex Therapeutics, Hampton, NJ, USA
| | - G Webster
- Innate Immunotherapeutics, Auckland, New Zealand
| | - B Lichty
- McMaster Immunology Research Centre, McMaster University, Hamilton, ON, Canada
| | - D Kozbor
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - K Odunsi
- Center for Immunotherapy, Roswell Park Comprehensive Cancer Center, Roswell Park Cancer Institute, Elm and Carlton Sts, Buffalo, NY, 14263, USA.
- University of Chicago Comprehensive Cancer Center, 5841 South Maryland Avenue, MC1140, Chicago, IL, 60637, USA.
| |
Collapse
|
12
|
Shi Y, Tomczak K, Li J, Ochieng JK, Lee Y, Haymaker C. Next-Generation Immunotherapies to Improve Anticancer Immunity. Front Pharmacol 2021; 11:566401. [PMID: 33505304 PMCID: PMC7831045 DOI: 10.3389/fphar.2020.566401] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Accepted: 10/28/2020] [Indexed: 12/15/2022] Open
Abstract
Checkpoint inhibitors are widely used immunotherapies for advanced cancer. Nonetheless, checkpoint inhibitors have a relatively low response rate, work in a limited range of cancers, and have some unignorable side effects. Checkpoint inhibitors aim to reinvigorate exhausted or suppressed T cells in the tumor microenvironment (TME). However, the TME contains various other immune cell subsets that interact to determine the fate of cytotoxic T cells. Activation of cytotoxic T cells is initiated by antigen cross-presentation of dendritic cells. Dendritic cells could also release chemokines and cytokines to recruit and foster T cells. B cells, another type of antigen-presenting cell, also foster T cells and can produce tumor-specific antibodies. Neutrophils, a granulocyte cell subset in the TME, impede the proliferation and activation of T cells. The TME also consists of cytotoxic innate natural killer cells, which kill tumor cells efficiently. Natural killer cells can eradicate major histocompatibility complex I-negative tumor cells, which escape cytotoxic T cell–mediated destruction. A thorough understanding of the immune mechanism of the TME, as reviewed here, will lead to further development of more powerful therapeutic strategies. We have also reviewed the clinical outcomes of patients treated with drugs targeting these immune cells to identify strategies for improvement and possible immunotherapy combinations.
Collapse
Affiliation(s)
- Yaoyao Shi
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Katarzyna Tomczak
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - June Li
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Joshua K Ochieng
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Younghee Lee
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Cara Haymaker
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| |
Collapse
|
13
|
Kandalaft LE, Odunsi K, Coukos G. Immune Therapy Opportunities in Ovarian Cancer. Am Soc Clin Oncol Educ Book 2021; 40:1-13. [PMID: 32412818 DOI: 10.1200/edbk_280539] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Immunotherapy has emerged as a highly promising approach in the treatment of epithelial ovarian cancer (EOC). Immune checkpoint blockade (ICB) therapy, PARP inhibitors (PARPis), neoantigen vaccines, and personalized T-cell therapy have been associated with encouraging clinical activity in a small subset of patients. To increase the proportion of patients who are likely to derive benefit, it will be important not only to generate sufficient numbers of antitumor T cells but also to overcome multiple inhibitory networks in the ovarian tumor microenvironment (TME). Therefore, a major direction is to develop biomarkers that would predict responsiveness to different types of immunotherapies and allow treatment selection based on the results. Moreover, such biomarkers would allow rational combination of immunotherapies while minimizing toxicities. In this review, we provide progress on immune therapies and future directions for maximally exploiting immune-based strategies for the treatment of ovarian cancer.
Collapse
Affiliation(s)
- Lana E Kandalaft
- Ludwig Institute for Cancer Research, University of Lausanne, and Department of Oncology, Lausanne University Hospital, Lausanne, Switzerland
| | - Kunle Odunsi
- Center for Immunotherapy, Roswell Park Comprehensive Cancer Center, Buffalo, NY.,Department of Gynecologic Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY
| | - George Coukos
- Ludwig Institute for Cancer Research, University of Lausanne, and Department of Oncology, Lausanne University Hospital, Lausanne, Switzerland
| |
Collapse
|
14
|
Hartnett EG, Knight J, Radolec M, Buckanovich RJ, Edwards RP, Vlad AM. Immunotherapy Advances for Epithelial Ovarian Cancer. Cancers (Basel) 2020; 12:cancers12123733. [PMID: 33322601 PMCID: PMC7764119 DOI: 10.3390/cancers12123733] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Revised: 12/01/2020] [Accepted: 12/07/2020] [Indexed: 12/23/2022] Open
Abstract
Simple Summary The overall five-year survival rate in epithelial ovarian cancer is 44% and has only marginally improved in the past two decades. Despite an initial response to standard treatment consisting of chemotherapy and surgical removal of tumor, the lesions invariably recur, and patients ultimately die of chemotherapy resistant disease. New treatment modalities are needed in order to improve the prognosis of women diagnosed with ovarian cancer. One such modality is immunotherapy, which aims to boost the capacity of the patient’s immune system to recognize and attack the tumor cells. We performed a retrospective study to identify some of the most promising immune therapies for epithelial ovarian cancer. Special emphasis was given to immuno-oncology clinical trials. Abstract New treatment modalities are needed in order to improve the prognosis of women diagnosed with epithelial ovarian cancer (EOC), the most aggressive gynecologic cancer type. Most ovarian tumors are infiltrated by immune effector cells, providing the rationale for targeted approaches that boost the existing or trigger new anti-tumor immune mechanisms. The field of immuno-oncology has experienced remarkable progress in recent years, although the results seen with single agent immunotherapies in several categories of solid tumors have yet to extend to ovarian cancer. The challenge remains to determine what treatment combinations are most suitable for this disease and which patients are likely to benefit and to identify how immunotherapy should be incorporated into EOC standard of care. We review here some of the most promising immune therapies for EOC and focus on those currently tested in clinical trials.
Collapse
Affiliation(s)
- Erin G. Hartnett
- Department of Obstetrics and Gynecology and Reproductive Sciences, Magee-Womens Research Institute and Foundation and Magee-Womens Hospital of UPMC, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA; (E.G.H.); (M.R.); (R.J.B.); (R.P.E.)
| | - Julia Knight
- School of Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA;
| | - Mackenzy Radolec
- Department of Obstetrics and Gynecology and Reproductive Sciences, Magee-Womens Research Institute and Foundation and Magee-Womens Hospital of UPMC, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA; (E.G.H.); (M.R.); (R.J.B.); (R.P.E.)
| | - Ronald J. Buckanovich
- Department of Obstetrics and Gynecology and Reproductive Sciences, Magee-Womens Research Institute and Foundation and Magee-Womens Hospital of UPMC, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA; (E.G.H.); (M.R.); (R.J.B.); (R.P.E.)
| | - Robert P. Edwards
- Department of Obstetrics and Gynecology and Reproductive Sciences, Magee-Womens Research Institute and Foundation and Magee-Womens Hospital of UPMC, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA; (E.G.H.); (M.R.); (R.J.B.); (R.P.E.)
| | - Anda M. Vlad
- Department of Obstetrics and Gynecology and Reproductive Sciences, Magee-Womens Research Institute and Foundation and Magee-Womens Hospital of UPMC, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA; (E.G.H.); (M.R.); (R.J.B.); (R.P.E.)
- Correspondence:
| |
Collapse
|
15
|
García-Martínez E, Pérez-Fidalgo JA. Immunotherapies in ovarian cancer. EJC Suppl 2020; 15:87-95. [PMID: 33240447 PMCID: PMC7573463 DOI: 10.1016/j.ejcsup.2020.02.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2019] [Revised: 02/18/2020] [Accepted: 02/29/2020] [Indexed: 01/21/2023] Open
Abstract
Ovarian cancer is the leading cause of death for gynaecological cancer, and new therapies are urgently awaited. Although the presence of tumour-infiltrating lymphocytes has been confirmed to be associated to a better prognosis, immunotherapy is not yet incorporated to the armamentarium in ovarian cancer. This review briefly summarises the strategies that have been tested or are under study for the three different groups of tumours: immune desert, inflamed and immune-excluded ovarian tumours. Finally, a better knowledge of the biology and immune microenvironment is needed for successfully developing new immunotherapy strategies. Immune ovarian cancer subtypes could improve the selection patients for immunotherapy. Very frequently ovarian cancer needs to be converted in an inflamed tumour. Checkpoints inhibitor combinations are well designed and very promising in ovarian cancer.
Collapse
Affiliation(s)
- Elena García-Martínez
- Department of Medical Oncology, Hospital Universitario Morales Meseguer, Instituto Murciano de Investigación Biosanitaria (IMIB), Grupo Español de Investigación en Cáncer de Ovario (GEICO), Murcia, Spain
| | - J Alejandro Pérez-Fidalgo
- Department of Medical Oncology, Hospital Clínico Universitario de Valencia, Instituto de Investigación Sanitaria INCLIVA, Grupo Español de Investigación en Cáncer de Ovario (GEICO), Valencia, Spain
| |
Collapse
|
16
|
Dafni U, Martín-Lluesma S, Balint K, Tsourti Z, Vervita K, Chenal J, Coukos G, Zaman K, Sarivalasis A, Kandalaft LE. Efficacy of cancer vaccines in selected gynaecological breast and ovarian cancers: A 20-year systematic review and meta-analysis. Eur J Cancer 2020; 142:63-82. [PMID: 33221598 DOI: 10.1016/j.ejca.2020.10.014] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 09/29/2020] [Accepted: 10/08/2020] [Indexed: 12/30/2022]
Abstract
BACKGROUND Therapeutic cancer vaccination is an area of interest, even though promising efficacy has not been demonstrated so far. DESIGN A systematic review and meta-analysis was conducted to evaluate vaccines' efficacy on breast cancer (BC) and ovarian cancer (OC) patients. Our search was based on the PubMed electronic database, from 1st January 2000 to 4th February 2020. OBJECTIVE response rate (ORR) was the primary end-point of interest, while progression-free survival (PFS), overall survival (OS) and toxicity were secondary end-points. Analysis was performed separately for BC and OC patients. Pooled ORRs were estimated by fixed or random effects models, depending on the detected degree of heterogeneity, for all studies with more than five patients. Subgroup analyses by vaccine type and treatment schema as well as sensitivity analyses, were implemented. RESULTS Among 315 articles initially identified, 67 were eligible for our meta-analysis (BC: 46, 1698 patients; OC: 32, 426 patients; where both BC/OC in 11). Dendritic-cell and peptide vaccines were found in more studies, 6/10 BC and 10/13 OC studies, respectively. In our primary BC analysis (21 studies; 428 patients), the pooled ORR estimate was 9% (95%CI[5%,13%]). The primary OC analysis (12 studies; 182 patients), yielded pooled ORR estimate of 4% (95%CI[1%,7%]). Similar were the results derived in sensitivity analyses. No statistically significant differences were detected by vaccine type or treatment schema. Median PFS was 2.6 months (95% confidence interval (CI)[1.9,2.9]) and 13.0 months (95%CI[8.5,16.3]) for BC and OC respectively, while corresponding median OS was 24.8 months (95%CI[15.0,46.0]) and 39.0 months (95%CI[31.0,49.0]). In almost all cases, the observed toxicity was only moderate. CONCLUSION Despite their modest results in terms of ORR, therapeutic vaccines in the last 20 years display relatively long survival rates and low toxicity. Since a plethora of different approaches have been tested, a better understanding of the underlying mechanisms is needed in order to further improve vaccine efficacy.
Collapse
Affiliation(s)
- U Dafni
- Department of Oncology, CHUV, University of Lausanne, Lausanne, Switzerland; Faculty of Nursing, National and Kapodistrian University of Athens, Athens, Greece
| | - S Martín-Lluesma
- Departamento de Ciencias Médicas Básicas, Facultad de Medicina, Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, Boadilla Del Monte, Madrid, 28668, Spain
| | - K Balint
- Department of Oncology, CHUV, University of Lausanne, Lausanne, Switzerland
| | - Z Tsourti
- Scientific Research Consulting Hellas, Statistics Center, Athens, Greece
| | - K Vervita
- Scientific Research Consulting Hellas, Statistics Center, Athens, Greece
| | - J Chenal
- Department of Oncology, CHUV, University of Lausanne, Lausanne, Switzerland
| | - G Coukos
- Department of Oncology, CHUV, University of Lausanne, Lausanne, Switzerland; Ludwig Institute for Cancer Research, University of Lausanne, Lausanne, Switzerland
| | - K Zaman
- Department of Oncology, CHUV, University of Lausanne, Lausanne, Switzerland
| | - A Sarivalasis
- Department of Oncology, CHUV, University of Lausanne, Lausanne, Switzerland
| | - L E Kandalaft
- Department of Oncology, CHUV, University of Lausanne, Lausanne, Switzerland; Ludwig Institute for Cancer Research, University of Lausanne, Lausanne, Switzerland.
| |
Collapse
|
17
|
Salewski I, Gladbach YS, Kuntoff S, Irmscher N, Hahn O, Junghanss C, Maletzki C. In vivo vaccination with cell line-derived whole tumor lysates: neoantigen quality, not quantity matters. J Transl Med 2020; 18:402. [PMID: 33087163 PMCID: PMC7579816 DOI: 10.1186/s12967-020-02570-y] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Accepted: 10/12/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Cancer vaccines provide a complex source of neoantigens. Still, increasing evidence reveals that the neoantigen quality rather than the quantity is predictive for treatment outcome. METHODS Using the preclinical Mlh1-/- tumor model, we performed a side-by side comparison of two autologous cell-line derived tumor lysates (namely 328 and A7450 T1 M1) harboring different tumor mutational burden (TMB; i.e. ultra-high: 328; moderate-high: A7450 T1 M1). Mice received repetitive prophylactic or therapeutic applications of the vaccine. Tumor incidence, immune responses and tumor microenvironment was examined. RESULTS Both tumor cell lysates delayed tumor formation in the prophylactic setting, with the A7450 T1 M1 lysate being more effective in decelerating tumor growth than the 328 lysate (median overall survival: 37 vs. 25 weeks). Comparable results were achieved in therapeutic setting and could be traced back to antigen-driven immune stimulation. Reactive T cells isolated from A7450 T1 M1-treated mice recognized autologous Mlh1-/- tumor cells in IFNγ ELISpot, but likewise YAC-1 cells, indicative for stimulation of both arms of the immune system. By deciphering local effects, vaccines shaped the tumor microenvironment differently. While A7450 T1 M1 prophylactically vaccinated tumors harbored low numbers of myeloid-derived suppressor cells (MDSC) and elevated CD8-T cell infiltrates, vaccination with the 328 lysate evoked MDSC infiltration. Similar effects were seen in the therapeutic setting with stable disease induction only upon A7450 T1 M1 vaccination. Untangling individual response profiles revealed strong infiltration with LAG3+ and PD-L1+ immune cells when treatments failed, but almost complete exclusion of checkpoint-expressing lymphocytes in long-term survivors. CONCLUSIONS By applying two tumor cell lysates we demonstrate that neoantigen quality outranks quantity. This should be considered prior to designing cancer vaccine-based combination approaches.
Collapse
Affiliation(s)
- Inken Salewski
- Department of Medicine, Clinic III-Hematology, Oncology, Palliative Care, Rostock University Medical Center, Ernst-Heydemann-Str. 6, 18057, Rostock, Germany
| | - Yvonne Saara Gladbach
- Institute for Biostatistics and Informatics in Medicine and Ageing Research (IBIMA), Rostock University Medical Center, University of Rostock, 18057, Rostock, Germany.,Faculty of Biosciences, Heidelberg University, 69120, Heidelberg, Germany.,Division of Applied Bioinformatics, German Cancer Research Center (DKFZ) and National Center for Tumor Diseases (NCT), Heidelberg, Germany
| | - Steffen Kuntoff
- Department of Medicine, Clinic III-Hematology, Oncology, Palliative Care, Rostock University Medical Center, Ernst-Heydemann-Str. 6, 18057, Rostock, Germany
| | - Nina Irmscher
- Department of Medicine, Clinic III-Hematology, Oncology, Palliative Care, Rostock University Medical Center, Ernst-Heydemann-Str. 6, 18057, Rostock, Germany
| | - Olga Hahn
- Department of Cell Biology, Rostock University Medical Center, 18057, Rostock, Germany
| | - Christian Junghanss
- Department of Medicine, Clinic III-Hematology, Oncology, Palliative Care, Rostock University Medical Center, Ernst-Heydemann-Str. 6, 18057, Rostock, Germany
| | - Claudia Maletzki
- Department of Medicine, Clinic III-Hematology, Oncology, Palliative Care, Rostock University Medical Center, Ernst-Heydemann-Str. 6, 18057, Rostock, Germany.
| |
Collapse
|
18
|
Yang C, Xia BR, Zhang ZC, Zhang YJ, Lou G, Jin WL. Immunotherapy for Ovarian Cancer: Adjuvant, Combination, and Neoadjuvant. Front Immunol 2020; 11:577869. [PMID: 33123161 PMCID: PMC7572849 DOI: 10.3389/fimmu.2020.577869] [Citation(s) in RCA: 196] [Impact Index Per Article: 39.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Accepted: 09/16/2020] [Indexed: 12/13/2022] Open
Abstract
Ovarian cancer is the most lethal gynecologic malignancy. Surgery and chemotherapy are the primary treatments for ovarian cancer; however, patients often succumb to recurrence with chemotherapeutic resistance within several years after the initial treatment. In the past two decades, immunotherapy has rapidly developed, and has revolutionized the treatment of various types of cancer. Despite the fact that immunotherapy response rates among ovarian cancer patients remain modest, treatment with immune checkpoint inhibitors (ICIs), chimeric antigen receptor (CAR)- and TCR-engineered T cells is rapidly developing. Therapeutic efficiency could be improved significantly if immunotherapy is included as an adjuvant therapy, in combination with chemotherapy, radiation therapy, and the use of anti-angiogenesis drugs, and poly ADP ribose polymerase inhibitors (PARPi). Newly developed technologies that identify therapeutic targets, predict treatment efficacy, rapidly screen potential immunotherapy drugs, provide neoadjuvant immunotherapy, and utilize nanomedicine technology provide new opportunities for the treatment of ovarian cancer, and have the potential to prolong patient survival. However, important issues that may hinder the efficacy of such approaches, including hyperprogressive disease (HPD), immunotherapy-resistance, and toxicity of the treatments, including neurotoxicity, must be taken into account and addressed for these therapies to be effective.
Collapse
Affiliation(s)
- Chang Yang
- Department of Gynecology Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Bai-Rong Xia
- Department of Gynecology Oncology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Zhao-Cong Zhang
- Department of Gynecology Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Yong-Jian Zhang
- Department of Gynecology Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Ge Lou
- Department of Gynecology Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Wei-Lin Jin
- Department of Instrument Science and Engineering, Institute of Nano Biomedicine and Engineering, Shanghai Engineering Center for Intelligent Diagnosis and Treatment Instrument, Key Laboratory for Thin Film and Microfabrication Technology of Ministry of Education, School of Electronic Information and Electronic Engineering, Shanghai Jiao Tong University, Shanghai, China
- National Center for Translational Medicine, Collaborative Innovational Center for System Biology, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
19
|
Macpherson AM, Barry SC, Ricciardelli C, Oehler MK. Epithelial Ovarian Cancer and the Immune System: Biology, Interactions, Challenges and Potential Advances for Immunotherapy. J Clin Med 2020; 9:E2967. [PMID: 32937961 PMCID: PMC7564553 DOI: 10.3390/jcm9092967] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 08/28/2020] [Accepted: 09/03/2020] [Indexed: 12/11/2022] Open
Abstract
Recent advances in the understanding of immune function and the interactions with tumour cells have led to the development of various cancer immunotherapies and strategies for specific cancer types. However, despite some stunning successes with some malignancies such as melanomas and lung cancer, most patients receive little or no benefit from immunotherapy, which has been attributed to the tumour microenvironment and immune evasion. Although the US Food and Drug Administration have approved immunotherapies for some cancers, to date, only the anti-angiogenic antibody bevacizumab is approved for the treatment of epithelial ovarian cancer. Immunotherapeutic strategies for ovarian cancer are still under development and being tested in numerous clinical trials. A detailed understanding of the interactions between cancer and the immune system is vital for optimisation of immunotherapies either alone or when combined with chemotherapy and other therapies. This article, in two main parts, provides an overview of: (1) components of the normal immune system and current knowledge regarding tumour immunology, biology and their interactions; (2) strategies, and targets, together with challenges and potential innovative approaches for cancer immunotherapy, with attention given to epithelial ovarian cancer.
Collapse
Affiliation(s)
- Anne M. Macpherson
- Discipline of Obstetrics and Gynaecology, Adelaide Medical School, Robinson Research Institute, University of Adelaide, Adelaide 5000, Australia; (A.M.M.); (C.R.)
| | - Simon C. Barry
- Molecular Immunology, Robinson Research Institute, University of Adelaide, Adelaide 5005, Australia;
| | - Carmela Ricciardelli
- Discipline of Obstetrics and Gynaecology, Adelaide Medical School, Robinson Research Institute, University of Adelaide, Adelaide 5000, Australia; (A.M.M.); (C.R.)
| | - Martin K. Oehler
- Discipline of Obstetrics and Gynaecology, Adelaide Medical School, Robinson Research Institute, University of Adelaide, Adelaide 5000, Australia; (A.M.M.); (C.R.)
- Department of Gynaecological Oncology, Royal Adelaide Hospital, Adelaide 5000, Australia
| |
Collapse
|
20
|
Ahmed R, Crespo I, Tuyaerts S, Bekkar A, Graciotti M, Xenarios I, Kandalaft LE. Predicting combinations of immunomodulators to enhance dendritic cell-based vaccination based on a hybrid experimental and computational platform. Comput Struct Biotechnol J 2020; 18:2217-2227. [PMID: 32952936 PMCID: PMC7475195 DOI: 10.1016/j.csbj.2020.08.001] [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: 05/13/2020] [Revised: 07/28/2020] [Accepted: 08/01/2020] [Indexed: 12/24/2022] Open
Abstract
Dendritic cell (DC)-based vaccines have been largely used in the adjuvant setting for the treatment of cancer, however, despite their proven safety, clinical outcomes still remain modest. In order to improve their efficacy, DC-based vaccines are often combined with one or multiple immunomodulatory agents. However, the selection of the most promising combinations is hampered by the plethora of agents available and the unknown interplay between these different agents. To address this point, we developed a hybrid experimental and computational platform to predict the effects and immunogenicity of dual combinations of stimuli once combined with DC vaccination, based on the experimental data of a variety of assays to monitor different aspects of the immune response after a single stimulus. To assess the stimuli behavior when used as single agents, we first developed an in vitro co-culture system of T cell priming using monocyte-derived DCs loaded with whole tumor lysate to prime autologous peripheral blood mononuclear cells in the presence of the chosen stimuli, as single adjuvants, and characterized the elicited response assessing 18 different phenotypic and functional traits important for an efficient anti-cancer response. We then developed and applied a prediction algorithm, generating a ranking for all possible dual combinations of the different single stimuli considered here. The ranking generated by the prediction tool was then validated with experimental data showing a strong correlation with the predicted scores, confirming that the top ranked conditions globally significantly outperformed the worst conditions. Thus, the method developed here constitutes an innovative tool for the selection of the best immunomodulatory agents to implement in future DC-based vaccines.
Collapse
Affiliation(s)
- Rita Ahmed
- Department of Oncology, University Hospital of Lausanne, Lausanne, Switzerland
- Ludwig Institute for Cancer Research, University of Lausanne (UNIL), Lausanne, Switzerland
| | - Isaac Crespo
- Department of Oncology, University Hospital of Lausanne, Lausanne, Switzerland
- Ludwig Institute for Cancer Research, University of Lausanne (UNIL), Lausanne, Switzerland
- Vital-IT group, SIB Swiss Institute of Bioinformatics, University of Lausanne (UNIL), Lausanne, Switzerland
| | - Sandra Tuyaerts
- Department of Oncology, Leuven Cancer Institute (LKI), University of Leuven (KU Leuven), Leuven, Belgium
| | - Amel Bekkar
- Center for Integrative Genomics, University of Lausanne (UNIL), Lausanne, Switzerland
| | - Michele Graciotti
- Department of Oncology, University Hospital of Lausanne, Lausanne, Switzerland
- Ludwig Institute for Cancer Research, University of Lausanne (UNIL), Lausanne, Switzerland
| | - Ioannis Xenarios
- Center for Integrative Genomics, University of Lausanne (UNIL), Lausanne, Switzerland
| | - Lana E. Kandalaft
- Department of Oncology, University Hospital of Lausanne, Lausanne, Switzerland
- Ludwig Institute for Cancer Research, University of Lausanne (UNIL), Lausanne, Switzerland
| |
Collapse
|
21
|
Harari A, Graciotti M, Bassani-Sternberg M, Kandalaft LE. Antitumour dendritic cell vaccination in a priming and boosting approach. Nat Rev Drug Discov 2020; 19:635-652. [PMID: 32764681 DOI: 10.1038/s41573-020-0074-8] [Citation(s) in RCA: 186] [Impact Index Per Article: 37.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/19/2020] [Indexed: 02/06/2023]
Abstract
Mobilizing antitumour immunity through vaccination potentially constitutes a powerful anticancer strategy but has not yet provided robust clinical benefits in large patient populations. Although major hurdles still exist, we believe that currently available strategies for vaccines that target dendritic cells or use them to present antitumour antigens could be integrated into existing clinical practice using prime-boost approaches. In the priming phase, these approaches capitalize on either standard treatment modalities to trigger in situ vaccination and release tumour antigens or vaccination with dendritic cells loaded with tumour lysates or patient-specific neoantigens. In a second boost phase, personalized synthetic vaccines specifically boost T cells that were triggered during the priming phase. This immunotherapy approach has been enabled by the substantial recent improvements in dendritic cell vaccines. In this Perspective, we discuss these improvements, highlight how the prime-boost approach can be translated into clinical practice and provide solutions for various anticipated hurdles.
Collapse
Affiliation(s)
- Alexandre Harari
- Center of Experimental Therapeutics, Department of Oncology, University Hospital of Lausanne, Lausanne, Switzerland.,Ludwig Institute for Cancer Research, University of Lausanne, Lausanne, Switzerland
| | - Michele Graciotti
- Center of Experimental Therapeutics, Department of Oncology, University Hospital of Lausanne, Lausanne, Switzerland.,Ludwig Institute for Cancer Research, University of Lausanne, Lausanne, Switzerland
| | - Michal Bassani-Sternberg
- Center of Experimental Therapeutics, Department of Oncology, University Hospital of Lausanne, Lausanne, Switzerland.,Ludwig Institute for Cancer Research, University of Lausanne, Lausanne, Switzerland
| | - Lana E Kandalaft
- Center of Experimental Therapeutics, Department of Oncology, University Hospital of Lausanne, Lausanne, Switzerland. .,Ludwig Institute for Cancer Research, University of Lausanne, Lausanne, Switzerland.
| |
Collapse
|
22
|
Guo Q, Yang Q, Li J, Liu G, Nikoulin I, Jia S. Advanced clinical trials of dendritic cell vaccines in ovarian cancer. J Investig Med 2020; 68:1223-1227. [PMID: 32718940 DOI: 10.1136/jim-2020-001355] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/23/2020] [Indexed: 12/11/2022]
Abstract
Epithelial ovarian cancer (EOC) is the most common and leading cause of death for gynecologic cancer in the western world. Current standard treatments with limited selection of chemotherapies cannot meet patients' urgent needs. Immunotherapies have recently demonstrated clinical benefits in a variety of solid tumors and may offer a promising frontier for treating EOC. Dendritic cells (DCs) are key coordinators of the innate and adaptive immune system in induction of antitumor immunity. DC-based vaccinations showed clinical benefits and encouraging safety profiles in a few phase II clinical trials for patients with EOC and currently are in a phase III double-blind, randomized, placebo-controlled clinical trial. In this review, we have searched Pubmed and Clinicaltrials. gov databases for past and current phase II or phase III clinical trials with focus on EOC and DC vaccines. Outcomes and implications of the completed and ongoing trials are discussed.
Collapse
Affiliation(s)
- Quan Guo
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Qing Yang
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Jun Li
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Guipeng Liu
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Igor Nikoulin
- Research and Development, IriSys, LLC, San Diego, California, USA
| | - Steve Jia
- RD Center, Pacificbio Inc, Irvine, California, USA
| |
Collapse
|
23
|
Brunekreeft KL, Paijens ST, Wouters MC, Komdeur FL, Eggink FA, Lubbers JM, Workel HH, Van Der Slikke EC, Pröpper NE, Leffers N, Adam J, Pijper H, Plat A, Kol A, Nijman HW, De Bruyn M. Deep immune profiling of ovarian tumors identifies minimal MHC-I expression after neoadjuvant chemotherapy as negatively associated with T-cell-dependent outcome. Oncoimmunology 2020; 9:1760705. [PMID: 32923120 PMCID: PMC7458665 DOI: 10.1080/2162402x.2020.1760705] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Accepted: 01/30/2020] [Indexed: 12/12/2022] Open
Abstract
Epithelial Ovarian cancer (EOC) is the most lethal gynecological malignancy and has limited curative therapeutic options. Immunotherapy for EOC is promising, but clinical efficacy remains restricted to a small percentage of patients. Several lines of evidence suggest that the low response rate might be improved by combining immunotherapy with carboplatin and paclitaxel, the standard-of-care chemotherapy for EOC. Here, we assessed the immune contexture of EOC tumors, draining lymph nodes, and peripheral blood mononuclear cells during carboplatin/paclitaxel chemotherapy. We observed that the immune contexture of EOC patients is defined by the tissue of origin, independent of exposure to chemotherapy. Summarized, draining lymph nodes were characterized by a quiescent microenvironment composed of mostly non-proliferating naïve CD4 + T cells. Circulating T cells shared phenotypic features of both lymph nodes and tumor-infiltrating immune cells. Immunologically 'hot' ovarian tumors were characterized by ICOS, GITR, and PD-1 expression on CD4 + and CD8 + cells, independent of chemotherapy. The presence of PD-1 + cells in tumors prior to, but not after, chemotherapy was associated with disease-specific survival (DSS). Accordingly, we observed high MHC-I expression in tumors prior to chemotherapy, but minimal MHC-I expression in tumors after neoadjuvant chemotherapy, even though there were no differences in the number of tumor-infiltrating lymphocytes (TIL) in both groups. We therefore speculate that the TIL influx into the chemotherapy tumor microenvironment may be a consequence of the general inflammatory nature of chemotherapy-experienced tumors. Strategies to upregulate MHC-I during or after neoadjuvant chemotherapy may thus improve treatment outcome in these patients.
Collapse
Affiliation(s)
- Kim L. Brunekreeft
- Department of Obstetrics and Gynecology, University of Groningen, University Medical Center Groningen, The Netherlands
| | - Sterre T. Paijens
- Department of Obstetrics and Gynecology, University of Groningen, University Medical Center Groningen, The Netherlands
| | | | - Fenne L. Komdeur
- Department of Obstetrics and Gynecology, University of Groningen, University Medical Center Groningen, The Netherlands
| | - Florine A. Eggink
- Department of Obstetrics and Gynecology, University of Groningen, University Medical Center Groningen, The Netherlands
| | - Joyce M. Lubbers
- Department of Obstetrics and Gynecology, University of Groningen, University Medical Center Groningen, The Netherlands
| | - Hagma H. Workel
- Department of Obstetrics and Gynecology, University of Groningen, University Medical Center Groningen, The Netherlands
| | - Elisabeth C. Van Der Slikke
- Department of Obstetrics and Gynecology, University of Groningen, University Medical Center Groningen, The Netherlands
| | - Noor E.J. Pröpper
- Department of Obstetrics and Gynecology, University of Groningen, University Medical Center Groningen, The Netherlands
| | - Ninke Leffers
- Department of Obstetrics and Gynecology, University of Groningen, University Medical Center Groningen, The Netherlands
| | - Julien Adam
- Department of Clinical Biology, Institut De Cancérologie Gustave Roussy, Paris, France
| | - Harry Pijper
- Department of Obstetrics and Gynecology, University of Groningen, University Medical Center Groningen, The Netherlands
| | - Annechien Plat
- Department of Obstetrics and Gynecology, University of Groningen, University Medical Center Groningen, The Netherlands
| | - Arjan Kol
- Department of Obstetrics and Gynecology, University of Groningen, University Medical Center Groningen, The Netherlands
| | - Hans W. Nijman
- Department of Obstetrics and Gynecology, University of Groningen, University Medical Center Groningen, The Netherlands
| | - Marco De Bruyn
- Department of Obstetrics and Gynecology, University of Groningen, University Medical Center Groningen, The Netherlands
| |
Collapse
|
24
|
Krotova K, Day A, Aslanidi G. An Engineered AAV6-Based Vaccine Induces High Cytolytic Anti-Tumor Activity by Directly Targeting DCs and Improves Ag Presentation. Mol Ther Oncolytics 2019; 15:166-177. [PMID: 31720373 PMCID: PMC6838889 DOI: 10.1016/j.omto.2019.10.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Accepted: 10/01/2019] [Indexed: 12/30/2022] Open
Abstract
We have previously shown that an AAV6-based vaccine generates high levels of antigen-specific CD8+ T cells. Further modifications described here led to significantly increased levels of antigen-specific CD8+ and CD4+ T cells, enhanced formation of memory cells, and superior antigen-specific killing capacity in a murine model. By tracking reporter-gene-positive dendritic cells, we showed that they were directly targeted with modified AAV6 in vivo. Our vaccine's anti-cancer potential was evaluated with the antigen ovalbumin against a B16F10 melanoma cell line stably expressing ovalbumin. The vaccination showed superior protection in a murine model of metastatic melanoma. The vaccination significantly delayed solid tumor growth but did not completely prevent tumor development. We show that tumors in immunized mice escaped vaccine-induced killing by losing ovalbumin expression. The vaccine induced massive tumor infiltration with NK and CD8+ T cells with upregulated PD-1 expression. Thus, a vaccination of a combination of anti-PD-1 antibodies demonstrated significant improvement in the treatment efficacy. To summarize, we showed that a bioengineered AAV6-based vaccine elicits strong and long-lasting cellular and humoral responses against an encoded antigen. To increase AAV vaccine efficiency and mitigate tumor escape through antigen loss, we intended to target several antigens in combination with treatments targeting the tumor microenvironment.
Collapse
Affiliation(s)
- Karina Krotova
- The Hormel Institute, University of Minnesota, Austin, MN, USA
| | - Andrew Day
- The Hormel Institute, University of Minnesota, Austin, MN, USA
| | - George Aslanidi
- The Hormel Institute, University of Minnesota, Austin, MN, USA
| |
Collapse
|
25
|
Sarivalasis A, Boudousquié C, Balint K, Stevenson BJ, Gannon PO, Iancu EM, Rossier L, Martin Lluesma S, Mathevet P, Sempoux C, Coukos G, Dafni U, Harari A, Bassani-Sternberg M, Kandalaft LE. A Phase I/II trial comparing autologous dendritic cell vaccine pulsed either with personalized peptides (PEP-DC) or with tumor lysate (OC-DC) in patients with advanced high-grade ovarian serous carcinoma. J Transl Med 2019; 17:391. [PMID: 31771601 PMCID: PMC6880492 DOI: 10.1186/s12967-019-02133-w] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Accepted: 11/09/2019] [Indexed: 02/07/2023] Open
Abstract
Background Most ovarian cancer patients are diagnosed at a late stage with 85% of them relapsing after surgery and standard chemotherapy; for this reason, new treatments are urgently needed. Ovarian cancer has become a candidate for immunotherapy by reason of their expression of shared tumor-associated antigens (TAAs) and private mutated neoantigens (NeoAgs) and the recognition of the tumor by the immune system. Additionally, the presence of intraepithelial tumor infiltrating lymphocytes (TILs) is associated with improved progression-free and overall survival of patients with ovarian cancer. The aim of active immunotherapy, including vaccination, is to generate a new anti-tumor response and amplify an existing immune response. Recently developed NeoAgs-based cancer vaccines have the advantage of being more tumor specific, reducing the potential for immunological tolerance, and inducing robust immunogenicity. Methods We propose a randomized phase I/II study in patients with advanced ovarian cancer to compare the immunogenicity and to assess safety and feasibility of two personalized DC vaccines. After standard of care surgery and chemotherapy, patients will receive either a novel vaccine consisting of autologous DCs pulsed with up to ten peptides (PEP-DC), selected using an agnostic, yet personalized, epitope discovery algorithm, or a sequential combination of a DC vaccine loaded with autologous oxidized tumor lysate (OC-DC) prior to an equivalent PEP-DC vaccine. All vaccines will be administered in combination with low-dose cyclophosphamide. This study is the first attempt to compare the two approaches and to use NeoAgs-based vaccines in ovarian cancer in the adjuvant setting. Discussion The proposed treatment takes advantage of the beneficial effects of pre-treatment with OC-DC prior to PEP-DC vaccination, prompting immune response induction against a wide range of patient-specific antigens, and amplification of pre-existing NeoAgs-specific T cell clones. Trial registration This trial is already approved by Swissmedic (Ref.: 2019TpP1004) and will be registered at http://www.clinicaltrials.gov before enrollment opens.
Collapse
Affiliation(s)
- Apostolos Sarivalasis
- Department of Oncology, Centre Hospitalier Universitaire Vaudois and University of Lausanne, Lausanne, Switzerland
| | - Caroline Boudousquié
- Department of Oncology, Centre Hospitalier Universitaire Vaudois and University of Lausanne, Lausanne, Switzerland
| | - Klara Balint
- Department of Oncology, Centre Hospitalier Universitaire Vaudois and University of Lausanne, Lausanne, Switzerland.,Ludwig Institute for Cancer Research, University of Lausanne, Lausanne, Switzerland
| | | | - Philippe O Gannon
- Department of Oncology, Centre Hospitalier Universitaire Vaudois and University of Lausanne, Lausanne, Switzerland
| | - Emanuela Marina Iancu
- Department of Oncology, Centre Hospitalier Universitaire Vaudois and University of Lausanne, Lausanne, Switzerland
| | - Laetitia Rossier
- Department of Oncology, Centre Hospitalier Universitaire Vaudois and University of Lausanne, Lausanne, Switzerland.,Ludwig Institute for Cancer Research, University of Lausanne, Lausanne, Switzerland
| | - Silvia Martin Lluesma
- Department of Oncology, Centre Hospitalier Universitaire Vaudois and University of Lausanne, Lausanne, Switzerland.,Ludwig Institute for Cancer Research, University of Lausanne, Lausanne, Switzerland
| | - Patrice Mathevet
- Women-Mother-Child Department, Service of Gynecology, University Hospital of Lausanne, Lausanne, Switzerland
| | - Christine Sempoux
- Department of Pathology, University Hospital of Lausanne, Lausanne, Switzerland
| | - George Coukos
- Department of Oncology, Centre Hospitalier Universitaire Vaudois and University of Lausanne, Lausanne, Switzerland.,Ludwig Institute for Cancer Research, University of Lausanne, Lausanne, Switzerland
| | - Urania Dafni
- Laboratory of Biostatistics, School of Health Sciences, National and Kapodistrian, University of Athens, Athens, Greece
| | - Alexandre Harari
- Department of Oncology, Centre Hospitalier Universitaire Vaudois and University of Lausanne, Lausanne, Switzerland.,Ludwig Institute for Cancer Research, University of Lausanne, Lausanne, Switzerland
| | - Michal Bassani-Sternberg
- Department of Oncology, Centre Hospitalier Universitaire Vaudois and University of Lausanne, Lausanne, Switzerland.,Ludwig Institute for Cancer Research, University of Lausanne, Lausanne, Switzerland
| | - Lana E Kandalaft
- Department of Oncology, Centre Hospitalier Universitaire Vaudois and University of Lausanne, Lausanne, Switzerland. .,Ludwig Institute for Cancer Research, University of Lausanne, Lausanne, Switzerland.
| |
Collapse
|
26
|
Dendritic Cells Loaded with Heat Shock-Conditioned Ovarian Epithelial Carcinoma Cell Lysates Elicit T Cell-Dependent Antitumor Immune Responses In Vitro. J Immunol Res 2019; 2019:9631515. [PMID: 31886313 PMCID: PMC6899292 DOI: 10.1155/2019/9631515] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Revised: 09/08/2019] [Accepted: 10/16/2019] [Indexed: 12/13/2022] Open
Abstract
Ovarian epithelial carcinoma (OEC) is the most frequent ovarian tumor, characterized by a high mortality in advanced stages where conventional therapies are not effective. Based on the role of the immune system in the progression of this disease, immunotherapy using checkpoint blockade has been considered as a therapeutic alternative. Nevertheless, its results do not match up to the positive results in entities like melanoma and other malignancies, suggesting the need to find other therapies to be used alone or in combination. Dendritic cell- (DC-) based vaccines have shown promising results in several types of cancer, such as melanoma, prostate, and lung cancers, due to the essential role played by DCs in the activation of specific T cells, thus using other ways of activating the immune response than immune checkpoint blockade. During the last decade, we have used DC-based vaccines loaded with an allogeneic heat shock-conditioned melanoma cell lysate in the treatment of advanced stage patients in a series of clinical trials. In these studies, 60% of treated patients showed immunological responses which correlated positively with improved survival. Considering the relevance of ovarian cancer and the promising results of our DC-based vaccine, we show here that heat shock-conditioned cell lysates derived from ovarian epithelial carcinoma cell lines have the potential to induce the phenotypic and functional maturation of human DC, which in turn, is able to induce an efficient CD4+ and CD8+ T cell-mediated immune responses against ovarian cancer cell lines in vitro. In summary, OEC heat shock-conditioned cell lysate-loaded DCs may be considered for future combined immunotherapy approaches against ovarian tumors.
Collapse
|
27
|
Kandalaft LE, Odunsi K, Coukos G. Immunotherapy in Ovarian Cancer: Are We There Yet? J Clin Oncol 2019; 37:2460-2471. [PMID: 31403857 DOI: 10.1200/jco.19.00508] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Affiliation(s)
- Lana E Kandalaft
- Ludwig Institute for Cancer Research and University of Lausanne, Lausanne, Switzerland
| | - Kunle Odunsi
- Roswell Park Comprehensive Cancer Center, Buffalo, NY
| | - George Coukos
- Ludwig Institute for Cancer Research and University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
28
|
McGray AJR, Huang RY, Battaglia S, Eppolito C, Miliotto A, Stephenson KB, Lugade AA, Webster G, Lichty BD, Seshadri M, Kozbor D, Odunsi K. Oncolytic Maraba virus armed with tumor antigen boosts vaccine priming and reveals diverse therapeutic response patterns when combined with checkpoint blockade in ovarian cancer. J Immunother Cancer 2019; 7:189. [PMID: 31315674 PMCID: PMC6637574 DOI: 10.1186/s40425-019-0641-x] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Accepted: 06/17/2019] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Cancer immunotherapies are emerging as promising treatment strategies for ovarian cancer patients that experience disease relapse following first line therapy. As such, identifying strategies to bolster anti-tumor immunity and limit immune suppression, while recognizing diverse patterns of tumor response to immunotherapy is critical to selecting treatment combinations that lead to durable therapeutic benefit. METHODS Using a pre-clinical mouse model, we evaluated a heterologous prime/boost vaccine in combination with checkpoint blockade to treat metastatic intraperitoneal ovarian cancer. Vaccine-elicited CD8+ T cell responses and changes in the tumor microenvironment following treatment were analyzed and compared to treatment outcome. Kinetics of intraperitoneal tumor growth were assessed using non-invasive magnetic resonance imaging (MRI). RESULTS Vaccine priming followed by antigen-armed oncolytic Maraba virus boosting elicited robust tumor-specific CD8+ T cell responses that improved tumor control and led to unique immunological changes in the tumor, including a signature that correlated with improved clinical outcome of ovarian cancer patients. However, this treatment was not curative and T cells in the tumor microenvironment (TME) were functionally suppressed. Combination PD-1 blockade partially overcame the adaptive resistance in the tumor observed in response to prime/boost vaccination, restoring CD8+ T cell function in the TME and enhancing the therapeutic response. Non-invasive MRI of tumors during the course of combination treatment revealed heterogeneous radiologic response patterns following treatment, including pseudo-progression, which was associated with improved tumor control prior to relapse. CONCLUSIONS Our findings point to a key hierarchical role for PD-1 signaling and adaptive immune resistance in the ovarian TME in determining the functional fate of tumor-specific CD8+ T cells, even in the context of robust therapy mediated anti-tumor immunity, as well as the ability of multiple unique patterns of therapeutic response to result in durable tumor control.
Collapse
Affiliation(s)
- A J Robert McGray
- Center for Immunotherapy, Roswell Park Comprehensive Cancer Center, Elm and Carlton Sts, Buffalo, NY, 14263, USA
| | - Ruea-Yea Huang
- Center for Immunotherapy, Roswell Park Comprehensive Cancer Center, Elm and Carlton Sts, Buffalo, NY, 14263, USA
| | - Sebastiano Battaglia
- Center for Immunotherapy, Roswell Park Comprehensive Cancer Center, Elm and Carlton Sts, Buffalo, NY, 14263, USA
| | - Cheryl Eppolito
- Center for Immunotherapy, Roswell Park Comprehensive Cancer Center, Elm and Carlton Sts, Buffalo, NY, 14263, USA
| | - Anthony Miliotto
- Center for Immunotherapy, Roswell Park Comprehensive Cancer Center, Elm and Carlton Sts, Buffalo, NY, 14263, USA
| | - Kyle B Stephenson
- McMaster Immunology Research Centre, McMaster University, Hamilton, ON, Canada.,Turnstone Biologics, Ottawa, ON, Canada
| | - Amit A Lugade
- Center for Immunotherapy, Roswell Park Comprehensive Cancer Center, Elm and Carlton Sts, Buffalo, NY, 14263, USA
| | - Gill Webster
- Innate Immunotherapeutics, Auckland, NZ, New Zealand
| | - Brian D Lichty
- McMaster Immunology Research Centre, McMaster University, Hamilton, ON, Canada.,Turnstone Biologics, Ottawa, ON, Canada
| | - Mukund Seshadri
- Department of Dentistry and Maxillofacial Prosthetics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Danuta Kozbor
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Kunle Odunsi
- Center for Immunotherapy, Roswell Park Comprehensive Cancer Center, Elm and Carlton Sts, Buffalo, NY, 14263, USA.
| |
Collapse
|
29
|
Li GC, Qin XL, Song HH, Li YN, Qiu YY, Cui SC, Wang YS, Wang H, Gong JL. Upregulated microRNA-15b alleviates ovarian cancer through inhitbition of the PI3K/Akt pathway by targeting LPAR3. J Cell Physiol 2019; 234:22331-22342. [PMID: 31140597 DOI: 10.1002/jcp.28799] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2017] [Revised: 04/15/2019] [Accepted: 04/17/2019] [Indexed: 12/21/2022]
Abstract
Ovarian cancer characterizes as the fourth leading consequence of death associated with cancer for women. Accumulating evidence underscores the vital roles of microRNAs (miRNAs) in preventing ovarian cancer development. Besides, induction of the phosphatidylinositol-3 kinase/serine/threonine kinase (PI3K/Akt) pathway associated with the ovarian cancer cell migration and invasion. The study aims to examine the effects of miR-15b on the proliferation, apoptosis, and senescence of human ovarian cancer cells by binding to lysophosphatidic acid receptor 3 (LPAR3) with the involvement of the PI3K/Akt pathway. The positive expression of LPAR3 protein was detected by immunohistochemistry. Then the interaction between miR-15b and LPAR3 was examined. The possible role of miR-15b in ovarian cancer was explored using gain- and loss-of-function experiments. Subsequently, the functions of miR-15b on PI3K/Akt pathway, proliferation, migration, invasion, senescence and apoptosis of ovarian cancer cells were assessed. Furthermore, in vivo tumorigenicity assay in nude mice was performed. LPAR3 was overexpressed, whereas miR-15b was poorly expressed in ovarian cancer tissues. LPAR3 is a direct target of miR-15b. Restored miR-15b promoted Bax expression, apoptosis, and senescence, inhibited expression of LPAR3 and Bcl-2, the extent of PI3K and Akt phosphorylation, as well as ovarian cancer cell proliferation, migration, and invasion. Further, tumor growth was observed to be prevented by miR-15b overexpression. Collectively, our study demonstrates that miR-15b represses the proliferation and drives the senescence and apoptosis of ovarian cancer cells through the suppression of LPAR3 and the PI3K/Akt pathway, highlighting an antitumorigenic role of miR-15b.
Collapse
Affiliation(s)
- Guang-Cai Li
- Department of Obstetrics and Gynecology, Linyi People's Hospital, Linyi, People's Republic of China
| | - Xu-Ling Qin
- Department of Emergency, Economic and Technological Development Zone People's Hospital of Linyi, Linyi, People's Republic of China
| | - Huai-Hua Song
- Department of Obstetrics and Gynecology, The Third People's Hospital of Linyi, Linyi, People's Republic of China
| | - Ying-Ni Li
- Department of Obstetrics and Gynecology, Economic and Technological Development Zone People's Hospital of Linyi, Linyi, People's Republic of China
| | - Yu-Yan Qiu
- Department of Obstetrics and Gynecology, Economic and Technological Development Zone People's Hospital of Linyi, Linyi, People's Republic of China
| | - Shi-Chang Cui
- Department of General Surgery, Linyi Central Hospital, Linyi, People's Republic of China
| | - Yong-Sheng Wang
- Department of Obstetrics and Gynecology, Linyi People's Hospital, Linyi, People's Republic of China
| | - Hui Wang
- Department of Obstetrics and Gynecology, Linyi People's Hospital, Linyi, People's Republic of China
| | - Jun-Ling Gong
- Department of Obstetrics and Gynecology, Linyi People's Hospital, Linyi, People's Republic of China
| |
Collapse
|
30
|
Abstract
The phenotype and functionalities of the major immune cell subsets including myeloid cells, macrophages, dendritic cells, and T cells are altered in the ovarian cancer microenvironment. Immunosuppressive networks including inhibitory B7 family members and regulatory T cell-associated adenosine pathway have been defined in human ovarian cancer. In this review, the authors integrate emerging information on immunosuppressive mechanisms and T cell phenotype and discuss strategies of immunotherapeutic and vaccine regimens. Finally, crucial points regarding design of immuno-oncology clinical trials are reviewed.
Collapse
Affiliation(s)
- Weimin Wang
- Department of Surgery, University of Michigan School of Medicine, BSRB 5448, 109 Zina Pitcher Place, Ann Arbor, MI 48109-0669, USA
| | - Janice Rebecca Liu
- Department of Obstetrics and Gynecology, University of Michigan School of Medicine, L4604 WH, 1500 East Medical Center, Ann Arbor, MI 48109, USA
| | - Weiping Zou
- Department of Surgery, University of Michigan School of Medicine, BSRB 5071, 109 Zina Pitcher Place, Ann Arbor, MI 48109-0669, USA.
| |
Collapse
|
31
|
Ovarian Cancer Stem Cells with High ROR1 Expression Serve as a New Prophylactic Vaccine for Ovarian Cancer. J Immunol Res 2019; 2019:9394615. [PMID: 31008116 PMCID: PMC6441513 DOI: 10.1155/2019/9394615] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Revised: 11/25/2018] [Accepted: 01/20/2019] [Indexed: 01/16/2023] Open
Abstract
Tumor vaccines offer a number of advantages for cancer treatment. In the study, the vaccination with cancer stem cells (CSCs) with high expression of the type I receptor tyrosine kinase-like orphan receptor (ROR1) was evaluated in a murine model for the vaccine's immunogenicity and protective efficacy against epithelial ovarian carcinoma (EOC). CD117+CD44+ CSCs were isolated from human EOC HO8910 cell line using a magnetic-activated cell sorting system; murine ID8 EOC suspension sphere cells, which are collectively known as cancer stem-like cells, were acquired from serum-free suspension sphere-forming culture. Mice were subcutaneously immunized with the repeat cycles of freezing and thawing whole HO8910 CD117+CD44+ CSCs and ID8 cancer stem-like cells, respectively, followed by a challenge with HO8910 or ID8 cells at one week after final vaccination. The results showed that the CSC vaccination significantly induced immunity against EOC growth and markedly prolonged the survival of EOC-bearing mice in the prophylactic setting compared with non-CSC vaccination. Flow cytometry showed significantly increased immunocyte cytotoxicities and remarkably reduced CSC counts in the CSC-vaccinated mice. Moreover, the protective efficacy against EOC was decreased when the ROR1 expression was downregulated by shRNA in CSC vaccines. The findings from the study suggest that CSC vaccines with high ROR1 expression were highly effective in triggering immunity against EOC in vaccinated mice and may serve as an effective vaccine for EOC immunoprophylaxis.
Collapse
|
32
|
Moufarrij S, Dandapani M, Arthofer E, Gomez S, Srivastava A, Lopez-Acevedo M, Villagra A, Chiappinelli KB. Epigenetic therapy for ovarian cancer: promise and progress. Clin Epigenetics 2019; 11:7. [PMID: 30646939 PMCID: PMC6334391 DOI: 10.1186/s13148-018-0602-0] [Citation(s) in RCA: 204] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Accepted: 12/19/2018] [Indexed: 12/14/2022] Open
Abstract
Ovarian cancer is the deadliest gynecologic malignancy, with a 5-year survival rate of approximately 47%, a number that has remained constant over the past two decades. Early diagnosis improves survival, but unfortunately only 15% of ovarian cancers are diagnosed at an early or localized stage. Most ovarian cancers are epithelial in origin and treatment prioritizes surgery and cytoreduction followed by cytotoxic platinum and taxane chemotherapy. While most tumors will initially respond to this treatment, recurrence is likely to occur within a median of 16 months for patients who present with advanced stage disease. New treatment options separate from traditional chemotherapy that take advantage of advances in understanding of the pathophysiology of ovarian cancer are needed to improve outcomes. Recent work has shown that mutations in genes encoding epigenetic regulators are mutated in ovarian cancer, driving tumorigenesis and resistance to treatment. Several of these epigenetic modifiers have emerged as promising drug targets for ovarian cancer therapy. In this article, we delineate epigenetic abnormalities in ovarian cancer, discuss key scientific advances using epigenetic therapies in preclinical ovarian cancer models, and review ongoing clinical trials utilizing epigenetic therapies in ovarian cancer.
Collapse
Affiliation(s)
- Sara Moufarrij
- Department of Microbiology, Immunology, & Tropical Medicine, The George Washington University, Washington, D.C., 20052 USA
- Department of Obstetrics & Gynecology, The George Washington University, Washington, D.C., 20052 USA
- Department of Biochemistry & Molecular Medicine, The George Washington University, Washington, D.C., 20052 USA
- The George Washington Cancer Center, The George Washington University, Washington, D.C., 20052 USA
| | - Monica Dandapani
- Department of Microbiology, Immunology, & Tropical Medicine, The George Washington University, Washington, D.C., 20052 USA
- Department of Obstetrics & Gynecology, The George Washington University, Washington, D.C., 20052 USA
- The George Washington Cancer Center, The George Washington University, Washington, D.C., 20052 USA
| | - Elisa Arthofer
- Department of Microbiology, Immunology, & Tropical Medicine, The George Washington University, Washington, D.C., 20052 USA
- The George Washington Cancer Center, The George Washington University, Washington, D.C., 20052 USA
| | - Stephanie Gomez
- Department of Microbiology, Immunology, & Tropical Medicine, The George Washington University, Washington, D.C., 20052 USA
- The George Washington Cancer Center, The George Washington University, Washington, D.C., 20052 USA
| | - Aneil Srivastava
- Department of Microbiology, Immunology, & Tropical Medicine, The George Washington University, Washington, D.C., 20052 USA
- The George Washington Cancer Center, The George Washington University, Washington, D.C., 20052 USA
| | - Micael Lopez-Acevedo
- Department of Obstetrics & Gynecology, The George Washington University, Washington, D.C., 20052 USA
- The George Washington Cancer Center, The George Washington University, Washington, D.C., 20052 USA
| | - Alejandro Villagra
- Department of Biochemistry & Molecular Medicine, The George Washington University, Washington, D.C., 20052 USA
- The George Washington Cancer Center, The George Washington University, Washington, D.C., 20052 USA
| | - Katherine B. Chiappinelli
- Department of Microbiology, Immunology, & Tropical Medicine, The George Washington University, Washington, D.C., 20052 USA
- The George Washington Cancer Center, The George Washington University, Washington, D.C., 20052 USA
| |
Collapse
|
33
|
Maletzki C, Wiegele L, Nassar I, Stenzel J, Junghanss C. Chemo-immunotherapy improves long-term survival in a preclinical model of MMR-D-related cancer. J Immunother Cancer 2019; 7:8. [PMID: 30630527 PMCID: PMC6329128 DOI: 10.1186/s40425-018-0476-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Accepted: 12/06/2018] [Indexed: 01/12/2023] Open
Abstract
BACKGROUND Mismatch Repair Deficiency (MMR-D)-related tumors are highly immunogenic and constitute ideal vaccination targets. In a proof-of-concept study delayed tumorigenesis and prolonged survival has been shown in a clinically-relevant mouse model for MMR-D-related diseases (=MLH1 knock out mice). To refine this approach, vaccination was combined with immune modulatory low-dose chemotherapy to polarize immune regulatory subtypes. METHODS Mice (prophylactic: 8-10 weeks; therapeutic: > 36 weeks) received a single injection of cyclophosphamide (CPX, 120 mg/kg bw, i.p.) or gemcitabine (GEM, 100 mg/kg bw, i.p.) prior to vaccination (lysate of a gastrointestinal tumor allograft, 10 mg/kg bw, n = 9 mice/group). The vaccine was given repetitively (10 mg/kg bw, s.c., 4 x / once a week, followed by monthly boosts) until tumor formation or progression. Tumor growth ([18F] FDG PET/CT imaging) and immune responses were monitored (flow cytometry, IFNγ ELISpot). The microenvironment was analyzed by immunofluorescence. RESULTS Prophylactic application of GEM + lysate delayed tumorigenesis compared to lysate monotherapy and CPX-pre-treatment (median time of onset: 53 vs. 47 vs. 48 weeks). 33% of mice even remained tumor-free until the experimental endpoint (= 65 weeks). This was accompanied by long-term effect on cytokine plasma levels; splenic myeloid derived suppressor cells (MDSC) as well as regulatory T cell numbers. Assessment of tumor microenvironment from GEM + lysate treated mice revealed low numbers of MDSCs, but enhanced T cell infiltration, in some cases co-expressing PD-L1. Therapeutic chemo-immunotherapy (GEM + lysate) had minor impact on overall survival (median time: 12 (GEM + lysate) vs. 11.5 (lysate) vs. 3 weeks (control)), but induced complete remission in one case. Dendritic and T cell infiltrates increased in both treatment groups. Reactive T cells specifically recognized MLH1-/- tumor cells in IFNγ ELISpot, but lacked response towards NK cell targets YAC-1. CONCLUSIONS Combined chemo-immunotherapy impairs tumor onset and growth likely attributable to modulation of immune responses. Depleting or 're-educating' immunosuppressive cell types, such as MDSC, may help moving a step closer to combat cancer.
Collapse
Affiliation(s)
- Claudia Maletzki
- Department of Medicine, Clinic III - Hematology, Oncology, Palliative Medicine, Rostock University Medical Center, Ernst-Heydemann-Str. 6, 18057, Rostock, Germany.
| | - Leonie Wiegele
- Department of Medicine, Clinic III - Hematology, Oncology, Palliative Medicine, Rostock University Medical Center, Ernst-Heydemann-Str. 6, 18057, Rostock, Germany
| | - Ingy Nassar
- Department of Medicine, Clinic III - Hematology, Oncology, Palliative Medicine, Rostock University Medical Center, Ernst-Heydemann-Str. 6, 18057, Rostock, Germany
| | - Jan Stenzel
- Core Facility Multimodal Small Animal Imaging, Rostock University Medical Center, Schillingallee 69a, 18057, Rostock, Germany
| | - Christian Junghanss
- Department of Medicine, Clinic III - Hematology, Oncology, Palliative Medicine, Rostock University Medical Center, Ernst-Heydemann-Str. 6, 18057, Rostock, Germany
| |
Collapse
|
34
|
IL-15 and a Two-Step Maturation Process Improve Bone Marrow-Derived Dendritic Cell Cancer Vaccine. Cancers (Basel) 2019; 11:cancers11010040. [PMID: 30621204 PMCID: PMC6356194 DOI: 10.3390/cancers11010040] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Revised: 12/20/2018] [Accepted: 12/21/2018] [Indexed: 12/12/2022] Open
Abstract
In the last 20 years, dendritic cells (DCs) have been largely used as a platform for therapeutic vaccination in cancer patients. However, despite its proven safety and ability to induce cancer specific immune responses, the clinical benefits of DC-based immunotherapy are currently very limited. Thus, novel approaches are still needed to boost its efficacy. Our group recently showed that squaric acid treatment of antigens is an important adjuvant that can increase vaccine-induced downstream immune responses and therapeutic outcomes. Here we further improved this dendritic cell vaccine formulation by developing a new method for differentiating and maturing DCs from their bone marrow precursors. Our data demonstrate that bone marrow-derived DCs differentiated with GM-CSF and IL-15 and matured with a maturation cocktail in two steps present a more mature and immunogenic phenotype, compared to standard DC preparations. Further suppression of the prostaglandin E₂ pathway achieved even more immunogenic DC phenotypes. This vaccine was more potent at delaying tumor growth, improved animal survival and induced a more immunogenic and Th1-skewed T cell response in an ovarian cancer mouse model. These promising results support future efforts for the clinical translation of this approach.
Collapse
|
35
|
Martin Lluesma S, Graciotti M, Chiang CLL, Kandalaft LE. Does the Immunocompetent Status of Cancer Patients Have an Impact on Therapeutic DC Vaccination Strategies? Vaccines (Basel) 2018; 6:E79. [PMID: 30477198 PMCID: PMC6313858 DOI: 10.3390/vaccines6040079] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Revised: 11/09/2018] [Accepted: 11/21/2018] [Indexed: 12/24/2022] Open
Abstract
Although different types of therapeutic vaccines against established cancerous lesions in various indications have been developed since the 1990s, their clinical benefit is still very limited. This observed lack of effectiveness in cancer eradication may be partially due to the often deficient immunocompetent status of cancer patients, which may facilitate tumor development by different mechanisms, including immune evasion. The most frequently used cellular vehicle in clinical trials are dendritic cells (DCs), thanks to their crucial role in initiating and directing immune responses. Viable vaccination options using DCs are available, with a positive toxicity profile. For these reasons, despite their limited therapeutic outcomes, DC vaccination is currently considered an additional immunotherapeutic option that still needs to be further explored. In this review, we propose potential actions aimed at improving DC vaccine efficacy by counteracting the detrimental mechanisms recognized to date and implicated in establishing a poor immunocompetent status in cancer patients.
Collapse
Affiliation(s)
- Silvia Martin Lluesma
- Center of Experimental Therapeutics, Ludwig Center for Cancer Research, Department of Oncology, University of Lausanne, Lausanne 1011, Switzerland.
| | - Michele Graciotti
- Vaccine development laboratory, Ludwig Center for Cancer Research, Lausanne 1011, Switzerland.
| | - Cheryl Lai-Lai Chiang
- Vaccine development laboratory, Ludwig Center for Cancer Research, Lausanne 1011, Switzerland.
| | - Lana E Kandalaft
- Center of Experimental Therapeutics, Ludwig Center for Cancer Research, Department of Oncology, University of Lausanne, Lausanne 1011, Switzerland.
- Vaccine development laboratory, Ludwig Center for Cancer Research, Lausanne 1011, Switzerland.
| |
Collapse
|
36
|
Gao Q, Xiang SD, Wilson K, Madondo M, Stephens AN, Plebanski M. Sperm Protein 17 Expression by Murine Epithelial Ovarian Cancer Cells and Its Impact on Tumor Progression. Cancers (Basel) 2018; 10:cancers10080276. [PMID: 30127274 PMCID: PMC6115966 DOI: 10.3390/cancers10080276] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Revised: 08/13/2018] [Accepted: 08/14/2018] [Indexed: 12/12/2022] Open
Abstract
The cancer testis antigen sperm protein 17 (Sp17) is a promising antigenic target in epithelial ovarian cancer (EOC) vaccine development. However, its role in ovarian cancer is unclear. We isolated and expanded Sp17+ and Sp17− clones from the murine EOC cell line ID8, and compared their in-vitro cell growth characteristics and in-vivo tumorigenicity. We also examined the potential co-expression of molecules that may influence cancer cell survival and interaction with immune cells. These include stimulatory and immunosuppressive molecules, such as major histocompatibility class I molecules (MHC I), MHC II, cytotoxic T lymphocyte associated antigen-4 (CTLA-4), CD73, CD39, tumor necrosis factor receptor II (TNFRII), signal transducer and activator of transcription 3 (STAT3) and programmed death-ligand 1 (PD-L1). Whilst the presence of Sp17 was not correlated with the ID8 cell proliferation/growth capacity in vitro, it was critical to enable progressive tumor formation in vivo. Flow cytometry revealed that Sp17+ ID8 cells displayed higher expression of both STAT3 and PD-L1, whilst MHC II expression was lower. Moreover, Sp17high (PD-L1+MHCII−) cell populations showed significantly enhanced resistance to Paclitaxel-induced cell death in vitro compared to Sp17low (PD-L1−MHCII+) cells, which was associated in turn with increased STAT3 expression. Together, the data support Sp17 as a factor associated with in-vivo tumor progression and chemo-resistance, validating it as a suitable target for vaccine development.
Collapse
Affiliation(s)
- Qian Gao
- Department of Immunology and Pathology, Central Clinical School, Faculty of Medicine, Nursing and Health Science, Monash University, Melbourne, Victoria 3004, Australia.
- Department of Clinical Laboratory, Xiangya Hospital, Central South University, Changsha 410008, China.
| | - Sue D Xiang
- Department of Immunology and Pathology, Central Clinical School, Faculty of Medicine, Nursing and Health Science, Monash University, Melbourne, Victoria 3004, Australia.
- Centre for Cancer Research, Hudson Institute of Medical Research, Clayton, Victoria 3168, Australia.
- Department of Molecular and Translational Sciences, Monash University, Clayton, Victoria 3168, Australia.
| | - Kirsty Wilson
- Department of Immunology and Pathology, Central Clinical School, Faculty of Medicine, Nursing and Health Science, Monash University, Melbourne, Victoria 3004, Australia.
| | - Mutsa Madondo
- Department of Immunology and Pathology, Central Clinical School, Faculty of Medicine, Nursing and Health Science, Monash University, Melbourne, Victoria 3004, Australia.
| | - Andrew N Stephens
- Centre for Cancer Research, Hudson Institute of Medical Research, Clayton, Victoria 3168, Australia.
- Department of Molecular and Translational Sciences, Monash University, Clayton, Victoria 3168, Australia.
| | - Magdalena Plebanski
- Department of Immunology and Pathology, Central Clinical School, Faculty of Medicine, Nursing and Health Science, Monash University, Melbourne, Victoria 3004, Australia.
- School of Health and Biomedical Sciences, RMIT University, Bundoora, Victoria 3083, Australia.
| |
Collapse
|
37
|
Qu X, Tang Y, Hua S. Immunological Approaches Towards Cancer and Inflammation: A Cross Talk. Front Immunol 2018; 9:563. [PMID: 29662489 PMCID: PMC5890100 DOI: 10.3389/fimmu.2018.00563] [Citation(s) in RCA: 197] [Impact Index Per Article: 28.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Accepted: 03/06/2018] [Indexed: 12/12/2022] Open
Abstract
The inflammation is the protective response of the body against various harmful stimuli; however, the aberrant and inappropriate activation tends to become harmful. The acute inflammatory response tends to resolved once the offending agent is subside but this acute response becomes chronic in nature when the body is unable to successfully neutralized the noxious stimuli. This chronic inflammatory microenvironment is associated with the release of various pro-inflammatory and oncogenic mediators such as nitric oxide (NO), cytokines [IL-1β, IL-2, interleukin-6 (IL-6), and tumor necrosis factor-α (TNF-α)], growth factor, and chemokines. These mediators make the inflammatory microenvironment more vulnerable toward tumorigenesis. The pro-inflammatory mediators released during the chronic inflammation tends to induce several molecular signaling cascades such as nuclear factor kappa B, MAPKinase, nuclear factor erythroid 2-related factor 2, phosphoinositide-3-kinase, Janus kinases/STAT, Wnt/B-catenin, and cyclic AMP response element binding protein. The immune system and its components have a pleiotropic effect on inflammation and cancer progression. Immune components such as T cells, natural killer cells, macrophages, and neutrophils either inhibit or enhance tumor initiation depending on the type of tumor and immune cells involved. Tumor-associated macrophages and tumor-associated neutrophils are pro-tumorigenic cells highly prevalent in inflammation-mediated tumors. Similarly, presence of T regulatory (Treg) cells in an inflammatory and tumor setting suppresses the immune system, thus paving the way for oncogenesis. However, Treg cells also inhibit autoimmune inflammation. By contrast, cytotoxic T cells and T helper cells confer antitumor immunity and are associated with better prognosis in patients with cancer. Cytotoxic T cells inflict a direct cytotoxic effect on cells expressing oncogenic markers. Currently, several anti-inflammatory and antitumor therapies are under trials in which these immune cells are exploited. Adoptive cell transfer composed of tumor-infiltrating lymphocytes has been tried for the treatment of tumors after their ex vivo expansion. Mediators released by cells in a tumorigenic and inflammatory microenvironment cross talk with nearby cells, either promoting or inhibiting inflammation and cancer. Recently, several cytokine-based therapies are either being developed or are under trial to treat such types of manifestations. Monoclonal antibodies directed against TNF-α, VEGF, and IL-6 has shown promising results to ameliorate inflammation and cancer, while direct administration of IL-2 has been shown to cause tumor regression.
Collapse
Affiliation(s)
- Xinglong Qu
- Department of Respiration, The First Hospital of Jilin University, Changchun, China
| | - Ying Tang
- Department of Respiration, The First Hospital of Jilin University, Changchun, China
| | - Shucheng Hua
- Department of Respiration, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
38
|
Pang J, Jiang P, Wang Y, Jiang L, Qian H, Tao Y, Shi R, Gao J, Chen Y, Wu Y. Cross-linked hyaluronan gel inhibits the growth and metastasis of ovarian carcinoma. J Ovarian Res 2018; 11:22. [PMID: 29510732 PMCID: PMC5840805 DOI: 10.1186/s13048-018-0394-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2017] [Accepted: 02/28/2018] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND The recurrence, metastasis and poor prognosis are important characteristics of ovarian carcinoma (OC), which are associated with exfoliation of cells from the primary tumor and colonization of the cells in pelvic cavity. On the other hand, the life quality of the patients undergoing surgical resection of OC was influenced by postoperative adhesions. Therefore, preventing postoperative implant tumor and adhesion may be effective methods to improve OC treatment. HyaRegen Gel, a cross-linked hyaluronan gel (CHAG), has been widely used as an anti-adhesive agent following pelvic operation in clinic. However, whether it can affect the implantation and growth of OC cells or not is still not clear. METHODS Migration and invasion assays were applied to detect the effect of CHAG on migration and invasion of OC cells. Western blotting was performed to detect the phosphorylation/activation of EGFR and ERK, and the expression of PCNA and MMP7. Pull down assay was used to analyze the effect of CHAG on the activation of small G protein Rac1. Nude mice implantation tumor model was applied to observe the effect of CHAG on implantation tumor of OC cells. RESULTS The results of in vitro experiments showed that CHAG suppressed both basic and EGF-induced migration and invasion of OC cells, blocked the activation of EGF-initiated EGFR activation, inhibited downstream signal transduction of EGFR, and decreased expression of proliferation and migration/invasion related proteins. Meanwhile, results of in vivo experiments showed that CHAG not only inhibited the formation of implantation tumor of OC cells but also delayed the of the growth of the tumors. CONCLUSIONS CHAG inhibited migration, invasion and proliferation of OC cells in vitro, and suppressed development of implantation tumor of OC in vivo. This made it as both anti-tumor and anti-adhesion agents.
Collapse
Affiliation(s)
- Ji Pang
- Department of Physiology, School of Medicine, Jiangsu University, Zhenjiang City, Jiangsu Province, 212013, People's Republic of China
| | - Pengcheng Jiang
- Department of Obstetrics and Gynecology, Changzhou Second People's Hospital Affiliated to Nanjing Medical University, Changzhou City, Jiangsu Province, China
| | - Ying Wang
- Department of Physiology, School of Medicine, Jiangsu University, Zhenjiang City, Jiangsu Province, 212013, People's Republic of China
| | - Lu Jiang
- Department of Physiology, School of Medicine, Jiangsu University, Zhenjiang City, Jiangsu Province, 212013, People's Republic of China
| | - Hai Qian
- Department of Physiology, School of Medicine, Jiangsu University, Zhenjiang City, Jiangsu Province, 212013, People's Republic of China
| | - Yan Tao
- Department of Physiology, School of Medicine, Jiangsu University, Zhenjiang City, Jiangsu Province, 212013, People's Republic of China
| | - Ruxia Shi
- Department of Obstetrics and Gynecology, Changzhou Second People's Hospital Affiliated to Nanjing Medical University, Changzhou City, Jiangsu Province, China
| | - Jizong Gao
- R & D Department, Changzhou BioRegen Biomedical (Changzhou) Co., Ltd., Changzhou City, Jiangsu Province, China
| | - Yongchang Chen
- Department of Physiology, School of Medicine, Jiangsu University, Zhenjiang City, Jiangsu Province, 212013, People's Republic of China
| | - Yan Wu
- Department of Physiology, School of Medicine, Jiangsu University, Zhenjiang City, Jiangsu Province, 212013, People's Republic of China.
| |
Collapse
|
39
|
Chen Z, Lin H, Hu K, Su R, Lai N, Yang Z, Kang S. [Soluble PD-1 over-expression enhances the anti-tumor effect of senescence tumor cell vaccine against breast cancer cell growth in tumor-bearing mice]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2018; 38:20-26. [PMID: 33177023 DOI: 10.3969/j.issn.1673-4254.2018.01.04] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
OBJECTIVE To investigate whether soluble PD-1 overexpression in 4T1 senescence tumor cells enhances the antitumor effect of senescence tumor cell vaccine (STCV) against breast tumor cells in a tumor-bearing mouse model. METHODS 4T1 cells were treated with interferon-γ (IFN-γ) and the expression of PD-L1 was detected by flow cytometry. CCK8 assay was used to compare the cell proliferation activity between 4T1 cells and 4T1 cells infected by a lentiviral vector of sPD-1 (4T1/sPD-1 cells), and the expressions of sPD-1 mRNA and protein in 4T1/sPD-1 cells were detected using qPCR and Western blotting. The culture supernatant of 4T1/sPD-1 cells was added in 4T1 cells pre-treated with IFN-γ, and PD-1-positive 4T1 cells were detected with flow cytometry. Senescence β-galactosidase staining kit was used to detect the senescent 4T1 and 4T1/sPD-1 cells following exposure to X-ray radiation and Veliparib. Balb/c mice bearing subcutaneous 4T1 tumor xenografts were treated with injections of PBS, 4T1 STCV, or 4T1/sPD-1 STCV, and tumor growth was observed. RESULTS Stimulation with IFN-γ concentration-dependently up-regulated PD-L1 expression by as much as (84.80 ± 1.03)% in 4T1 cells (P < 0.001). sPD-1 overexpression in 4T1 cells did not significantly affect the cell proliferation. Treatment of 4T1 cells with 4T1/sPD-1 cell culture supernatant significantly increased the proportion of PD-1 + cells from (6.893 ± 0.271)% to (55.450 ± 0.555)% (P < 0.001). X-ray irradiation combined with Veliparib caused obvious senescence in 4T1 and 4T1/sPD-1 cells. In the tumor-preventing experiment, tumor formation occurred in all the mice in PBS group; 28.787% of the mice in 4T1 STCV group and 55.556% in 4T1/sPD-1 STCV group showed no tumor formation. In the tumor treatment experiment, tumor formation occurred in all the mice in PBS groups while in 4T1 STCV and 4T1/sPD-1 STCV groups, 11.111% and 38.89% of the mice were tumor-free during the observation period, respectively. CONCLUSIONS Senescence tumor cells vaccine has antitumor effect against breast cancer in mice, and sPD-1 overexpression can enhance this effect of the vaccine.
Collapse
Affiliation(s)
- Zehong Chen
- Department of Oncology, Cancer Center, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510315, China
| | - Huiwen Lin
- Department of Oncology, Cancer Center, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510315, China
| | - Kang Hu
- Department of Oncology, Cancer Center, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510315, China
| | - Ruxiong Su
- Department of Pharmacy, Puning Peoples' Hospital, Southern Medical University, Puning 515300, China
| | - Nan Lai
- Department of Oncology, Cancer Center, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510315, China
| | - Zike Yang
- Department of Oncology, Cancer Center, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510315, China
| | - Shijun Kang
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| |
Collapse
|
40
|
Hardwick NR, Frankel P, Ruel C, Kilpatrick J, Tsai W, Kos F, Kaltcheva T, Leong L, Morgan R, Chung V, Tinsley R, Eng M, Wilczynski S, Ellenhorn JDI, Diamond DJ, Cristea M. p53-Reactive T Cells Are Associated with Clinical Benefit in Patients with Platinum-Resistant Epithelial Ovarian Cancer After Treatment with a p53 Vaccine and Gemcitabine Chemotherapy. Clin Cancer Res 2018; 24:1315-1325. [PMID: 29301826 DOI: 10.1158/1078-0432.ccr-17-2709] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Revised: 11/27/2017] [Accepted: 12/19/2017] [Indexed: 01/05/2023]
Abstract
Purpose: To conduct a phase I trial of a Modified Vaccinia Ankara vaccine delivering wild-type human p53 (p53MVA) in combination with gemcitabine chemotherapy in patients with platinum-resistant ovarian cancer.Experimental Design: Patients received gemcitabine on days 1 and 8 and p53MVA vaccine on day 15, during the first 3 cycles of chemotherapy. Toxicity was classified using the NCI Common Toxicity Criteria and clinical response assessed by CT scan. Peripheral blood samples were collected for immunophenotyping and monitoring of anti-p53 immune responses.Results: Eleven patients were evaluated for p53MVA/gemcitabine toxicity, clinical outcome, and immunologic response. TOXICITY there were no DLTs, but 3 of 11 patients came off study early due to gemcitabine-attributed adverse events (AE). Minimal AEs were attributed to p53MVA vaccination. Immunologic and clinical response: enhanced in vitro recognition of p53 peptides was detectable after immunization in both the CD4+ and CD8+ T-cell compartments in 5 of 11 and 6 of 11 patients, respectively. Changes in peripheral T regulatory cells (Tregs) and myeloid-derived suppressor cells (MDSC) did not correlate significantly with vaccine response or progression-free survival (PFS). Patients with the greatest expansion of p53-reactive T cells had significantly longer PFS than patients with lower p53-reactivity after therapy. Tumor shrinkage or disease stabilization occurred in 4 patients.Conclusions: p53MVA was well tolerated, but gemcitabine without steroid pretreatment was intolerable in some patients. However, elevated p53-reactive CD4+ and CD8+ T-cell responses after therapy correlated with longer PFS. Therefore, if responses to p53MVA can be enhanced with alternative agents, superior clinical responses may be achievable. Clin Cancer Res; 24(6); 1315-25. ©2018 AACR.
Collapse
Affiliation(s)
- Nicola R Hardwick
- Department of Experimental Therapeutics, Beckman Research Institute, Duarte, California
| | - Paul Frankel
- Division of Biostatistics, Beckman Research Institute, Duarte, California
| | - Christopher Ruel
- Division of Biostatistics, Beckman Research Institute, Duarte, California
| | - Julie Kilpatrick
- Department of Medical Oncology and Therapeutics Research, City of Hope National Medical Center, Duarte, California
| | - Weimin Tsai
- Department of Hematology & Hematopoietic Cell Transplantation, City of Hope National Medical Center, Duarte, California
| | - Ferdynand Kos
- Department of Experimental Therapeutics, Beckman Research Institute, Duarte, California
| | - Teodora Kaltcheva
- Department of Experimental Therapeutics, Beckman Research Institute, Duarte, California
| | - Lucille Leong
- Department of Medical Oncology and Therapeutics Research, City of Hope National Medical Center, Duarte, California
| | - Robert Morgan
- Department of Medical Oncology and Therapeutics Research, City of Hope National Medical Center, Duarte, California
| | - Vincent Chung
- Department of Medical Oncology and Therapeutics Research, City of Hope National Medical Center, Duarte, California
| | - Raechelle Tinsley
- Clinical Trials Office, City of Hope National Medical Center, Duarte, California
| | - Melissa Eng
- Department of Medical Oncology and Therapeutics Research, City of Hope National Medical Center, Duarte, California
| | - Sharon Wilczynski
- Department of Pathology, City of Hope National Medical Center, Duarte, California
| | | | - Don J Diamond
- Department of Experimental Therapeutics, Beckman Research Institute, Duarte, California.
| | - Mihaela Cristea
- Department of Medical Oncology and Therapeutics Research, City of Hope National Medical Center, Duarte, California
| |
Collapse
|
41
|
Cortez AJ, Tudrej P, Kujawa KA, Lisowska KM. Advances in ovarian cancer therapy. Cancer Chemother Pharmacol 2018; 81:17-38. [PMID: 29249039 PMCID: PMC5754410 DOI: 10.1007/s00280-017-3501-8] [Citation(s) in RCA: 393] [Impact Index Per Article: 56.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Accepted: 12/11/2017] [Indexed: 02/06/2023]
Abstract
Epithelial ovarian cancer is typically diagnosed at an advanced stage. Current state-of-the-art surgery and chemotherapy result in the high incidence of complete remissions; however, the recurrence rate is also high. For most patients, the disease eventually becomes a continuum of symptom-free periods and recurrence episodes. Different targeted treatment approaches and biological drugs, currently under development, bring the promise of turning ovarian cancer into a manageable chronic disease. In this review, we discuss the current standard in the therapy for ovarian cancer, major recent studies on the new variants of conventional therapies, and new therapeutic approaches, recently approved and/or in clinical trials. The latter include anti-angiogenic therapies, polyADP-ribose polymerase (PARP) inhibitors, inhibitors of growth factor signaling, or folate receptor inhibitors, as well as several immunotherapeutic approaches. We also discuss cost-effectiveness of some novel therapies and the issue of better selection of patients for personalized treatment.
Collapse
Affiliation(s)
- Alexander J Cortez
- Maria Skłodowska-Curie Institute - Oncology Center, Gliwice Branch, Wybrzeże Armii Krajowej 15, Gliwice, 44-100, Poland
| | - Patrycja Tudrej
- Maria Skłodowska-Curie Institute - Oncology Center, Gliwice Branch, Wybrzeże Armii Krajowej 15, Gliwice, 44-100, Poland
| | - Katarzyna A Kujawa
- Maria Skłodowska-Curie Institute - Oncology Center, Gliwice Branch, Wybrzeże Armii Krajowej 15, Gliwice, 44-100, Poland
| | - Katarzyna M Lisowska
- Maria Skłodowska-Curie Institute - Oncology Center, Gliwice Branch, Wybrzeże Armii Krajowej 15, Gliwice, 44-100, Poland.
| |
Collapse
|
42
|
MtHsp70-CLIC1-pulsed dendritic cells enhance the immune response against ovarian cancer. Biochem Biophys Res Commun 2017; 494:13-19. [PMID: 29061300 DOI: 10.1016/j.bbrc.2017.10.094] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2017] [Accepted: 10/17/2017] [Indexed: 12/11/2022]
Abstract
Approximately 80% of ovarian cancer (OC) is diagnosed at late stages, and most patients die within 5 years of diagnosis due to recurrence or drug resistance. Novel treatments are required to improve patient survival. Immune therapy against cancer is promising; however, therapeutic vaccination has been limited by the inability of tumor antigens to induce effective immune responses. Chloride intracellular channel 1 (CLIC1) was previously identified as a possible tumor marker for OC. In this study, we constructed a recombinant protein by conjugating the extracellular domain of CLIC1 to the carboxyl terminus of Mycobacterium tuberculosis heat shock protein 70 (MtHsp70). Human dendritic cells (DCs) derived from cortical blood were pulsed with the fusion protein, and the antitumor effect of human cytotoxic T lymphocytes (CTLs) stimulated by autologous DCs was assessed in NOG mice. MtHsp70-CLIC1 promoted the phenotypic maturation of human DCs and the secretion of Th1-associated cytokines in vitro. MtHsp70-CLIC1-stimulated CTLs generated a CLIC1-specific immune response both in vitro and in vivo. These results indicate that DCs pulsed with MtHsp70-CLIC1 can enhance antitumor immunity against OC, providing a novel immune therapeutic strategy.
Collapse
|
43
|
Li W, Chen Y, Sheng Y, Xie Z, Tang Y. Synthesis and inhibitory effect of 10-chlorocanthin-6-one on ovarian cancer HO8910PM cells. Biotechnol Lett 2017; 40:23-30. [PMID: 28948407 DOI: 10.1007/s10529-017-2438-7] [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: 07/08/2017] [Accepted: 09/07/2017] [Indexed: 11/27/2022]
Abstract
OBJECTIVE To synthesize and determine the antitumor activity of 10-chlorocanthin-6-one in ovarian cancer HO8910PM cells. RESULTS Among the synthesized canthin-6-one analogs, 10-chlorocanthin-6-one was the most cytotoxic (IC50 = 4.9 μM), as demonstrated by a dose-dependent cytotoxicity assay. Moreover, 10-chlorocanthin-6-one induced apoptosis through the activation of poly(ADP-ribose) polymerase and caspase-3 cleavage, upregulation of Bcl-2, and downregulation of Bim, x-linked inhibitor of apoptosis protein (XIAP), and survivin in HO8910PM cells. Furthermore, Bim RNA, upregulated in a concentration-dependent manner, and knockdown of Bim via short-hairpin RNAs attenuated the inhibitory effects of 10-chlorocanthin-6-one on HO8910PM cell growth. CONCLUSIONS 10-Chlorocanthin-6-one inhibits cell proliferation and induces apoptosis in H08910PM cells. The underlying molecular mechanisms of 10-chlorocanthin-6-one include activation of the Bim-mediated mitochondrial apoptotic pathway via upregulation of Bim and downregulation of Bcl-2, XIAP, and survivin. These data suggest that Bim is a potential target of 10-chlorocanthin-6-one, further demonstrating its potential use in the prevention and treatment of ovarian cancer.
Collapse
Affiliation(s)
- Wenhua Li
- Chengdu Institute of Biology, Chinese Academy of Sciences, No 9 Section 4, Renmin Nan Road, Chengdu, 610041, Sichuan, People's Republic of China
| | - Yuwen Chen
- Chengdu Institute of Biology, Chinese Academy of Sciences, No 9 Section 4, Renmin Nan Road, Chengdu, 610041, Sichuan, People's Republic of China
- School of Life Science and Engineering, Sichuan University, Chengdu, China
| | - Yuwen Sheng
- Chengdu Institute of Biology, Chinese Academy of Sciences, No 9 Section 4, Renmin Nan Road, Chengdu, 610041, Sichuan, People's Republic of China
| | - Zeyu Xie
- Chengdu Institute of Biology, Chinese Academy of Sciences, No 9 Section 4, Renmin Nan Road, Chengdu, 610041, Sichuan, People's Republic of China
| | - Yaxiong Tang
- Chengdu Institute of Biology, Chinese Academy of Sciences, No 9 Section 4, Renmin Nan Road, Chengdu, 610041, Sichuan, People's Republic of China.
| |
Collapse
|
44
|
Ferrell B, Cullinane CA, Ervine K, Melancon C, Uman GC, Juarez G. Perspectives on the Impact of Ovarian Cancer: Women's Views of Quality of Life. Oncol Nurs Forum 2007; 32:1143-9. [PMID: 16270110 DOI: 10.1188/05.onf.1143-1149] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
PURPOSE/OBJECTIVES To describe quality-of-life (QOL) concerns particular to women with ovarian cancer and to examine whether subgroups of patients with ovarian cancer have significantly different QOL concerns. DESIGN Mailed survey. SAMPLE Readership of an ovarian cancer newsletter. METHODS A total of 1,383 surveys were received in response to the survey's inclusion in the November 2002 issue of Conversations! The International Newsletter for Those Fighting Ovarian Cancer, a monthly newsletter circulated to 3,300 women with ovarian cancer (response rate = 42%). Women were asked to complete the 45-item City of Hope QOL Ovarian Cancer Tool (QOL-OVCA) and a short demographic questionnaire. MAIN RESEARCH VARIABLES Patients' QOL-OVCA scores were compared across six independent variables, including disease status, age at diagnosis, stage at diagnosis, marital status, household income, and use of alternative therapy, controlling for survival time. FINDINGS Ovarian cancer survivors reported significant QOL concerns across dimensions of physical, psychological, social, and spiritual well-being. CONCLUSIONS Patients with ovarian cancer experience particular QOL concerns requiring support. Future research is needed to describe the needs of survivors in more diverse populations and to develop and test interventions that can address these QOL concerns. IMPLICATIONS FOR NURSING Women with ovarian cancer experience QOL concerns common to other patients with cancer and some that are particular to ovarian cancer. Nurses should assess for and aggressively address these QOL concerns.
Collapse
Affiliation(s)
- Betty Ferrell
- City of Hope National Medical Center in Duarte, CA, USA.
| | | | | | | | | | | |
Collapse
|