1
|
Wolters-Eisfeld G, Oliveira-Ferrer L. Glycan diversity in ovarian cancer: Unraveling the immune interplay and therapeutic prospects. Semin Immunopathol 2024; 46:16. [PMID: 39432076 PMCID: PMC11493797 DOI: 10.1007/s00281-024-01025-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Accepted: 09/12/2024] [Indexed: 10/22/2024]
Abstract
Ovarian cancer remains a formidable challenge in oncology due to its late-stage diagnosis and limited treatment options. Recent research has revealed the intricate interplay between glycan diversity and the immune microenvironment within ovarian tumors, shedding new light on potential therapeutic strategies. This review seeks to investigate the complex role of glycans in ovarian cancer and their impact on the immune response. Glycans, complex sugar molecules decorating cell surfaces and secreted proteins, have emerged as key regulators of immune surveillance in ovarian cancer. Aberrant glycosylation patterns can promote immune evasion by shielding tumor cells from immune recognition, enabling disease progression. Conversely, certain glycan structures can modulate the immune response, leading to either antitumor immunity or immune tolerance. Understanding the intricate relationship between glycan diversity and immune interactions in ovarian cancer holds promise for the development of innovative therapeutic approaches. Immunotherapies that target glycan-mediated immune evasion, such as glycan-based vaccines or checkpoint inhibitors, are under investigation. Additionally, glycan profiling may serve as a diagnostic tool for patient stratification and treatment selection. This review underscores the emerging importance of glycan diversity in ovarian cancer, emphasizing the potential for unraveling immune interplay and advancing tailored therapeutic prospects for this devastating disease.
Collapse
Affiliation(s)
- Gerrit Wolters-Eisfeld
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | | |
Collapse
|
2
|
Tu HF, Wong M, Tseng SH, Ingavat N, Olczak P, Notarte KI, Hung CF, Roden RBS. Virus-like particle vaccine displaying an external, membrane adjacent MUC16 epitope elicits ovarian cancer-reactive antibodies. J Ovarian Res 2024; 17:19. [PMID: 38225646 PMCID: PMC10790439 DOI: 10.1186/s13048-023-01325-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Accepted: 12/12/2023] [Indexed: 01/17/2024] Open
Abstract
BACKGROUND MUC16 is a heavily glycosylated cell surface mucin cleaved in the tumor microenvironment to shed CA125. CA125 is a serum biomarker expressed by > 95% of non-mucinous advanced stage epithelial ovarian cancers. MUC16/CA125 contributes to the evasion of anti-tumor immunity, peritoneal spread and promotes carcinogenesis; consequently, it has been targeted with antibody-based passive and active immunotherapy. However, vaccination against this self-antigen likely requires breaking B cell tolerance and may trigger autoimmune disease. Display of self-antigens on virus-like particles (VLPs), including those produced with human papillomavirus (HPV) L1, can efficiently break B cell tolerance. RESULTS A 20 aa juxta-membrane peptide of the murine MUC16 (mMUC16) or human MUC16 (hMUC16) ectodomain was displayed either via genetic insertion into an immunodominant loop of HPV16 L1-VLPs between residues 136/137, or by chemical coupling using malemide to cysteine sulfhydryl groups on their surface. Female mice were vaccinated intramuscularly three times with either DNA expressing L1-MUC16 fusions via electroporation, or with alum-formulated VLP chemically-coupled to MUC16 peptides. Both regimens were well tolerated, and elicited MUC16-specific serum IgG, although titers were higher in mice vaccinated with MUC16-coupled VLP on alum as compared to L1-MUC16 DNA vaccination. Antibody responses to mMUC16-targeted vaccination cross-reacted with hMUC16 peptide, and vice versa; both were reactive with the surface of CA125+ OVCAR3 cells, but not SKOV3 that lack detectable CA125 expression. Interestingly, vaccination of mice with mMUC16 peptide mixed with VLP and alum elicited mMUC16-specific IgG, implying VLPs provide robust T help and that coupling may not be required to break tolerance to this epitope. CONCLUSION Vaccination with VLP displaying the 20 aa juxta-membrane MUC16 ectodomain, which includes the membrane proximal cleavage site, is likely to be well tolerated and induce IgG targeting ovarian cancer cells, even after CA125 is shed.
Collapse
Affiliation(s)
- Hsin-Fang Tu
- Department of Pathology, Johns Hopkins University, Baltimore, MD, 21287, USA
| | - Margaret Wong
- Department of Pathology, Johns Hopkins University, Baltimore, MD, 21287, USA
| | - Ssu-Hsueh Tseng
- Department of Pathology, Johns Hopkins University, Baltimore, MD, 21287, USA
| | - Nattha Ingavat
- Downstream Processing (DSP), Bioprocessing Technology Institute (BTI), Agency for Science, Technology, and Research (A*STAR), Singapore, 138632, Singapore
| | - Pola Olczak
- Department of Pathology, Johns Hopkins University, Baltimore, MD, 21287, USA
| | - Kin Israel Notarte
- Department of Pathology, Johns Hopkins University, Baltimore, MD, 21287, USA
| | - Chien-Fu Hung
- Department of Pathology, Johns Hopkins University, Baltimore, MD, 21287, USA
- Department of Oncology, Johns Hopkins University, Baltimore, MD, 21287, USA
| | - Richard B S Roden
- Department of Pathology, Johns Hopkins University, Baltimore, MD, 21287, USA.
- Department of Oncology, Johns Hopkins University, Baltimore, MD, 21287, USA.
- Department of Gynecology and Obstetrics, Johns Hopkins University, Baltimore, MD, 21287, USA.
| |
Collapse
|
3
|
Guy H, Hawkes C, Walder L, Malinowska IA, Gupta D. Systematic literature review of efficacy and safety of first-line maintenance therapy trials in advanced ovarian cancer. Future Oncol 2022; 18:3435-3447. [DOI: 10.2217/fon-2022-0578] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Aim: To review safety and efficacy outcomes in studies of first-line maintenance therapies for advanced ovarian cancer. Methods: A systematic literature review was performed (27 February 2020) to identify clinical outcomes including progression-free survival (PFS), overall survival (OS) and Grade ≥3 adverse events. Results: Overall 50 references met prespecified criteria; 18 studies evaluated 10 different agents, including PARP inhibitors. PFS was an end point in 16 trials and OS in 12 trials. PARP inhibitors reported better PFS hazard ratios (HRs: 0.59–0.68) compared with other classes; no mature OS data were identified. Safety reporting was inconsistent. Conclusion: Reported PFS HRs were better for PARP inhibitors than for other ovarian cancer maintenance therapies; overall survival data remain immature.
Collapse
Affiliation(s)
- Holly Guy
- FIECON Ltd., 3 College Yard, Lower Dagnall St, St Albans, Hertfordshire, AL3 4PA, UK
| | - Carol Hawkes
- GlaxoSmithKline, 980 Great West Road, Brentford, TW8 9GS, UK
| | - Lydia Walder
- FIECON Ltd., 3 College Yard, Lower Dagnall St, St Albans, Hertfordshire, AL3 4PA, UK
| | | | - Divya Gupta
- GlaxoSmithKline, 1000 Winter St, Waltham, MA 02451, USA
| |
Collapse
|
4
|
Lorusso D, Guy H, Samyshkin Y, Hawkes C, Estenson K, Coleman RL. Feasibility Study of a Network Meta-Analysis and Unanchored Population-Adjusted Indirect Treatment Comparison of Niraparib, Olaparib, and Bevacizumab as Maintenance Therapies in Patients with Newly Diagnosed Advanced Ovarian Cancer. Cancers (Basel) 2022; 14:1285. [PMID: 35267593 PMCID: PMC8909094 DOI: 10.3390/cancers14051285] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 02/22/2022] [Accepted: 02/23/2022] [Indexed: 01/27/2023] Open
Abstract
Selecting a first-line (1L) maintenance option for ovarian cancer is challenging given the variety of therapies, differing trials, and the lack of head-to-head data for angiogenesis and poly(ADP-ribose) polymerase (PARP) inhibitors. Thus, indirect treatment comparisons (ITCs) can aid treatment decision making. This study assessed the feasibility of two ITCs, a network meta-analysis (NMA) and a population-adjusted ITC (PAIC), comparing the efficacy of the PARP inhibitor niraparib in the PRIMA trial (NCT02655016) with other 1L maintenance treatments. A systematic literature review was conducted to identify trials using the Cochrane Handbook for Systematic Reviews of Interventions to assess differences in trial design, population characteristics, treatment arms, and outcome measures. All 12 trials identified were excluded from the NMA due to the absence of a common comparator and differences in survival measures and/or inclusion criteria. The PAIC comparing PRIMA and PAOLA-1 trials was also not feasible due to differences in inclusion criteria, survival measures, and the previous receipt of chemotherapy/bevacizumab. Neither ITC met recommended guidelines for analysis; the results of such comparisons would not be considered appropriate evidence when selecting 1L maintenance options in ovarian cancer. ITCs in this setting should be performed cautiously, as many factors can preclude objective trial comparisons.
Collapse
Affiliation(s)
- Domenica Lorusso
- Fondazione Policlinico Gemelli of Rome, 00168 Rome, Italy
- Department of Gynecologic Oncology, Catholic University of Sacred Heart, 00168 Rome, Italy
| | | | | | - Carol Hawkes
- GlaxoSmithKline, Brentford TW8 9GS, UK; (Y.S.); (C.H.)
| | - Kasey Estenson
- GlaxoSmithKline, Philadelphia, PA 19112, USA;
- Eisai Inc., Nutley, NJ 07677, USA
| | | |
Collapse
|
5
|
Xu X, Gan M, Ge Y, Yi C, Feng T, Liu M, Wu C, Chen X, Zhang W, Zhao L, Zou J. Multifaceted glycoadjuvant@AuNPs inhibits tumor metastasis through promoting T cell activation and remodeling tumor microenvironment. J Nanobiotechnology 2021; 19:376. [PMID: 34794428 PMCID: PMC8600715 DOI: 10.1186/s12951-021-01129-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Accepted: 11/08/2021] [Indexed: 11/17/2022] Open
Abstract
ABSTARCT BACKGROUND: Cytosine-phosphate-guanine (CpG) dinucleotides has been used as adjuvants for cancer immunotherapy. However, unmodified CpG are not very efficient in clinical trials. Glucose, ligand of C-type lectin receptors (CLRs), can promote DC maturation and antigen presentation, which is the first step of induction of adaptive immune responses. Therefore, conjugation of type B CpG DNA to glucose-containing glycopolymers may enhance the therapeutic effects against tumor by CpG-based vaccine. METHODS gCpG was developed by chemical conjugation of type B CpG DNA to glucose-containing glycopolymers. The therapeutic effects of gCpG-based vaccine were tested in both murine primary melanoma model and its metastasis model. RESULTS gCpG based tumor vaccine inhibited both primary and metastasis of melanoma in mice which was dependent on CD8 + T cells and IFNγ. In tumor microenvironment, gCpG treatment increased Th1 and CTL infiltration, increased M1 macrophages, decreased Tregs and MDSCs populations, and promoted inflammatory milieu with enhanced secretion of IFNγ and TNFα. The anti-tumor efficacy of gCpG was dramatically enhanced when combined with anti-PD1 immunotherapy. CONCLUSIONS We confirmed that gCpG was a promising adjuvant for vaccine formulation by activating both tumor-specific Th1 and Tc1 responses, and regulating tumor microenvironments.
Collapse
Affiliation(s)
- Xiaojing Xu
- College of Basic Medicine and Biological Sciences, Medical Department, Soochow University, 215123, Suzhou, People's Republic of China
| | - Minfeng Gan
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, 215006, Jiangsu, China
| | - Youzhen Ge
- College of Basic Medicine and Biological Sciences, Medical Department, Soochow University, 215123, Suzhou, People's Republic of China
- Institute of Blood and Marrow Transplantation, Department of Hematology, Collaborative Innovation Center of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, People's Republic of China
| | - Cheng Yi
- College of Basic Medicine and Biological Sciences, Medical Department, Soochow University, 215123, Suzhou, People's Republic of China
- Institute of Blood and Marrow Transplantation, Department of Hematology, Collaborative Innovation Center of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, People's Republic of China
| | - Tianyun Feng
- College of Basic Medicine and Biological Sciences, Medical Department, Soochow University, 215123, Suzhou, People's Republic of China
- Institute of Blood and Marrow Transplantation, Department of Hematology, Collaborative Innovation Center of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, People's Republic of China
| | - Mengjie Liu
- Center for Soft Condensed Matter Physics and Interdisciplinary Research, Soochow University, Suzhou, 215006, People's Republic of China
| | - Cenhao Wu
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, 215006, Jiangsu, China
| | - Xiang Chen
- Jiangsu Key Lab of Zoonosis/Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, China
| | - Weidong Zhang
- Center for Soft Condensed Matter Physics and Interdisciplinary Research, Soochow University, Suzhou, 215006, People's Republic of China.
| | - Lixiang Zhao
- College of Basic Medicine and Biological Sciences, Medical Department, Soochow University, 215123, Suzhou, People's Republic of China.
| | - Jun Zou
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, 215006, Jiangsu, China.
| |
Collapse
|
6
|
Lee DH, Choi S, Park Y, Jin HS. Mucin1 and Mucin16: Therapeutic Targets for Cancer Therapy. Pharmaceuticals (Basel) 2021; 14:ph14101053. [PMID: 34681277 PMCID: PMC8537522 DOI: 10.3390/ph14101053] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 10/13/2021] [Accepted: 10/14/2021] [Indexed: 01/18/2023] Open
Abstract
The mucin (MUC) family is a group of highly glycosylated macromolecules that are abundantly expressed in mammalian epithelial cells. MUC proteins contribute to the formation of the mucus barrier and thus have protective functions against infection. Interestingly, some MUC proteins are aberrantly expressed in cancer cells and are involved in cancer development and progression, including cell growth, proliferation, the inhibition of apoptosis, chemoresistance, metabolic reprogramming, and immune evasion. With their unique biological and structural features, MUC proteins have been considered promising therapeutic targets and also biomarkers for human cancer. In this review, we discuss the biological roles of the transmembrane mucins MUC1 and MUC16 in the context of hallmarks of cancer and current efforts to develop MUC1- and MUC16-targeted therapies.
Collapse
Affiliation(s)
- Dong-Hee Lee
- Department of Convergence Medicine, Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Korea;
| | - Seunghyun Choi
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Korea;
| | - Yoon Park
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Korea;
- Correspondence: (Y.P.); (H.-s.J.)
| | - Hyung-seung Jin
- Department of Convergence Medicine, Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Korea;
- Correspondence: (Y.P.); (H.-s.J.)
| |
Collapse
|
7
|
Smith BAH, Bertozzi CR. The clinical impact of glycobiology: targeting selectins, Siglecs and mammalian glycans. Nat Rev Drug Discov 2021; 20:217-243. [PMID: 33462432 PMCID: PMC7812346 DOI: 10.1038/s41573-020-00093-1] [Citation(s) in RCA: 300] [Impact Index Per Article: 75.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/29/2020] [Indexed: 01/31/2023]
Abstract
Carbohydrates - namely glycans - decorate every cell in the human body and most secreted proteins. Advances in genomics, glycoproteomics and tools from chemical biology have made glycobiology more tractable and understandable. Dysregulated glycosylation plays a major role in disease processes from immune evasion to cognition, sparking research that aims to target glycans for therapeutic benefit. The field is now poised for a boom in drug development. As a harbinger of this activity, glycobiology has already produced several drugs that have improved human health or are currently being translated to the clinic. Focusing on three areas - selectins, Siglecs and glycan-targeted antibodies - this Review aims to tell the stories behind therapies inspired by glycans and to outline how the lessons learned from these approaches are paving the way for future glycobiology-focused therapeutics.
Collapse
Affiliation(s)
- Benjamin A H Smith
- Department of Chemical & Systems Biology and ChEM-H, Stanford School of Medicine, Stanford, CA, USA
| | - Carolyn R Bertozzi
- Department of Chemical & Systems Biology and ChEM-H, Stanford School of Medicine, Stanford, CA, USA.
- Department of Chemistry, Stanford University, Stanford, CA, USA.
- Howard Hughes Medical Institute, Stanford University, Stanford, CA, USA.
| |
Collapse
|
8
|
Zhang M, Cheng S, Jin Y, Zhao Y, Wang Y. Roles of CA125 in diagnosis, prediction, and oncogenesis of ovarian cancer. Biochim Biophys Acta Rev Cancer 2021; 1875:188503. [PMID: 33421585 DOI: 10.1016/j.bbcan.2021.188503] [Citation(s) in RCA: 171] [Impact Index Per Article: 42.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 01/03/2021] [Accepted: 01/04/2021] [Indexed: 12/12/2022]
Abstract
After it was discovered approximately 40 years ago, carbohydrate antigen 125 (CA125) became the most widely used and concerning biomarker in ovarian cancer screening. However, there is still controversy about its role in clinical practice. CA125 is not sufficiently reliable in diagnosis to screen for early-stage ovarian cancer. On the other hand, CA125 has been a valuable indicator for evaluating chemotherapeutic efficacy and prognosis. We still do not know much about its biological role, and several studies have indicated that this marker participates in the occurrence and development of ovarian cancer. Currently, an increasing number of scholars have begun to pay attention to CA125-targeted treatment strategies. In the interest of better design and development of anticancer therapies, a renewed and systematic understanding of the roles of CA125 in diagnosis, prediction, and tumorigenesis is warranted.
Collapse
Affiliation(s)
- Minghai Zhang
- Department of Obstetrics and Gynecology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200127, China
| | - Shanshan Cheng
- Department of Obstetrics and Gynecology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200127, China
| | - Yue Jin
- Department of Obstetrics and Gynecology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200127, China
| | - Yaqian Zhao
- Department of Obstetrics and Gynecology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200127, China
| | - Yu Wang
- Department of Obstetrics and Gynecology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200127, China; Shanghai Key Laboratory of Gynecologic Oncology, Shanghai 200127, China.
| |
Collapse
|
9
|
Palaia I, Tomao F, Sassu CM, Musacchio L, Benedetti Panici P. Immunotherapy For Ovarian Cancer: Recent Advances And Combination Therapeutic Approaches. Onco Targets Ther 2020; 13:6109-6129. [PMID: 32617007 PMCID: PMC7326187 DOI: 10.2147/ott.s205950] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Accepted: 05/20/2020] [Indexed: 01/05/2023] Open
Abstract
Epithelial ovarian cancer (EOC) is the most lethal gynaecological cancer. Although many advances have been made in therapeutic strategies, the global standard of care still remains radical surgery plus chemotherapy, but new scenarios need to be explored to improve survival. The role of immunotherapy in EOC treatment is controversial. Results obtained from studies evaluating immunotherapy are contradictory: in particular data on survival are not as good as expected when immunotherapy was administered alone, and other data are still immature. Thus, significant efforts must be devoted to finding new strategies for the use of immunotherapy. The aim of this paper is to review the most recent findings of the use of immunotherapy in ovarian cancer, with a particular focus on combination approaches.
Collapse
Affiliation(s)
- Innocenza Palaia
- Department of Maternal and Child Health and Urological Sciences, “Sapienza” University of Rome, Policlinico Umberto I, Rome00161, Italy
| | - Federica Tomao
- Department of Maternal and Child Health and Urological Sciences, “Sapienza” University of Rome, Policlinico Umberto I, Rome00161, Italy
| | - Carolina Maria Sassu
- Department of Maternal and Child Health and Urological Sciences, “Sapienza” University of Rome, Policlinico Umberto I, Rome00161, Italy
| | - Lucia Musacchio
- Department of Maternal and Child Health and Urological Sciences, “Sapienza” University of Rome, Policlinico Umberto I, Rome00161, Italy
| | - Pierluigi Benedetti Panici
- Department of Maternal and Child Health and Urological Sciences, “Sapienza” University of Rome, Policlinico Umberto I, Rome00161, Italy
| |
Collapse
|
10
|
Battaglia A, Buzzonetti A, Fossati M, Scambia G, Fattorossi A, Madiyalakan MR, Mahnke YD, Nicodemus C. Translational immune correlates of indirect antibody immunization in a randomized phase II study using scheduled combination therapy with carboplatin/paclitaxel plus oregovomab in ovarian cancer patients. Cancer Immunol Immunother 2020; 69:383-397. [PMID: 31897661 DOI: 10.1007/s00262-019-02456-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Accepted: 12/13/2019] [Indexed: 12/12/2022]
Abstract
The standard-of-care (SOC) first-line therapy for ovarian cancer (OC) patients is plagued with high relapse rates. Several studies indicated the immune system's prominent role changing the disease course in OC patients. Chemo-immunotherapy regimens, currently being explored, include oregovomab, which is a monoclonal antibody specific for the OC associated antigen carbohydrate/cancer antigen 125 (CA125) that yielded promising results when administered together with SOC in a previous study. The QPT-ORE-002 multi-site phase II randomized study demonstrated that in patients with advanced OC, oregovomab combined with first-line SOC improved overall and progression-free survival, compared to SOC alone. The study included an Italian cohort in which we demonstrated that adding oregovomab to SOC resulted in increased patient numbers with amplified CA125-specific CD8+T lymphocytes/ml peripheral blood counts, which might explain the improved therapeutic effect of SOC + oregovomab over SOC alone. Predictive for oregovomab efficacy was a less suppressive immune environment at baseline as indicated by low numbers of circulating myeloid-derived suppressor cells, subset type 4, and a low neutrophil-and-monocyte to lymphocyte ratio.
Collapse
Affiliation(s)
- Alessandra Battaglia
- Istituto di Clinica Ostetrica e Ginecologica, Università Cattolica del Sacro Cuore, L.go F.Vito 1, 00168, Rome, Italy.
| | - Alexia Buzzonetti
- Department of Woman and Child Health and Public Health, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Marco Fossati
- Department of Woman and Child Health and Public Health, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Giovanni Scambia
- Istituto di Clinica Ostetrica e Ginecologica, Università Cattolica del Sacro Cuore, L.go F.Vito 1, 00168, Rome, Italy.,Department of Woman and Child Health and Public Health, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Andrea Fattorossi
- Department of Woman and Child Health and Public Health, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | | | | | | |
Collapse
|
11
|
Want MY, Konstorum A, Huang RY, Jain V, Matsueda S, Tsuji T, Lugade A, Odunsi K, Koya R, Battaglia S. Neoantigens retention in patient derived xenograft models mediates autologous T cells activation in ovarian cancer. Oncoimmunology 2019; 8:e1586042. [PMID: 31069153 PMCID: PMC6492964 DOI: 10.1080/2162402x.2019.1586042] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Revised: 01/17/2019] [Accepted: 02/15/2019] [Indexed: 12/12/2022] Open
Abstract
Ovarian cancer (OC) has an overall modest number of mutations that facilitate a functional immune infiltrate able to recognize tumor mutated antigens, or neoantigens. Although patient-derived xenografts (PDXs) can partially model the tumor mutational load and mimic response to chemotherapy, no study profiled a neoantigen-driven response in OC PDXs. Here we demonstrate that the genomic status of the primary tumor from an OC patient can be recapitulated in vivo in a PDX model, with the goal of defining autologous T cells activation by neoantigens using in silico, in vitro and in vivo approaches. By profiling the PDX mutanome we discovered three main clusters of mutations defining the expansion, retraction or conservation of tumor clones based on their variant allele frequencies (VAF). RNASeq analyses revealed a strong functional conservation between the primary tumor and PDXs, highlighted by the upregulation of antigen presenting pathways. We tested in vitro a set of 30 neoantigens for recognition by autologous T cells and identified a core of six neoantigens that define a potent T cell activation able to slow tumor growth in vivo. The pattern of recognition of these six neoantigens indicates the pre-existence of anti-tumor immunity in the patient. To evaluate the breadth of T cell activation, we performed single cell sequencing profiling the TCR repertoire upon stimulation with neoantigenic moieties and identified sequence motifs that define an oligoclonal and autologous T cell response. Overall, these results indicate that OC PDXs can be a valid tool to model OC response to immunotherapy.
Collapse
Affiliation(s)
| | - Anna Konstorum
- Center for Quantitative Medicine, UConn Health, Farmington, CT, USA
| | - Ruea-Yea Huang
- Center For Immunotherapy, Comprehensive Cancer Center, Buffalo, NY, USA
| | - Vaibhav Jain
- Center For Immunotherapy, Comprehensive Cancer Center, Buffalo, NY, USA
| | - Satoko Matsueda
- Center For Immunotherapy, Comprehensive Cancer Center, Buffalo, NY, USA
| | - Takemasa Tsuji
- Center For Immunotherapy, Comprehensive Cancer Center, Buffalo, NY, USA
| | - Amit Lugade
- Center For Immunotherapy, Comprehensive Cancer Center, Buffalo, NY, USA
| | - Kunle Odunsi
- Center For Immunotherapy, Comprehensive Cancer Center, Buffalo, NY, USA
| | - Richard Koya
- Center For Immunotherapy, Comprehensive Cancer Center, Buffalo, NY, USA
| | - Sebastiano Battaglia
- Center For Immunotherapy, Comprehensive Cancer Center, Buffalo, NY, USA.,Department of Cancer Genetics and Genomics, Roswell Park, Comprehensive Cancer Center, Buffalo, NY, USA
| |
Collapse
|
12
|
Paijens ST, Leffers N, Daemen T, Helfrich W, Boezen HM, Cohlen BJ, Melief CJM, de Bruyn M, Nijman HW, Cochrane Gynaecological, Neuro‐oncology and Orphan Cancer Group. Antigen-specific active immunotherapy for ovarian cancer. Cochrane Database Syst Rev 2018; 9:CD007287. [PMID: 30199097 PMCID: PMC6513204 DOI: 10.1002/14651858.cd007287.pub4] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
BACKGROUND This is the second update of the review first published in the Cochrane Library (2010, Issue 2) and later updated (2014, Issue 9).Despite advances in chemotherapy, the prognosis of ovarian cancer remains poor. Antigen-specific active immunotherapy aims to induce tumour antigen-specific anti-tumour immune responses as an alternative treatment for ovarian cancer. OBJECTIVES Primary objective• To assess the clinical efficacy of antigen-specific active immunotherapy for the treatment of ovarian cancer as evaluated by tumour response measured by Response Evaluation Criteria In Solid Tumors (RECIST) and/or cancer antigen (CA)-125 levels, response to post-immunotherapy treatment, and survival differences◦ In addition, we recorded the numbers of observed antigen-specific humoral and cellular responsesSecondary objective• To establish which combinations of immunotherapeutic strategies with tumour antigens provide the best immunological and clinical results SEARCH METHODS: For the previous version of this review, we performed a systematic search of the Cochrane Central Register of Controlled Trials (CENTRAL; 2009, Issue 3), in the Cochrane Library, the Cochrane Gynaecological Cancer Group Specialised Register, MEDLINE and Embase databases, and clinicaltrials.gov (1966 to July 2009). We also conducted handsearches of the proceedings of relevant annual meetings (1996 to July 2009).For the first update of this review, we extended the searches to October 2013, and for this update, we extended the searches to July 2017. SELECTION CRITERIA We searched for randomised controlled trials (RCTs), as well as non-randomised studies (NRSs), that included participants with epithelial ovarian cancer, irrespective of disease stage, who were treated with antigen-specific active immunotherapy, irrespective of type of vaccine, antigen used, adjuvant used, route of vaccination, treatment schedule, and reported clinical or immunological outcomes. DATA COLLECTION AND ANALYSIS Two reviews authors independently extracted the data. We evaluated the risk of bias for RCTs according to standard methodological procedures expected by Cochrane, and for NRSs by using a selection of quality domains deemed best applicable to the NRS. MAIN RESULTS We included 67 studies (representing 3632 women with epithelial ovarian cancer). The most striking observations of this review address the lack of uniformity in conduct and reporting of early-phase immunotherapy studies. Response definitions show substantial variation between trials, which makes comparison of trial results unreliable. Information on adverse events is frequently limited. Furthermore, reports of both RCTs and NRSs frequently lack the relevant information necessary for risk of bias assessment. Therefore, we cannot rule out serious biases in most of the included trials. However, selection, attrition, and selective reporting biases are likely to have affected the studies included in this review. GRADE ratings were high only for survival; for other primary outcomes, GRADE ratings were very low.The largest body of evidence is currently available for CA-125-targeted antibody therapy (17 studies, 2347 participants; very low-certainty evidence). Non-randomised studies of CA-125-targeted antibody therapy suggest improved survival among humoral and/or cellular responders, with only moderate adverse events. However, four large randomised placebo-controlled trials did not show any clinical benefit, despite induction of immune responses in approximately 60% of participants. Time to relapse with CA-125 monoclonal antibody versus placebo, respectively, ranged from 10.3 to 18.9 months versus 10.3 to 13 months (six RCTs, 1882 participants; high-certainty evidence). Only one RCT provided data on overall survival, reporting rates of 80% in both treatment and placebo groups (three RCTs, 1062 participants; high-certainty evidence). Other small studies targeting many different tumour antigens have presented promising immunological results. As these strategies have not yet been tested in RCTs, no reliable inferences about clinical efficacy can be made. Given the promising immunological results and the limited side effects and toxicity reported, exploration of clinical efficacy in large well-designed RCTs may be worthwhile. AUTHORS' CONCLUSIONS We conclude that despite promising immunological responses, no clinically effective antigen-specific active immunotherapy is yet available for ovarian cancer. Results should be interpreted cautiously, as review authors found a significant dearth of relevant information for assessment of risk of bias in both RCTs and NRSs.
Collapse
Affiliation(s)
- Sterre T Paijens
- University Medical Center Groningen (UMCG)Obstetrics & GynaecologyGroningenNetherlands9713 GZ
| | - Ninke Leffers
- University Medical Center Groningen (UMCG)Obstetrics & GynaecologyGroningenNetherlands9713 GZ
| | - Toos Daemen
- University Medical Center Groningen (UMCG)GroningenNetherlands9713 GZ
| | - Wijnand Helfrich
- University Medical Center Groningen (UMCG)Department of Surgery. Translational Surgical OncologyGroningenNetherlands9713 GZ
| | - H Marike Boezen
- University Medical Center Groningen (UMCG)Unit Chronic Airway Diseases, Department of EpidemiologyGroningenNetherlands9713 GZ
| | - Ben J Cohlen
- Isala Clinics, Location SophiaDepartment of Obstetrics & GynaecologyDr van Heesweg 2P O Box 10400ZwolleNetherlands3515 BE
| | - Cornelis JM Melief
- Leiden University Medical CenterDepartment of Immunohaematology and Blood TransfusionPO Box 9600E3‐QLeidenNetherlands2300 RC
| | - Marco de Bruyn
- University Medical Center Groningen (UMCG)Obstetrics & GynaecologyGroningenNetherlands9713 GZ
| | - Hans W Nijman
- University Medical Center Groningen (UMCG)GroningenNetherlands9713 GZ
| | | |
Collapse
|
13
|
Aithal A, Rauth S, Kshirsagar P, Shah A, Lakshmanan I, Junker WM, Jain M, Ponnusamy MP, Batra SK. MUC16 as a novel target for cancer therapy. Expert Opin Ther Targets 2018; 22:675-686. [PMID: 29999426 PMCID: PMC6300140 DOI: 10.1080/14728222.2018.1498845] [Citation(s) in RCA: 155] [Impact Index Per Article: 22.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
INTRODUCTION MUC16 is overexpressed in multiple cancers and plays an important role in tumorigenicity and acquired resistance to therapy. Area covered: In this review, we describe the role of MUC16 under normal physiological conditions and during tumorigenesis. First, we provide a summary of research on MUC16 from its discovery as CA125 to present anti-MUC16 therapy trials that are currently in the initial phases of clinical testing. Finally, we discuss the reasons for the limited effectiveness of these therapies and discuss the direction and focus of future research. Expert opinion: Apart from its protective role in normal physiology, MUC16 contributes to disease progression and metastasis in several malignancies. Due to its aberrant overexpression, it is a promising target for diagnosis and therapy. Cleavage and shedding of its extracellular domain is the major barrier for efficient targeting of MUC16-expressing cancers. Concerted efforts should be undertaken to target the noncleaved cell surface retained portion of MUC16. Such efforts should be accompanied by basic research to understand MUC16 cleavage and decipher the functioning of MUC16 cytoplasmic tail. While previous efforts to activate anti-MUC16 immune response using anti-CA125 idiotype antibodies have met with limited success, ideification of neo-antigenic epitopes in MUC16 that correlate with improved survival have raised raised hopes for developing MUC16-targeted immunotherapy.
Collapse
Affiliation(s)
- Abhijit Aithal
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, United States of America
| | - Sanchita Rauth
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, United States of America
| | - Prakash Kshirsagar
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, United States of America
| | - Ashu Shah
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, United States of America
| | - Imayavaramban Lakshmanan
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, United States of America
| | - Wade M. Junker
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, United States of America
| | - Maneesh Jain
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, United States of America
- Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, United States of America
| | - Moorthy P. Ponnusamy
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, United States of America
- Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, United States of America
| | - Surinder K. Batra
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, United States of America
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, United States of America
- Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, United States of America
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE, United States of America
| |
Collapse
|
14
|
A robust immune system conditions the response to abagovomab (anti-idiotypic monoclonal antibody mimicking the CA125 protein) vaccination in ovarian cancer patients. Immunol Lett 2017; 191:35-39. [PMID: 28919454 DOI: 10.1016/j.imlet.2017.09.006] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Revised: 09/13/2017] [Accepted: 09/13/2017] [Indexed: 11/21/2022]
Abstract
INTRODUCTION Despite encouraging phase I and II study results, vaccination of ovarian cancer patients with abagovomab - an anti-idiotypic mAb that mimics the ovarian cancer CA125 protein - failed to demonstrate efficacy in the phase III trial named MIMOSA (NCT00418574). We postulated that in this trial patients with a more robust immune system did respond to abagovomab but went undetected among a larger number of non-responders. We also postulated that assessment of the immune system status ahead of abagovomab administration might predict patients' propensity to respond to abagovomab. MATERIALS AND METHODS The immune system status was assessed as percentage and absolute count of CD8+ T cells producing IFN-γ after stimulation with Staphylococcal Enterotoxin B (SEB) in 80 patients on abagovomab and 31 patients on placebo from the MIMOSA trial ahead of treatment. Optimal cutoffs of the two variables were calculated by the web application "Cutoff Finder" as the points with most significant (log-rank test) splits based on relapse-free survival (RFS). The Kaplan-Meier curves and log-rank test served to estimate and compare RFS in patients with percentage and absolute count of IFN-γ producing CD8+ T cells around the cutoffs. RESULTS Patients on abagovomab with IFN-γ producing CD8+T cell percentage above the cutoff had a better RFS (p=0.042) than those with IFN-γ producing CD8+T cell percentage below the cutoff. Patients on abagovomab with IFN-γ producing CD8+T cell absolute count above the cutoff had a better RFS (p=0.019) than those with IFN-γ producing CD8+T cell absolute counts below the cutoff. Consistently, the RFS of patients on abagovomab with IFN-γ producing CD8+T cell percentage and absolute counts values below the respective cutoffs was identical to that of patients on placebo. Neither the percentage nor the absolute count of IFN-γ producing CD8+T cells correlated with RFS in patients on placebo. CONCLUSIONS A robust immune system is essential to obtain a clinical response in OC patients undergoing abagovomab immunotherapy whereas a robust immune system does not confer per se a survival advantage. Further work will clarify whether the results shown here apply only in the present setting or extend to other types of cancer and/or immunotherapeutic agents.
Collapse
|
15
|
Aalipour S, Zoghi S, Khalili N, Hirbod-Mobarakeh A, Emens LA, Rezaei N. Specific immunotherapy in ovarian cancer: a systematic review. Immunotherapy 2017; 8:1193-204. [PMID: 27605068 DOI: 10.2217/imt-2016-0034] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Epithelial ovarian cancer (EOC) is the most lethal gynecological cancer. Several approaches of active and passive immunotherapy for EOC have been studied. The aim of this systematic review was to assess the clinical efficacy of specific immunotherapy in patients with EOC. We found 4524 references in seven databases and we included ten controlled clinical trials with 2285 patients with EOC reporting five active immunotherapeutic agents and three passive immunotherapies. Meta-analysis of six studies showed that overall there was not any significant difference in overall survival and recurrence-free survival between patients undergoing specific immunotherapy and those in control group. Most of the studies we evaluated reported a positive outcome from treatment with specific immunotherapy, although this was not significant.
Collapse
Affiliation(s)
- Soroush Aalipour
- Border of Immune Tolerance Education & Research Network (BITERN), Universal Scientific Education & Research Network (USERN), Tehran, Iran
| | - Samaneh Zoghi
- Border of Immune Tolerance Education & Research Network (BITERN), Universal Scientific Education & Research Network (USERN), Tehran, Iran
| | - Nastaran Khalili
- Border of Immune Tolerance Education & Research Network (BITERN), Universal Scientific Education & Research Network (USERN), Tehran, Iran
| | - Armin Hirbod-Mobarakeh
- Border of Immune Tolerance Education & Research Network (BITERN), Universal Scientific Education & Research Network (USERN), Tehran, Iran.,Molecular Immunology Research Center; and Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.,Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Leisha A Emens
- Department of Oncology, Kimmel Cancer Center at Johns Hopkins, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Nima Rezaei
- Molecular Immunology Research Center; and Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.,Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran.,Systematic Review and Meta-Analysis Expert Group (SRMEG), Universal Scientific Education and Research Network (USERN), Boston, MA, USA
| |
Collapse
|
16
|
Wurz GT, Kao CJ, DeGregorio MW. Novel cancer antigens for personalized immunotherapies: latest evidence and clinical potential. Ther Adv Med Oncol 2016; 8:4-31. [PMID: 26753003 DOI: 10.1177/1758834015615514] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The clinical success of monoclonal antibody immune checkpoint modulators such as ipilimumab, which targets cytotoxic T lymphocyte-associated antigen 4 (CTLA-4), and the recently approved agents nivolumab and pembrolizumab, which target programmed cell death receptor 1 (PD-1), has stimulated renewed enthusiasm for anticancer immunotherapy, which was heralded by Science as 'Breakthrough of the Year' in 2013. As the potential of cancer immunotherapy has been recognized since the 1890s when William Coley showed that bacterial products could be beneficial in cancer patients, leveraging the immune system in the treatment of cancer is certainly not a new concept; however, earlier attempts to develop effective therapeutic vaccines and antibodies against solid tumors, for example, melanoma, frequently met with failure due in part to self-tolerance and the development of an immunosuppressive tumor microenvironment. Increased knowledge of the mechanisms through which cancer evades the immune system and the identification of tumor-associated antigens (TAAs) and negative immune checkpoint regulators have led to the development of vaccines and monoclonal antibodies targeting specific tumor antigens and immune checkpoints such as CTLA-4 and PD-1. This review first discusses the established targets of currently approved cancer immunotherapies and then focuses on investigational cancer antigens and their clinical potential. Because of the highly heterogeneous nature of tumors, effective anticancer immunotherapy-based treatment regimens will likely require a personalized combination of therapeutic vaccines, antibodies and chemotherapy that fit the specific biology of a patient's disease.
Collapse
Affiliation(s)
- Gregory T Wurz
- Department of Internal Medicine, Division of Hematology and Oncology, University of California, Davis, Sacramento, CA, USA
| | - Chiao-Jung Kao
- Department of Obstetrics and Gynecology, University of California, Davis Sacramento, CA, USA
| | - Michael W DeGregorio
- Department of Internal Medicine, Division of Hematology and Oncology, University of California, Davis, 4501 X Street Suite 3016, Sacramento, CA 95817, USA
| |
Collapse
|
17
|
Noujaim J, Thway K, Bajwa Z, Bajwa A, Maki RG, Jones RL, Keller C. Epithelioid Sarcoma: Opportunities for Biology-Driven Targeted Therapy. Front Oncol 2015; 5:186. [PMID: 26347853 PMCID: PMC4538302 DOI: 10.3389/fonc.2015.00186] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2015] [Accepted: 08/03/2015] [Indexed: 12/31/2022] Open
Abstract
Epithelioid sarcoma (ES) is a soft tissue sarcoma of children and young adults for which the preferred treatment for localized disease is wide surgical resection. Medical management is to a great extent undefined, and therefore for patients with regional and distal metastases, the development of targeted therapies is greatly desired. In this review, we will summarize clinically relevant biomarkers (e.g., SMARCB1, CA125, dysadherin, and others) with respect to targeted therapeutic opportunities. We will also examine the role of EGFR, mTOR, and polykinase inhibitors (e.g., sunitinib) in the management of local and disseminated disease. Toward building a consortium of pharmaceutical, academic, and non-profit collaborators, we will discuss the state of resources for investigating ES with respect to cell line resources, tissue banks, and registries so that a roadmap can be developed toward effective biology-driven therapies.
Collapse
Affiliation(s)
| | | | - Zia Bajwa
- Children's Cancer Therapy Development Institute , Fort Collins, CO , USA
| | - Ayeza Bajwa
- Children's Cancer Therapy Development Institute , Fort Collins, CO , USA
| | - Robert G Maki
- Adult and Paediatric Sarcoma Program, Tisch Cancer Institute, Mount Sinai School of Medicine , New York, NY , USA
| | | | - Charles Keller
- Children's Cancer Therapy Development Institute , Fort Collins, CO , USA
| |
Collapse
|