101
|
O’Brien RM, Cannon A, Reynolds JV, Lysaght J, Lynam-Lennon N. Complement in Tumourigenesis and the Response to Cancer Therapy. Cancers (Basel) 2021; 13:1209. [PMID: 33802004 PMCID: PMC7998562 DOI: 10.3390/cancers13061209] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Revised: 03/04/2021] [Accepted: 03/05/2021] [Indexed: 12/16/2022] Open
Abstract
In recent years, our knowledge of the complement system beyond innate immunity has progressed significantly. A modern understanding is that the complement system has a multifaceted role in malignancy, impacting carcinogenesis, the acquisition of a metastatic phenotype and response to therapies. The ability of local immune cells to produce and respond to complement components has provided valuable insights into their regulation, and the subsequent remodeling of the tumour microenvironment. These novel discoveries have advanced our understanding of the immunosuppressive mechanisms supporting tumour growth and uncovered potential therapeutic targets. This review discusses the current understanding of complement in cancer, outlining both direct and immune cell-mediated roles. The role of complement in response to therapies such as chemotherapy, radiation and immunotherapy is also presented. While complement activities are largely context and cancer type-dependent, it is evident that promising therapeutic avenues have been identified, in particular in combination therapies.
Collapse
Affiliation(s)
- Rebecca M. O’Brien
- Department of Surgery, Trinity St. James’s Cancer Institute, Trinity Translational Medicine Institute, Trinity College Dublin and St. James’s Hospital, Dublin 8, Ireland; (R.M.O.); (A.C.); (J.V.R.); (J.L.)
- Cancer Immunology and Immunotherapy Group, Trinity St. James’s Cancer Institute, Trinity Translational Medicine Institute, Trinity College Dublin and St. James’s Hospital, Dublin 8, Ireland
| | - Aoife Cannon
- Department of Surgery, Trinity St. James’s Cancer Institute, Trinity Translational Medicine Institute, Trinity College Dublin and St. James’s Hospital, Dublin 8, Ireland; (R.M.O.); (A.C.); (J.V.R.); (J.L.)
| | - John V. Reynolds
- Department of Surgery, Trinity St. James’s Cancer Institute, Trinity Translational Medicine Institute, Trinity College Dublin and St. James’s Hospital, Dublin 8, Ireland; (R.M.O.); (A.C.); (J.V.R.); (J.L.)
| | - Joanne Lysaght
- Department of Surgery, Trinity St. James’s Cancer Institute, Trinity Translational Medicine Institute, Trinity College Dublin and St. James’s Hospital, Dublin 8, Ireland; (R.M.O.); (A.C.); (J.V.R.); (J.L.)
- Cancer Immunology and Immunotherapy Group, Trinity St. James’s Cancer Institute, Trinity Translational Medicine Institute, Trinity College Dublin and St. James’s Hospital, Dublin 8, Ireland
| | - Niamh Lynam-Lennon
- Department of Surgery, Trinity St. James’s Cancer Institute, Trinity Translational Medicine Institute, Trinity College Dublin and St. James’s Hospital, Dublin 8, Ireland; (R.M.O.); (A.C.); (J.V.R.); (J.L.)
| |
Collapse
|
102
|
Redin E, Garmendia I, Lozano T, Serrano D, Senent Y, Redrado M, Villalba M, De Andrea CE, Exposito F, Ajona D, Ortiz-Espinosa S, Remirez A, Bertolo C, Sainz C, Garcia-Pedrero J, Pio R, Lasarte J, Agorreta J, Montuenga LM, Calvo A. SRC family kinase (SFK) inhibitor dasatinib improves the antitumor activity of anti-PD-1 in NSCLC models by inhibiting Treg cell conversion and proliferation. J Immunother Cancer 2021; 9:jitc-2020-001496. [PMID: 33658304 PMCID: PMC7931761 DOI: 10.1136/jitc-2020-001496] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/12/2021] [Indexed: 12/22/2022] Open
Abstract
INTRODUCTION The use of immune-checkpoint inhibitors has drastically improved the management of patients with non-small cell lung cancer (NSCLC), but innate and acquired resistances are hurdles needed to be solved. Immunomodulatory drugs that can reinvigorate the immune cytotoxic activity, in combination with antiprogrammed cell death 1 (PD-1) antibody, are a great promise to overcome resistance. We evaluated the impact of the SRC family kinases (SFKs) on NSCLC prognosis, and the immunomodulatory effect of the SFK inhibitor dasatinib, in combination with anti-PD-1, in clinically relevant mouse models of NSCLC. METHODS A cohort of patients from University Clinic of Navarra (n=116) was used to study immune infiltrates by multiplex immunofluorescence (mIF) and YES1 protein expression in tumor samples. Publicly available resources (TCGA, Km Plotter, and CIBERSORT) were used to study patient's survival based on expression of SFKs and tumor infiltrates. Syngeneic NSCLC mouse models 393P and UNSCC680AJ were used for in vivo drug testing. RESULTS Among the SFK members, YES1 expression showed the highest association with poor prognosis. Patients with high YES1 tumor levels also showed high infiltration of CD4+/FOXP3+ cells (regulatory T cells (Tregs)), suggesting an immunosuppressive phenotype. After testing for YES1 expression in a panel of murine cell lines, 393P and UNSCC680AJ were selected for in vivo studies. In the 393P model, dasatinib+anti-PD-1 treatment resulted in synergistic activity, with 87% tumor regressions and development of immunological memory that impeded tumor growth when mice were rechallenged. In vivo depletion experiments further showed that CD8+ and CD4+ cells are necessary for the therapeutic effect of the combination. The antitumor activity was accompanied by a very significant decrease in the number of Tregs, which was validated by mIF in tumor sections. In the UNSCC680AJ model, the antitumor effects of dasatinib+anti-PD-1 were milder but similar to the 393P model. In in vitro assays, we demonstrated that dasatinib blocks proliferation and transforming growth factor beta-driven conversion of effector CD4+ cells into Tregs through targeting of phospholymphocyte-specific protein tyrosine kinase and downstream effectors pSTAT5 and pSMAD3. CONCLUSIONS YES1 protein expression is associated with increased numbers of Tregs in patients with NSCLC. Dasatinib synergizes with anti-PD-1 to impair tumor growth in NSCLC experimental models. This study provides the preclinical rationale for the combined use of dasatinib and PD-1/programmed death-ligand 1 blockade to improve outcomes of patients with NSCLC.
Collapse
Affiliation(s)
- Esther Redin
- IDISNA and Program in Solid Tumors, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain.,CIBERONC, ISCIII, Madrid, Spain.,Department of Pathology, Anatomy and Physiology, School of Medicine, University of Navarra, Pamplona, Spain
| | - Irati Garmendia
- IDISNA and Program in Solid Tumors, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain.,Department of Pathology, Anatomy and Physiology, School of Medicine, University of Navarra, Pamplona, Spain
| | - Teresa Lozano
- Immunology and Immunotherapy Program, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain
| | - Diego Serrano
- IDISNA and Program in Solid Tumors, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain.,Department of Pathology, Anatomy and Physiology, School of Medicine, University of Navarra, Pamplona, Spain
| | - Yaiza Senent
- IDISNA and Program in Solid Tumors, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain
| | - Miriam Redrado
- IDISNA and Program in Solid Tumors, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain
| | - Maria Villalba
- Department of Pathology, University Clinic of Navarra, Pamplona, Spain
| | - Carlos E De Andrea
- Department of Pathology, Anatomy and Physiology, School of Medicine, University of Navarra, Pamplona, Spain.,Department of Pathology, University Clinic of Navarra, Pamplona, Spain
| | - Francisco Exposito
- IDISNA and Program in Solid Tumors, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain.,CIBERONC, ISCIII, Madrid, Spain.,Department of Pathology, Anatomy and Physiology, School of Medicine, University of Navarra, Pamplona, Spain
| | - Daniel Ajona
- IDISNA and Program in Solid Tumors, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain.,CIBERONC, ISCIII, Madrid, Spain.,Department of Biochemistry and Genetics, School of Sciences, University of Navarra, Pamplona, Spain
| | - Sergio Ortiz-Espinosa
- IDISNA and Program in Solid Tumors, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain.,Department of Biochemistry and Genetics, School of Sciences, University of Navarra, Pamplona, Spain
| | - Ana Remirez
- IDISNA and Program in Solid Tumors, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain
| | - Cristina Bertolo
- IDISNA and Program in Solid Tumors, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain.,CIBERONC, ISCIII, Madrid, Spain
| | - Cristina Sainz
- IDISNA and Program in Solid Tumors, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain
| | - Juana Garcia-Pedrero
- CIBERONC, ISCIII, Madrid, Spain.,Department of Otolaryngology, Hospital Universitario Central de Asturias and Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Instituto Universitario de Oncología del Principado de Asturias, University of Oviedo, Oviedo, Spain
| | - Ruben Pio
- IDISNA and Program in Solid Tumors, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain.,CIBERONC, ISCIII, Madrid, Spain.,Department of Biochemistry and Genetics, School of Sciences, University of Navarra, Pamplona, Spain
| | - Juan Lasarte
- Immunology and Immunotherapy Program, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain
| | - Jackeline Agorreta
- IDISNA and Program in Solid Tumors, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain.,CIBERONC, ISCIII, Madrid, Spain.,Department of Pathology, Anatomy and Physiology, School of Medicine, University of Navarra, Pamplona, Spain
| | - Luis M Montuenga
- IDISNA and Program in Solid Tumors, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain.,CIBERONC, ISCIII, Madrid, Spain.,Department of Pathology, Anatomy and Physiology, School of Medicine, University of Navarra, Pamplona, Spain
| | - Alfonso Calvo
- IDISNA and Program in Solid Tumors, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain .,CIBERONC, ISCIII, Madrid, Spain.,Department of Pathology, Anatomy and Physiology, School of Medicine, University of Navarra, Pamplona, Spain
| |
Collapse
|
103
|
Gadwa J, Bickett TE, Darragh LB, Knitz MW, Bhatia S, Piper M, Van Court B, Bhuvane S, Nguyen D, Nangia V, Kleczko EK, Nemenoff RA, Karam SD. Complement C3a and C5a receptor blockade modulates regulatory T cell conversion in head and neck cancer. J Immunother Cancer 2021; 9:e002585. [PMID: 33789881 PMCID: PMC8016081 DOI: 10.1136/jitc-2021-002585] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/09/2021] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Resistance to therapy is a major problem in treating head and neck squamous cell carcinomas (HNSCC). Complement system inhibition has been shown to reduce tumor growth, metastasis, and therapeutic resistance in other tumor models, but has yet to be explored in the context of HNSCC. Here, we tested the effects of complement inhibition and its therapeutic potential in HNSCC. METHODS We conducted our studies using two Human Papilloma Virus (HPV)-negative HNSCC orthotopic mouse models. Complement C3aR and C5aR1 receptor antagonists were paired with radiation therapy (RT). Tumor growth was measured and immune populations from tumor, lymph node, and peripheral blood were compared among various treatment groups. Genetically engineered mouse models DEREG and C3-/- were used in addition to standard wild type models. Flow cytometry, clinical gene sets, and in vitro assays were used to evaluate the role complement receptor blockade has on the immunological makeup of the tumor microenvironment. RESULTS In contrast to established literature, inhibition of complement C3a and C5a signaling using receptor antagonists accelerated tumor growth in multiple HNSCC cell lines and corresponded with increased frequency of regulatory T cell (Treg) populations. Local C3a and C5a signaling has importance for CD4 T cell homeostasis and eventual development into effector phenotypes. Interruption of this signaling axis drives a phenotypic conversion of CD4+ T cells into Tregs, characterized by enhanced expression of Foxp3. Depletion of Tregs reversed tumor growth, and combination of Treg depletion and C3a and C5a receptor inhibition decreased tumor growth below that of the control groups. Complete knockout of C3 does not harbor the expected effect on tumor growth, indicating a still undetermined compensatory mechanism. Dexamethasone is frequently prescribed to patients undergoing RT and inhibits complement activation. We report no deleterious effects associated with dexamethasone due to complement inhibition. CONCLUSIONS Our data establish Tregs as a pro-tumorigenic driver during complement inhibition and provide evidence that targeted C3a and C5a receptor inhibition may add therapeutic advantage when coupled with anti-Treg therapy.
Collapse
MESH Headings
- Animals
- Cell Line, Tumor
- Cell Proliferation/drug effects
- Complement C3/genetics
- Complement C3/metabolism
- Complement Inactivating Agents/toxicity
- Dexamethasone/toxicity
- Forkhead Transcription Factors/metabolism
- Head and Neck Neoplasms/genetics
- Head and Neck Neoplasms/immunology
- Head and Neck Neoplasms/metabolism
- Head and Neck Neoplasms/pathology
- Humans
- Lymphocytes, Tumor-Infiltrating/drug effects
- Lymphocytes, Tumor-Infiltrating/immunology
- Lymphocytes, Tumor-Infiltrating/metabolism
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Mice, Knockout
- Receptor, Anaphylatoxin C5a/antagonists & inhibitors
- Receptor, Anaphylatoxin C5a/metabolism
- Receptors, Complement/antagonists & inhibitors
- Receptors, Complement/metabolism
- Signal Transduction
- Squamous Cell Carcinoma of Head and Neck/genetics
- Squamous Cell Carcinoma of Head and Neck/immunology
- Squamous Cell Carcinoma of Head and Neck/metabolism
- Squamous Cell Carcinoma of Head and Neck/pathology
- T-Lymphocytes, Regulatory/drug effects
- T-Lymphocytes, Regulatory/immunology
- T-Lymphocytes, Regulatory/metabolism
- Time Factors
- Tumor Burden/drug effects
- Mice
Collapse
Affiliation(s)
- Jacob Gadwa
- Radiation Oncology, University of Colorado-Anschutz Medical Campus, Aurora, Colorado, USA
| | - Thomas E Bickett
- Radiation Oncology, University of Colorado-Anschutz Medical Campus, Aurora, Colorado, USA
| | - Laurel B Darragh
- Radiation Oncology, University of Colorado-Anschutz Medical Campus, Aurora, Colorado, USA
| | - Michael William Knitz
- Radiation Oncology, University of Colorado-Anschutz Medical Campus, Aurora, Colorado, USA
| | - Shilpa Bhatia
- Radiation Oncology, University of Colorado-Anschutz Medical Campus, Aurora, Colorado, USA
| | - Miles Piper
- Radiation Oncology, University of Colorado-Anschutz Medical Campus, Aurora, Colorado, USA
| | - Benjamin Van Court
- Radiation Oncology, University of Colorado-Anschutz Medical Campus, Aurora, Colorado, USA
| | - Shiv Bhuvane
- Radiation Oncology, University of Colorado-Anschutz Medical Campus, Aurora, Colorado, USA
| | - Diemmy Nguyen
- Radiation Oncology, University of Colorado-Anschutz Medical Campus, Aurora, Colorado, USA
| | - Varuna Nangia
- Radiation Oncology, University of Colorado-Anschutz Medical Campus, Aurora, Colorado, USA
| | - Emily K Kleczko
- Medicine, University of Colorado Denver-Anschutz Medical Campus, Aurora, Colorado, USA
| | - Raphael A Nemenoff
- Medicine, University of Colorado Denver-Anschutz Medical Campus, Aurora, Colorado, USA
| | - Sana D Karam
- Radiation Oncology, University of Colorado-Anschutz Medical Campus, Aurora, Colorado, USA
| |
Collapse
|
104
|
Emrani S, Lamar M, Price CC, Baliga S, Wasserman V, Matusz E, Swenson R, Baliga G, Libon DJ. Assessing the capacity for mental manipulation in patients with statically-determined mild cognitive impairment using digital technology. EXPLORATION OF MEDICINE 2021. [DOI: 10.37349/emed.2021.00034] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Aims: Prior research employing a standard backward digit span test has been successful in operationally defining neurocognitive constructs associated with the Fuster’s model of executive attention. The current research sought to test if similar behavior could be obtained using a cross-modal mental manipulation test.
Methods: Memory clinic patients were studied. Using Jak-Bondi criteria, 24 patients were classified with mild cognitive impairment (MCI), and 33 memory clinic patients did not meet criteria for MCI (i.e. non-MCI). All patients were assessed with the digital version of the WRAML-2 Symbolic Working Memory Test-Part 1, a cross-modal mental manipulation task where patients hear digits, but respond by touching digits from lowest to highest on an answer key. Only 4 and 5-span trials were analyzed. Using an iPad, all test stimuli were played; and, all responses were obtained with a touch key. Only correct trials were analyzed. Average time to complete trials and latency for each digit was recorded.
Results: Groups did not differ when average time to complete 4-span trials was calculated. MCI patients displayed slower latency, or required more time to re-order the 1st and 3rd digits. Regression analyses, primarily involving initial and latter response latencies, were associated with better, but different underlying neuropsychological abilities. Almost no 5-span analyses were significant.
Conclusions: This cross-modal test paradigm found no difference for total average time. MCI patients generated slower 1st and 3rd response latency, suggesting differences in time allocation to achieve correct serial order recall. Moreover, different neuropsychological abilities were associated with different time-based test components. These data extend prior findings using a standard backward digit span test. Differences in time epochs are consistent with constructs underlying the model of executive attention and help explain mental manipulation deficits in MCI. These latency measures could constitute neurocognitive biomarkers that track emergent disease.
Collapse
Affiliation(s)
- Sheina Emrani
- Department of Psychology, Rowan University, Glassboro, NJ 08028, USA
| | - Melissa Lamar
- Department of Behavioral Sciences and the Rush Alzheimer’s Disease Center, Rush University Medical Center, Chicago, IL 60612, USA
| | - Catherine C. Price
- Department of Clinical and Health Psychology, University of Florida, Gainesville, FL 32610, USA
| | - Satya Baliga
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Victor Wasserman
- Department of Psychology, Rowan University, Glassboro, NJ 08028, USA
| | - Emily Matusz
- 5New Jersey Institute for Successful Aging, School of Osteopathic Medicine, Rowan University, Stratford, NJ 08084, USA
| | - Rod Swenson
- Department Psychiatry and Behavioral Science, University of North Dakota School of Medicine and Health Sciences, Grand Fork, Fargo, ND 58103, USA
| | - Ganesh Baliga
- Department of Computer Science, Rowan University, Glassboro, NJ 08028, USA
| | - David J. Libon
- Department of Psychology, Rowan University, Glassboro, NJ 08028, USA 5New Jersey Institute for Successful Aging, School of Osteopathic Medicine, Rowan University, Stratford, NJ 08084, USA
| |
Collapse
|
105
|
Agnello L, Camorani S, Fedele M, Cerchia L. Aptamers and antibodies: rivals or allies in cancer targeted therapy? EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2021; 2:107-121. [PMID: 36046085 PMCID: PMC9400792 DOI: 10.37349/etat.2021.00035] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Accepted: 02/01/2021] [Indexed: 12/29/2022] Open
Abstract
The goal of an efficacious cancer therapy is to specifically target diseased cells at high accuracy while sparing normal, healthy cells. Over the past three decades, immunotherapy, based on the use of monoclonal antibodies (mAbs) directed against tumor-associated antigens, to inhibit their oncogenic function, or against immune checkpoints, to modulate specific T cell responses against cancer, has proven to be an important strategy for cancer therapy. Nevertheless, the number of mAbs approved for clinical use is still limited because of significant drawbacks to their applicability. Oligonucleotide aptamers, similarly to antibodies, form high-affinity bonds with their specific protein targets, thus representing an effective tool for active cancer targeting. Compared to antibodies, aptamers’ use as therapeutic agents benefits from their low size, low/no immunogenicity, simple synthesis and design flexibility for improving efficacy and stability. This review intends to highlight recently emerged applications of aptamers as recognition elements, from biomarker discovery to targeted drug delivery and targeted treatment, showing aptamers’ potential to work in conjunction with antibodies for attacking cancer from multiple flanks.
Collapse
Affiliation(s)
- Lisa Agnello
- Institute of Experimental Endocrinology and Oncology “Gaetano Salvatore”, National Research Council (CNR), Via S. Pansini 5, 80131 Naples, Italy
| | - Simona Camorani
- Institute of Experimental Endocrinology and Oncology “Gaetano Salvatore”, National Research Council (CNR), Via S. Pansini 5, 80131 Naples, Italy
| | - Monica Fedele
- Institute of Experimental Endocrinology and Oncology “Gaetano Salvatore”, National Research Council (CNR), Via S. Pansini 5, 80131 Naples, Italy
| | - Laura Cerchia
- Institute of Experimental Endocrinology and Oncology “Gaetano Salvatore”, National Research Council (CNR), Via S. Pansini 5, 80131 Naples, Italy
| |
Collapse
|
106
|
Wen J, Wang L, Ren J, Kranz E, Chen S, Wu D, Kanazawa T, Chen I, Lu Y, Kamata M. Nanoencapsulated rituximab mediates superior cellular immunity against metastatic B-cell lymphoma in a complement competent humanized mouse model. J Immunother Cancer 2021; 9:jitc-2020-001524. [PMID: 33593826 PMCID: PMC7888328 DOI: 10.1136/jitc-2020-001524] [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] [Accepted: 12/17/2020] [Indexed: 11/03/2022] Open
Abstract
BACKGROUND Despite the numerous applications of monoclonal antibodies (mAbs) in cancer therapeutics, animal models available to test the therapeutic efficacy of new mAbs are limited. NOD.Cg-Prkdcscid Il2rg tm1Wjl /SzJ (NSG) mice are one of the most highly immunodeficient strains and are universally used as a model for testing cancer-targeting mAbs. However, this strain lacks several factors necessary to fully support antibody-mediated effector functions-including antibody-dependent cellular cytotoxicity, antibody-dependent cellular phagocytosis, and complement-dependent cytotoxicity (CDC)-due to the absence of immune cells as well as a mutation in the Hc gene, which is needed for a functional complement system. METHODS We have developed a humanized mouse model using a novel NSG strain, NOD.Cg-Hc1 Prkdcscid Il2rgtm1Wjl/SzJ (NSG-Hc1), which contains the corrected mutation in the Hc gene to support CDC in addition to other mechanisms endowed by humanization. With this model, we reevaluated the anticancer efficacies of nanoencapsulated rituximab after xenograft of the human Burkitt lymphoma cell line 2F7-BR44. RESULTS As expected, xenografted humanized NSG-Hc1 mice supported superior lymphoma clearance of native rituximab compared with the parental NSG strain. Nanoencapsulated rituximab with CXCL13 conjugation as a targeting ligand for lymphomas further enhanced antilymphoma activity in NSG-Hc1 mice and, more importantly, mediated antilymphoma cellular responses. CONCLUSIONS These results indicate that NSG-Hc1 mice can serve as a feasible model for both studying antitumor treatment using cancer targeting as well as understanding induction mechanisms of antitumor cellular immune response.
Collapse
Affiliation(s)
- Jing Wen
- Department of Microbiology, Immunology and Molecular Genetics, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, USA.,UCLA AIDS Institute, University of California Los Angeles, Los Angeles, California, USA
| | - Lan Wang
- Department of Microbiology, Immunology and Molecular Genetics, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, USA.,UCLA AIDS Institute, University of California Los Angeles, Los Angeles, California, USA
| | - Jie Ren
- Department of Chemical and Biomolecular Engineering, University of California Los Angeles, Los Angeles, California, USA
| | - Emiko Kranz
- Department of Microbiology, Immunology and Molecular Genetics, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, USA.,UCLA AIDS Institute, University of California Los Angeles, Los Angeles, California, USA
| | - Shilin Chen
- Department of Chemical and Biomolecular Engineering, University of California Los Angeles, Los Angeles, California, USA
| | - Di Wu
- Department of Chemical and Biomolecular Engineering, University of California Los Angeles, Los Angeles, California, USA
| | - Toshio Kanazawa
- Department of Microbiology, Immunology and Molecular Genetics, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, USA.,UCLA AIDS Institute, University of California Los Angeles, Los Angeles, California, USA
| | - Irvin Chen
- Department of Microbiology, Immunology and Molecular Genetics, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, USA.,UCLA AIDS Institute, University of California Los Angeles, Los Angeles, California, USA
| | - Yunfeng Lu
- Department of Chemical and Biomolecular Engineering, University of California Los Angeles, Los Angeles, California, USA
| | - Masakazu Kamata
- Microbiology, University of Alabama at Birmingham School of Arts and Humanities, Birmingham, Alabama, USA
| |
Collapse
|
107
|
Bao D, Zhang C, Li L, Wang H, Li Q, Ni L, Lin Y, Huang R, Yang Z, Zhang Y, Hu Y. Integrative Analysis of Complement System to Prognosis and Immune Infiltrating in Colon Cancer and Gastric Cancer. Front Oncol 2021; 10:553297. [PMID: 33614473 PMCID: PMC7886994 DOI: 10.3389/fonc.2020.553297] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Accepted: 12/14/2020] [Indexed: 12/14/2022] Open
Abstract
Background The complement system acts as an integral part of the innate immune response, which acts primarily to remove pathogens and injured cells. Emerging evidence has shown the activation of the immune regulatory function of complements in the tumor microenvironment (TME). We revealed the expression levels of various complements in human cancers and their role in tumor prognosis and immune infiltration. Methods The differential expression of complements was explored via the Tumor Immune Estimation Resource (TIMER) site and the Oncomine database. To investigate whether these differentially expressed complements have correlation with the prognosis of gastric cancer (GC) and colon cancer, their impact on survival was assessed using the PrognoScan database and Kaplan-Meier plotter. The correlations between complements and tumor immune-infiltrating levels and immune gene markers were statistically explored in TIMER based on Spearman's correlation coefficients and p-values. Results In two colon cancer cohorts, an increased expression level of DAF (CD55) has statistically significant correlation with poor disease-free survival (DFS). High C3, CR4, and C5aR1 expression levels were significantly related with poor prognosis in GC patients. In addition, C3, CR4, and C5aR1 expression was positively related to the tumor purity and infiltration levels of multiple immune cells in stomach adenocarcinoma (STAD). Moreover, the expression levels of C3, CR4, and C5aR1 were also strongly correlated with various immune marker sets, such as those of tumor-associated macrophages (TAMs), M1 and M2 macrophages, T cell exhaustion, Tregs, and DCs, in STAD. Additionally, CD55 has positive correlation with few immune cell infiltration levels in colon adenocarcinoma (COAD), but its correlation with immune marker sets was not statistically significant. Conclusion These findings confirm the relationship between various complements and tumor prognosis and immune infiltration in colon cancer and GC. CD55 may serve as an indicator on the survival prognosis of patients with colon cancer. Furthermore, as biomarkers for poor prognosis in GC, complements C3, CR4, and C5aR1 may provide potential biological targets for GC immunotherapy.
Collapse
Affiliation(s)
- Dandan Bao
- Department of General Surgery, The Third Clinical Institute Affiliated to Wenzhou Medical University, Wenzhou People's Hospital, Wenzhou, China
| | - Chenghao Zhang
- Emergency department, Wenzhou People's Hospital, The Third Clinical Institute Affiliated to Wenzhou Medical University, Wenzhou, China
| | - Longlong Li
- Department of Gastrointestinal Surgery, People's Hospital of Deyang City, Sichuan, China
| | - Haihong Wang
- Department of General Surgery, The Third Clinical Institute Affiliated to Wenzhou Medical University, Wenzhou People's Hospital, Wenzhou, China
| | - Qiuyan Li
- Department of Oncology, Wenzhou Medical University, Wenzhou, China
| | - Leilei Ni
- Department of General Surgery, The Third Clinical Institute Affiliated to Wenzhou Medical University, Wenzhou People's Hospital, Wenzhou, China
| | - Yinfeng Lin
- Department of Oncology, The Third Clinical Institute Affiliated to Wenzhou Medical University, Wenzhou People's Hospital, Wenzhou, China
| | - Rong Huang
- Shanghai Institute for Food and Drug Control, Shanghai, China
| | - Zhangwei Yang
- Department of General Surgery, The Third Clinical Institute Affiliated to Wenzhou Medical University, Wenzhou People's Hospital, Wenzhou, China
| | - Yan Zhang
- Department of Gastroenterology, Yijishan Hospital, the First Affiliated Hospital of Wannan Medical College, Wuhu, China
| | - Yiren Hu
- Department of General Surgery, Medical College of Soochow University, The Third Clinical Institute Affiliated to Wenzhou Medical University, Wenzhou People's Hospital, Wenzhou, China
| |
Collapse
|
108
|
Complement activation promoted by the lectin pathway mediates C3aR-dependent sarcoma progression and immunosuppression. NATURE CANCER 2021; 2:218-232. [PMID: 34505065 PMCID: PMC8425276 DOI: 10.1038/s43018-021-00173-0] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Complement has emerged as a component of tumor promoting inflammation. We conducted a systematic assessment of the role of complement activation and effector pathways in sarcomas. C3-/-, MBL1/2-/- and C4-/- mice showed reduced susceptibility to 3-methylcholanthrene sarcomagenesis and transplanted sarcomas, whereas C1q and factor B deficiency had marginal effects. Complement 3a receptor (C3aR), but not C5aR1 and C5aR2, deficiency mirrored the phenotype of C3-/- mice. C3 and C3aR deficiency were associated with reduced accumulation and functional skewing of tumor-associated macrophages, increased T cell activation and response to anti-PD-1 therapy. Transcriptional profiling of sarcoma infiltrating macrophages and monocytes revealed the enrichment of MHC II-dependent antigen presentation pathway in C3-deficient cells. In patients, C3aR expression correlated with a macrophage population signature and C3 deficiency-associated signatures predicted better clinical outcome. These results suggest that the lectin pathway and C3a/C3aR axis are key components of complement and macrophage-mediated sarcoma promotion and immunosuppression.
Collapse
|
109
|
Imamura R, Kitagawa S, Kubo T, Irie A, Kariu T, Yoneda M, Kamba T, Imamura T. Prostate cancer C5a receptor expression and augmentation of cancer cell proliferation, invasion, and PD-L1 expression by C5a. Prostate 2021; 81:147-156. [PMID: 33368414 DOI: 10.1002/pros.24090] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Accepted: 11/21/2020] [Indexed: 01/09/2023]
Abstract
BACKGROUND The treatment of castration-resistant prostate cancer (CRPC) is a urological issue. Recent studies have revealed cancer promotion via the C5a-C5a receptor (C5aR) system. To establish a new therapeutic target for CRPC, we investigated an association of the system with CRPC progression and evasion from the antitumor immune responses. METHODS C5aR and PD-L1 were immunostained in the prostate cancer (PC) tissues. The relationship of PC C5aR expression to clinicopathological parameters was analyzed. CRPC cell lines were examined for C5aR expression by real-time reverse transcription polymerase chain reaction, immunoblotting, and flow cytometry. C5a effects were examined on CRPC cell glutamine consumption, proliferation, invasion, and PD-L1 expression. RESULTS PC cells expressed C5aR in 83 of the 161 patients (52%) and in three of the six CRPC patients. Basal cells, but not luminal cells, of noncancerous prostate glands expressed C5aR. Three CRPC cell lines expressed C5aR. C5a increased CRPC cell glutamine consumption 2.1-fold, proliferation 1.3-1.6-fold, and invasion 2-3-fold in a C5a-concentration and a C5aR-dependent manner. High expression of C5aR did not relate to the PC patients' clinical parameters but the PD-L1-positive rate was higher in the C5aR high-expression patients (37.5%) compared to low- or no expression patients (17.8%), and double-positive PC cells were present. C5a increased CRPC cell PD-L1 production 1.4-fold and cell-surface expression 2.6-fold. CONCLUSIONS C5aR expression of PC cells in patients' tissues and C5a augmentation of C5aR-dependent CRPC proliferation, invasion, and PD-L1 expression suggested participation of the C5a-C5aR system in CRPC promotion and evasion from antitumor immune responses. Targeting this signaling pathway may provide a useful therapeutic option for CRPC.
Collapse
Affiliation(s)
- Ryuji Imamura
- Department of Molecular Pathology, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
- Department of Urology, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Saki Kitagawa
- Department of Molecular Pathology, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Tatsuko Kubo
- Department of Molecular Pathology, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Atsushi Irie
- Department of Immunogenetics, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Toru Kariu
- Department of Life Science, Shokei University, Kumamoto, Japan
| | - Masakazu Yoneda
- Department of Oral Surgery, Oral and Maxillofacial Center, Kagoshima University Hospital, Kagoshima, Japan
| | - Tomomi Kamba
- Department of Urology, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Takahisa Imamura
- Department of Molecular Pathology, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
- Department of Life Science, Shokei University, Kumamoto, Japan
| |
Collapse
|
110
|
Hou J, Karin M, Sun B. Targeting cancer-promoting inflammation - have anti-inflammatory therapies come of age? Nat Rev Clin Oncol 2021; 18:261-279. [PMID: 33469195 DOI: 10.1038/s41571-020-00459-9] [Citation(s) in RCA: 205] [Impact Index Per Article: 51.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/25/2020] [Indexed: 02/07/2023]
Abstract
The immune system has crucial roles in cancer development and treatment. Whereas adaptive immunity can prevent or constrain cancer through immunosurveillance, innate immunity and inflammation often promote tumorigenesis and malignant progression of nascent cancer. The past decade has witnessed the translation of knowledge derived from preclinical studies of antitumour immunity into clinically effective, approved immunotherapies for cancer. By contrast, the successful implementation of treatments that target cancer-associated inflammation is still awaited. Anti-inflammatory agents have the potential to not only prevent or delay cancer onset but also to improve the efficacy of conventional therapeutics and next-generation immunotherapies. Herein, we review the current clinical advances and experimental findings supporting the utility of an anti-inflammatory approach to the treatment of solid malignancies. Gaining a better mechanistic understanding of the mode of action of anti-inflammatory agents and designing more effective treatment combinations would advance the clinical application of this therapeutic approach.
Collapse
Affiliation(s)
- Jiajie Hou
- Department of Hepatobiliary Surgery, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China.,Department of Liver Surgery, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Michael Karin
- Laboratory of Gene Regulation and Signal Transduction, Departments of Pharmacology and Pathology, University of California San Diego School of Medicine, La Jolla, CA, USA.
| | - Beicheng Sun
- Department of Hepatobiliary Surgery, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China.
| |
Collapse
|
111
|
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
|
112
|
Teijeira A, Garasa S, Ochoa MC, Villalba M, Olivera I, Cirella A, Eguren-Santamaria I, Berraondo P, Schalper KA, de Andrea CE, Sanmamed MF, Melero I. IL8, Neutrophils, and NETs in a Collusion against Cancer Immunity and Immunotherapy. Clin Cancer Res 2020; 27:2383-2393. [PMID: 33376096 DOI: 10.1158/1078-0432.ccr-20-1319] [Citation(s) in RCA: 147] [Impact Index Per Article: 29.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 11/18/2020] [Accepted: 12/23/2020] [Indexed: 12/12/2022]
Abstract
One of the most important mechanisms by which cancer fosters its own development is the generation of an immune microenvironment that inhibits or impairs antitumor immune responses. A cancer permissive immune microenvironment is present in a large proportion of the patients with cancer who do not respond to immunotherapy approaches intended to trigger preexisting antitumor immune responses, for instance, immune checkpoint blockade. High circulating levels of IL8 in patients with cancer quite accurately predict those who will not benefit from checkpoint-based immunotherapy. IL8 has been reported to favor cancer progression and metastases via different mechanisms, including proangiogenesis and the maintenance of cancer stem cells, but its ability to attract and functionally modulate neutrophils and macrophages is arguably one of the most important factors. IL8 does not only recruit neutrophils to tumor lesions, but also triggers the extrusion of neutrophil extracellular traps (NET). The relevance and mechanisms underlying the contribution of both neutrophils and NETs to cancer development and progression are starting to be uncovered and include both direct effects on cancer cells and changes in the tumor microenvironment, such as facilitating metastasis, awakening micrometastases from dormancy, and facilitating escape from cytotoxic immune cells. Blockade of IL8 or its receptors (CXCR1 and CXCR2) is being pursued in drug development, and clinical trials alone or in combination with anti-PD-L1 checkpoint inhibitors are already ongoing.
Collapse
Affiliation(s)
- Alvaro Teijeira
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, Pamplona, Navarra, Spain. .,Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain.,Navarra Institute for Health Research (IDISNA), Pamplona, Navarra, Spain
| | - Saray Garasa
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, Pamplona, Navarra, Spain.,Navarra Institute for Health Research (IDISNA), Pamplona, Navarra, Spain
| | - Maria C Ochoa
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, Pamplona, Navarra, Spain.,Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
| | - Maria Villalba
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain.,Department of Pathology, Clínica Universidad de Navarra, Pamplona, Navarra, Spain
| | - Irene Olivera
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, Pamplona, Navarra, Spain.,Navarra Institute for Health Research (IDISNA), Pamplona, Navarra, Spain
| | - Assunta Cirella
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, Pamplona, Navarra, Spain.,Navarra Institute for Health Research (IDISNA), Pamplona, Navarra, Spain
| | - Iñaki Eguren-Santamaria
- Departments of Oncology and Immunology, Clinica Universidad de Navarra, Pamplona, Navarra, Spain
| | - Pedro Berraondo
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, Pamplona, Navarra, Spain.,Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain.,Navarra Institute for Health Research (IDISNA), Pamplona, Navarra, Spain
| | - Kurt A Schalper
- Department of Pathology, Yale University School of Medicine, New Haven, Connecticut
| | - Carlos E de Andrea
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain.,Department of Pathology, Clínica Universidad de Navarra, Pamplona, Navarra, Spain
| | - Miguel F Sanmamed
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, Pamplona, Navarra, Spain.,Navarra Institute for Health Research (IDISNA), Pamplona, Navarra, Spain.,Departments of Oncology and Immunology, Clinica Universidad de Navarra, Pamplona, Navarra, Spain
| | - Ignacio Melero
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, Pamplona, Navarra, Spain. .,Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain.,Navarra Institute for Health Research (IDISNA), Pamplona, Navarra, Spain
| |
Collapse
|
113
|
Chen B, Zhou W, Tang C, Wang G, Yuan P, Zhang Y, Bhushan SC, Ma J, Leng J. Down-Regulation of C3aR/C5aR Inhibits Cell Proliferation and EMT in Hepatocellular Carcinoma. Technol Cancer Res Treat 2020; 19:1533033820970668. [PMID: 33176600 PMCID: PMC7672723 DOI: 10.1177/1533033820970668] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Complement 3a (C3a) and complement 5a (C5a), small cleavage fragments generated by complement activation, has been previously shown to be obviously up-regulated in highly metastatic hepatocellular carcinoma (HCC) cells. However, their functional roles in HCC cells remains unclear. Here, we investigated the biological function of G protein-coupled receptor C3aR/C5aR using small interference RNA in HCC cells. Our data showed that C3aR and C5aR knockdown significantly inhibited the proliferation, migration and invasion of HCC cells using CCK-8, colony formation and transwell assays. Flow cytometry assay showed C3aR and C5aR knockdown induced cell cycle G0/G1 phase arrest and apoptosis in HCC cells. Moreover, we found down-regulation of C3aR/C5aR obviously down-regulated the expression of PCNA, Ki-67 and suppressed the epithelial-mesenchymal transition (EMT) markers (E-cadherin, N-cadherin and vimentin) in HCC cells. Collectively, our data demonstrated that targeting C3aR/C5aR may hold promise for the treatment of HCC.
Collapse
Affiliation(s)
- Bendong Chen
- Department of Hepatobiliary Surgery, 105002General Hospital of Ningxia Medical University, Yinchuan, Ningxia, China
| | - Wenyan Zhou
- Department of ICU, 105002General Hospital of Ningxia Medical University, Yinchuan, Ningxia, China
| | - Chaofeng Tang
- Department of Hepatobiliary Surgery, 105002General Hospital of Ningxia Medical University, Yinchuan, Ningxia, China
| | - Genwang Wang
- Department of Hepatobiliary Surgery, 105002General Hospital of Ningxia Medical University, Yinchuan, Ningxia, China
| | - Peng Yuan
- Department of Hepatobiliary Surgery, 105002General Hospital of Ningxia Medical University, Yinchuan, Ningxia, China
| | - Yawen Zhang
- 105002Ningxia Medical University, Yinchuan, Ningxia, China
| | | | - Jinlong Ma
- 105002Ningxia Medical University, Yinchuan, Ningxia, China
| | - Junzhi Leng
- Department of Hepatobiliary Surgery, 105002General Hospital of Ningxia Medical University, Yinchuan, Ningxia, China
| |
Collapse
|
114
|
Reese B, Silwal A, Daugherity E, Daugherity M, Arabi M, Daly P, Paterson Y, Woolford L, Christie A, Elias R, Brugarolas J, Wang T, Karbowniczek M, Markiewski MM. Complement as Prognostic Biomarker and Potential Therapeutic Target in Renal Cell Carcinoma. THE JOURNAL OF IMMUNOLOGY 2020; 205:3218-3229. [PMID: 33158953 DOI: 10.4049/jimmunol.2000511] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Accepted: 09/29/2020] [Indexed: 12/21/2022]
Abstract
Preclinical studies demonstrated that complement promotes tumor growth. Therefore, we sought to determine the best target for complement-based therapy among common human malignancies. High expression of 11 complement genes was linked to unfavorable prognosis in renal cell carcinoma. Complement protein expression or deposition was observed mainly in stroma, leukocytes, and tumor vasculature, corresponding to a role of complement in regulating the tumor microenvironment. Complement abundance in tumors correlated with a high nuclear grade. Complement genes clustered within an aggressive inflammatory subtype of renal cancer characterized by poor prognosis, markers of T cell dysfunction, and alternatively activated macrophages. Plasma levels of complement proteins correlated with response to immune checkpoint inhibitors. Corroborating human data, complement deficiencies and blockade reduced tumor growth by enhancing antitumor immunity and seemingly reducing angiogenesis in a mouse model of kidney cancer resistant to PD-1 blockade. Overall, this study implicates complement in the immune landscape of renal cell carcinoma, and notwithstanding cohort size and preclinical model limitations, the data suggest that tumors resistant to immune checkpoint inhibitors might be suitable targets for complement-based therapy.
Collapse
Affiliation(s)
- Britney Reese
- Department of Immunotherapeutics and Biotechnology, School of Pharmacy, Texas Tech University Health Sciences Center, Abilene, TX 79601
| | - Ashok Silwal
- Department of Immunotherapeutics and Biotechnology, School of Pharmacy, Texas Tech University Health Sciences Center, Abilene, TX 79601
| | - Elizabeth Daugherity
- Department of Immunotherapeutics and Biotechnology, School of Pharmacy, Texas Tech University Health Sciences Center, Abilene, TX 79601
| | - Michael Daugherity
- Department of Engineering and Physics, Abilene Christian University, Abilene, TX 79601
| | - Mahshid Arabi
- Department of Immunotherapeutics and Biotechnology, School of Pharmacy, Texas Tech University Health Sciences Center, Abilene, TX 79601
| | - Pierce Daly
- Department of Immunotherapeutics and Biotechnology, School of Pharmacy, Texas Tech University Health Sciences Center, Abilene, TX 79601
| | - Yvonne Paterson
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - Layton Woolford
- Division of Hematology and Oncology, Department of Internal Medicine, University of Texas Southwestern, Dallas, TX 75390.,Kidney Cancer Program, Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX 75390; and
| | - Alana Christie
- Division of Hematology and Oncology, Department of Internal Medicine, University of Texas Southwestern, Dallas, TX 75390.,Kidney Cancer Program, Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX 75390; and
| | - Roy Elias
- Division of Hematology and Oncology, Department of Internal Medicine, University of Texas Southwestern, Dallas, TX 75390.,Kidney Cancer Program, Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX 75390; and
| | - James Brugarolas
- Division of Hematology and Oncology, Department of Internal Medicine, University of Texas Southwestern, Dallas, TX 75390.,Kidney Cancer Program, Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX 75390; and
| | - Tao Wang
- Kidney Cancer Program, Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX 75390; and.,The Quantitative Biomedical Research Center, Department of Population and Data Sciences, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Magdalena Karbowniczek
- Department of Immunotherapeutics and Biotechnology, School of Pharmacy, Texas Tech University Health Sciences Center, Abilene, TX 79601
| | - Maciej M Markiewski
- Department of Immunotherapeutics and Biotechnology, School of Pharmacy, Texas Tech University Health Sciences Center, Abilene, TX 79601;
| |
Collapse
|
115
|
Thurman JM, Laskowski J, Nemenoff RA. Complement and Cancer-A Dysfunctional Relationship? Antibodies (Basel) 2020; 9:antib9040061. [PMID: 33167384 PMCID: PMC7709115 DOI: 10.3390/antib9040061] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 09/08/2020] [Accepted: 11/03/2020] [Indexed: 12/13/2022] Open
Abstract
Although it was long believed that the complement system helps the body to identify and remove transformed cells, it is now clear that complement activation contributes to carcinogenesis and can also help tumors to escape immune-elimination. Complement is activated by several different mechanisms in various types of cancer, and complement activation fragments have multiple different downstream effects on cancer cells and throughout the tumor microenvironment. Thus, the role of complement activation in tumor biology may vary among different types of cancer and over time within a single tumor. In multiple different pre-clinical models, however, complement activation has been shown to recruit immunosuppressive myeloid cells into the tumor microenvironment. These cells, in turn, suppress anti-tumor T cell immunity, enabling the tumor to grow. Based on extensive pre-clinical work, therapeutic complement inhibitors hold great promise as a new class of immunotherapy. A greater understanding of the role of complement in tumor biology will improve our ability to identify those patients most likely to benefit from this treatment and to rationally combine complement inhibitors with other cancer therapies.
Collapse
|
116
|
Complement System: Promoter or Suppressor of Cancer Progression? Antibodies (Basel) 2020; 9:antib9040057. [PMID: 33113844 PMCID: PMC7709131 DOI: 10.3390/antib9040057] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Revised: 10/10/2020] [Accepted: 10/20/2020] [Indexed: 12/13/2022] Open
Abstract
Constituent of innate immunity, complement is present in the tumor microenvironment. The functions of complement include clearance of pathogens and maintenance of homeostasis, and as such could contribute to an anti-tumoral role in the context of certain cancers. However, multiple lines of evidence show that in many cancers, complement has pro-tumoral actions. The large number of complement molecules (over 30), the diversity of their functions (related or not to the complement cascade), and the variety of cancer types make the complement-cancer topic a very complex matter that has just started to be unraveled. With this review we highlight the context-dependent role of complement in cancer. Recent studies revealed that depending of the cancer type, complement can be pro or anti-tumoral and, even for the same type of cancer, different models presented opposite effects. We aim to clarify the current knowledge of the role of complement in human cancers and the insights from mouse models. Using our classification of human cancers based on the prognostic impact of the overexpression of complement genes, we emphasize the strong potential for therapeutic targeting the complement system in selected subgroups of cancer patients.
Collapse
|
117
|
Nguyen HD, Allaire A, Diamandis P, Bisaillon M, Scott MS, Richer M. A machine learning analysis of a "normal-like" IDH-WT diffuse glioma transcriptomic subgroup associated with prolonged survival reveals novel immune and neurotransmitter-related actionable targets. BMC Med 2020; 18:280. [PMID: 33059718 PMCID: PMC7565364 DOI: 10.1186/s12916-020-01748-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Accepted: 08/14/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Classification of primary central nervous system tumors according to the World Health Organization guidelines follows the integration of histologic interpretation with molecular information and aims at providing the most precise prognosis and optimal patient management. According to the cIMPACT-NOW update 3, diffuse isocitrate dehydrogenase-wild type (IDH-WT) gliomas should be graded as grade IV glioblastomas (GBM) if they possess one or more of the following molecular markers that predict aggressive clinical course: EGFR amplification, TERT promoter mutation, and whole-chromosome 7 gain combined with chromosome 10 loss. METHODS The Cancer Genome Atlas (TCGA) glioma expression datasets were reanalyzed in order to identify novel tumor subcategories which would be considered as GBM-equivalents with the current diagnostic algorithm. Unsupervised clustering allowed the identification of previously unrecognized transcriptomic subcategories. A supervised machine learning algorithm (k-nearest neighbor model) was also used to identify gene signatures specific to some of these subcategories. RESULTS We identified 14 IDH-WT infiltrating gliomas displaying a "normal-like" (NL) transcriptomic profile associated with a longer survival. Genes such as C5AR1 (complement receptor), SLC32A1 (vesicular gamma-aminobutyric acid transporter), MSR1 (or CD204, scavenger receptor A), and SYT5 (synaptotagmin 5) were differentially expressed and comprised in gene signatures specific to NL IDH-WT gliomas which were validated further using the Chinese Glioma Genome Atlas datasets. These gene signatures showed high discriminative power and correlation with survival. CONCLUSION NL IDH-WT gliomas represent an infiltrating glioma subcategory with a superior prognosis which can only be detected using genome-wide analysis. Differential expression of genes potentially involved in immune checkpoint and amino acid signaling pathways is providing insight into mechanisms of gliomagenesis and could pave the way to novel treatment targets for infiltrating gliomas.
Collapse
Affiliation(s)
- H. D. Nguyen
- Department of Biochemistry and Functional Genomics, Université de Sherbrooke, Sherbrooke, Québec Canada
| | - A. Allaire
- Department of Biochemistry and Functional Genomics, Université de Sherbrooke, Sherbrooke, Québec Canada
| | - P. Diamandis
- Department of Laboratory Medicine and Pathobiology and Princess Margaret Cancer Center, University of Toronto, Toronto, Ontario Canada
| | - M. Bisaillon
- Department of Biochemistry and Functional Genomics, Université de Sherbrooke, Sherbrooke, Québec Canada
| | - M. S. Scott
- Department of Biochemistry and Functional Genomics, Université de Sherbrooke, Sherbrooke, Québec Canada
| | - M. Richer
- Department of Pathology, Université de Sherbrooke, Sherbrooke, Québec Canada
| |
Collapse
|
118
|
Nguyen TL, Yin Y, Choi Y, Jeong JH, Kim J. Enhanced Cancer DNA Vaccine via Direct Transfection to Host Dendritic Cells Recruited in Injectable Scaffolds. ACS NANO 2020; 14:11623-11636. [PMID: 32808762 DOI: 10.1021/acsnano.0c04188] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Deoxyribonucleic acid (DNA) vaccines are a promising cancer immunotherapy approach. However, effective delivery of DNA to antigen-presenting cells (e.g., dendritic cells (DCs)) for the induction of an adaptive immune response is limited. Conventional DNA delivery via intramuscular, intradermal, and subcutaneous injection by hypodermal needles shows a low potency and immunogenicity. Here, we propose the enhanced cancer DNA vaccine by direct transfection to the high number of DCs recruited into the chemoattractant-loaded injectable mesoporous silica microrods (MSRs). Subcutaneous administration of the MSRs mixed with tumor-antigen coding DNA polyplexes resulted in DC recruitment in the macroporous space of the scaffold formed by the spontaneous assembly of high-aspect-ratio MSRs, thereby allowing for enhanced cellular uptake of antigen-coded DNA by host DCs. The MSR scaffolds delivering the DNA vaccine trigger a more robust DC activation, antigen-specific CD8+ T cell response, and Th1 immune response compared to the bolus DNA vaccine. Additionally, the immunological memory can be induced with a single administration of the vaccine. The combination of the vaccination and antiprogrammed cell death-1 antibody significantly eliminates established lung metastasis. These results indicate that MSRs serve as a powerful platform for DNA vaccine delivery to DCs for effective cancer immunotherapy.
Collapse
Affiliation(s)
- Thanh Loc Nguyen
- School of Chemical Engineering, Sungkyunkwan University (SKKU), Suwon 16419, Republic of Korea
| | - Yue Yin
- School of Pharmacy, Sungkyunkwan University (SKKU), Suwon 16419, Republic of Korea
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, China
| | - Youngjin Choi
- School of Chemical Engineering, Sungkyunkwan University (SKKU), Suwon 16419, Republic of Korea
| | - Ji Hoon Jeong
- School of Pharmacy, Sungkyunkwan University (SKKU), Suwon 16419, Republic of Korea
| | - Jaeyun Kim
- School of Chemical Engineering, Sungkyunkwan University (SKKU), Suwon 16419, Republic of Korea
- Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences & Technology (SAIHST), Sungkyunkwan University (SKKU), Suwon 16419, Republic of Korea
- Biomedical Institute for Convergence at SKKU (BICS), Sungkyunkwan University (SKKU), Suwon 16419, Republic of Korea
- Institute of Quantum Biophysics (IQB), Sungkyunkwan University (SKKU), Suwon 16419, Republic of Korea
| |
Collapse
|
119
|
Rotoli D, Santana-Viera L, Ibba ML, Esposito CL, Catuogno S. Advances in Oligonucleotide Aptamers for NSCLC Targeting. Int J Mol Sci 2020; 21:ijms21176075. [PMID: 32842557 PMCID: PMC7504093 DOI: 10.3390/ijms21176075] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 08/20/2020] [Accepted: 08/22/2020] [Indexed: 02/07/2023] Open
Abstract
Non-small-cell lung cancer (NSCLC) is the most common type of lung cancer worldwide, with the highest incidence in developed countries. NSCLC patients often face resistance to currently available therapies, accounting for frequent relapses and poor prognosis. Indeed, despite great recent advancements in the field of NSCLC diagnosis and multimodal therapy, most patients are diagnosed at advanced metastatic stage, with a very low overall survival. Thus, the identification of new effective diagnostic and therapeutic options for NSCLC patients is a crucial challenge in oncology. A promising class of targeting molecules is represented by nucleic-acid aptamers, short single-stranded oligonucleotides that upon folding in particular three dimensional (3D) structures, serve as high affinity ligands towards disease-associated proteins. They are produced in vitro by SELEX (systematic evolution of ligands by exponential enrichment), a combinatorial chemistry procedure, representing an important tool for novel targetable biomarker discovery of both diagnostic and therapeutic interest. Aptamer-based approaches are promising options for NSCLC early diagnosis and targeted therapy and may overcome the key obstacles of currently used therapeutic modalities, such as the high toxicity and patients’ resistance. In this review, we highlight the most important applications of SELEX technology and aptamers for NSCLC handling.
Collapse
Affiliation(s)
- Deborah Rotoli
- Institute Experimental Endocrinology and Oncology “Gaetano Salvatore” (IEOS), National Research Council (CNR), 80145 Naples, Italy; (D.R.); (L.S.-V.)
| | - Laura Santana-Viera
- Institute Experimental Endocrinology and Oncology “Gaetano Salvatore” (IEOS), National Research Council (CNR), 80145 Naples, Italy; (D.R.); (L.S.-V.)
| | - Maria L. Ibba
- Department of Molecular Medicine and Medical Biotechnology, “Federico II” University of Naples, 80131 Naples, Italy;
| | - Carla L. Esposito
- Institute Experimental Endocrinology and Oncology “Gaetano Salvatore” (IEOS), National Research Council (CNR), 80145 Naples, Italy; (D.R.); (L.S.-V.)
- Correspondence: (C.L.E.); (S.C.); Tel.: +39-081-3722343 (C.L.E. & S.C.)
| | - Silvia Catuogno
- Institute Experimental Endocrinology and Oncology “Gaetano Salvatore” (IEOS), National Research Council (CNR), 80145 Naples, Italy; (D.R.); (L.S.-V.)
- Correspondence: (C.L.E.); (S.C.); Tel.: +39-081-3722343 (C.L.E. & S.C.)
| |
Collapse
|
120
|
Wieleba I, Wojas-Krawczyk K, Krawczyk P. Aptamers in Non-Small Cell Lung Cancer Treatment. Molecules 2020; 25:molecules25143138. [PMID: 32659994 PMCID: PMC7396979 DOI: 10.3390/molecules25143138] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 06/16/2020] [Accepted: 07/08/2020] [Indexed: 12/12/2022] Open
Abstract
Aptamers are short, single-stranded oligonucleotides which are capable of specifically binding to single molecules and cellular structures. Aptamers are also known as “chemical antibodies”. Compared to monoclonal antibodies, they are characterized by higher reaction specificity, lower molecular weight, lower production costs, and lower variability in the production stage. Aptamer research has been extended during the past twenty years, but only Macugen® has been accepted by the Food and Drug Administration (FDA) to date, and few aptamers have been examined in clinical trials. In vitro studies with aptamers have shown that they may take part in the regulation of cancer progression, angiogenesis, and metastasis processes. In this article, we focus on the potential use of aptamers in non-small cell lung cancer treatment.
Collapse
|
121
|
Deciphering the Intricate Roles of Radiation Therapy and Complement Activation in Cancer. Int J Radiat Oncol Biol Phys 2020; 108:46-55. [PMID: 32629082 DOI: 10.1016/j.ijrobp.2020.06.067] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 06/29/2020] [Accepted: 06/30/2020] [Indexed: 12/22/2022]
Abstract
The complement system consists of a collection of serum proteins that act as the main frontline effector arm of the innate immune system. Activation of complement can occur through 3 individual induction pathways: the classical, mannose-binding lectin, and alternative pathways. Activation results in opsonization, recruitment of effector cells through potent immune mediators known as anaphylatoxins, and cell lysis via the formation of the membrane attack complex. Stringent regulation of complement is required to protect against inappropriate activation of the complement cascade. Complement activation within the tumor microenvironment does not increase antitumoral action; instead, it enhances tumor growth and disease progression. Radiation therapy (RT) is a staple in the treatment of malignancies and controls tumor growth through direct DNA damage and the influx of immune cells, reshaping the makeup of the tumor microenvironment. The relationship between RT and complement activity in the tumor microenvironment is uncertain at best. The following review will focus on the complex interaction of complement activation and the immune-modulating effects of RT and the overall effect on tumor progression. The clinical implications of complement activation in cancer and the use of therapeutics and potential biomarkers will also be covered.
Collapse
|
122
|
Hew BE, Pangburn MK, Vogel CW, Fritzinger DC. Identification of intermolecular bonds between human factor B and Cobra Venom Factor important for C3 convertase stability. Toxicon 2020; 184:68-77. [PMID: 32526239 DOI: 10.1016/j.toxicon.2020.05.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 05/28/2020] [Accepted: 05/31/2020] [Indexed: 10/24/2022]
Abstract
Cobra venom factor (CVF) is the complement-activating protein in cobra venom. CVF is a structural and functional analog of complement component C3. CVF, like C3b, forms a convertase with factor B. This bimolecular complex CVF, Bb is an enzyme that cleaves C3 and C5. However, CVF, Bb exhibits significantly different functional properties from C3b,Bb. Whereas both, CVF, Bb and C3b, Bb exhibit spontaneous decay-dissociation into the respective subunits, thereby eliminating the enzymatic activity, the CVF, Bb convertase is physico-chemically far more stable, decaying with a half-life that is more than two orders of magnitude slower than that of C3b,Bb. In addition, CVF, Bb is completely resistant to inactivation by Factors H and I. These two properties of CVF, Bb allow continuous activation of C3 and C5, and complement depletion in serum. In order to understand the structural basis for the physico-chemical stability of CVF,Bb, we have created recombinant hybrid proteins of CVF and human C3, based on structural differences between CVF and human C3b in the C-terminal C345C domain. Here we describe three human C3/CVF hybrid proteins which differ in only one, two, or five amino acid residues from earlier described hybrid proteins. In all three cases, the hybrid proteins containing CVF residues form more stable convertases, and exhibit stronger complement-depletion activity than hybrid proteins with human C3 residues. Three bonds between CVF residues and Factor Bb residues could be identified by crystallographic modeling that contribute to the greater stability of the convertases.
Collapse
Affiliation(s)
- Brian E Hew
- University of Hawaii Cancer Center, University of Hawaii at Manoa, 701 Ilalo Street, Honolulu, HI, 96813, USA
| | - Michael K Pangburn
- Biomedical Research Department, University of Texas Health Science Center, Tyler, TX, 75708, USA
| | - Carl-Wilhelm Vogel
- University of Hawaii Cancer Center, University of Hawaii at Manoa, 701 Ilalo Street, Honolulu, HI, 96813, USA; Department of Pathology, John A. Burns School of Medicine, University of Hawaii at Manoa, 651 Ilalo Street, Honolulu, HI, 96813, USA.
| | - David C Fritzinger
- University of Hawaii Cancer Center, University of Hawaii at Manoa, 701 Ilalo Street, Honolulu, HI, 96813, USA
| |
Collapse
|
123
|
A novel methodology of the myeloid-derived suppressor cells (MDSCs) generation with splenic stroma feeder cells. Exp Cell Res 2020; 394:112119. [PMID: 32485182 DOI: 10.1016/j.yexcr.2020.112119] [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] [Received: 01/21/2020] [Revised: 05/21/2020] [Accepted: 05/28/2020] [Indexed: 01/17/2023]
Abstract
Myeloid-derived suppressor cells (MDSCs) are a significant obstacle for immunotherapy of cancer. It is of great clinical relevance to study the mechanism of MDSCs accumulation in mouse spleens and establish a stable method to obtain high-purity MDSCs in vitro for further research. Here, we established a new method for amplifying a large number of highly pure MDSCs in vitro. To mimic the microenvironment of MDSCs development in vivo, mouse splenic stroma feeder cells and serum-free medium containing granulocyte-macrophage colony stimulating factor (GM-CSF) were used to induce myeloid precursors in mouse bone marrow cells, which differentiate into MDSCs. Development and immunological functions of the cells were monitored both in vivo and in vitro. A total of 4 × 108 MDSCs could be obtained from the bone marrow from one mouse, the ratio of CD11b+Gr-1+ MDSCs could reach 93.8% ± 3.3% after nine days of culture in vitro. Cultured MDSCs maintained a similar immunophenotype with MDSCs found in tumor-bearing mice. Colony forming assay in vitro and in vivo demonstrated that these were myeloid precursor cells. These cells generated high levels of reactive oxygen species and arginase 1 to prevent proliferation of CD8+ T cells in vitro. These also increased regulatory T (Treg) cells in blood while promoting the growth of lymphoma in vivo. In addition, cultured MDSCs effectively inhibited acute graft-versus-host disease (aGVHD). Our findings suggest that mouse splenic stroma plays an important role in the generation of MDSCs and represent a preliminary mechanism for the accumulation of MDSCs in spleens, and thereby lay the foundation for basic research and the clinical application of MDSCs.
Collapse
|
124
|
Golay J, Andrea AE. Combined Anti-Cancer Strategies Based on Anti-Checkpoint Inhibitor Antibodies. Antibodies (Basel) 2020; 9:E17. [PMID: 32443877 PMCID: PMC7345008 DOI: 10.3390/antib9020017] [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: 04/20/2020] [Revised: 05/11/2020] [Accepted: 05/12/2020] [Indexed: 12/14/2022] Open
Abstract
Therapeutic monoclonal antibodies for the treatment of cancer came of age in 1997, with the approval of anti-CD20 Rituximab. Since then, a wide variety of antibodies have been developed with many different formats and mechanisms of action. Among these, antibodies blocking immune checkpoint inhibitors (ICI) have revolutionized the field, based on the novelty of their concept and their demonstrated efficacy in several types of cancer otherwise lacking effective immunotherapy approaches. ICI are expressed by tumor, stromal or immune cells infiltrating the tumor microenvironment, and negatively regulate anti-tumor immunity. Antibodies against the first discovered ICI, CTLA-4, PD-1 and PD-L1, have shown significant activity in phase III studies against melanoma and other solid cancers, alone or in combination with chemotherapy or radiotherapy. However, not all cancers and not all patients respond to these drugs. Therefore, novel antibodies targeting additional ICI are currently being developed. In addition, CTLA-4, PD-1 and PD-L1 blocking antibodies are being combined with each other or with other antibodies targeting novel ICI, immunostimulatory molecules, tumor antigens, angiogenic factors, complement receptors, or with T cell engaging bispecific antibodies (BsAb), with the aim of obtaining synergistic effects with minimal toxicity. In this review, we summarize the biological aspects behind such combinations and review some of the most important clinical data on ICI-specific antibodies.
Collapse
Affiliation(s)
- Josée Golay
- Center of Cellular Therapy “G. Lanzani”, UOC Ematologia, Azienda Socio Sanitaria Territoriale Papa Giovanni XXIII, 24127 Bergamo, Italy
- Fondazione per la Ricerca Ospedale Maggiore, 24127 Bergamo, Italy
| | - Alain E. Andrea
- Laboratoire de Biochimie et Thérapies Moléculaires, Faculté de Pharmacie, Université Saint Joseph de Beyrouth, Beirut 1100, Lebanon;
| |
Collapse
|
125
|
A Network Pharmacology-Based Study on the Anti-Lung Cancer Effect of Dipsaci Radix. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2020; 2020:7424061. [PMID: 32419823 PMCID: PMC7204368 DOI: 10.1155/2020/7424061] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Revised: 03/31/2020] [Accepted: 04/08/2020] [Indexed: 12/17/2022]
Abstract
Objective Dipsaci Radix (DR) has been used to treat fracture and osteoporosis. Recent reports have shown that myeloid cells from bone marrow can promote the proliferation of lung cancer. However, the action and mechanism of DR has not been well defined in lung cancer. The aim of the present study was to define molecular mechanisms of DR as a potential therapeutic approach to treat lung cancer. Methods Active compounds of DR with oral bioavailability ≥30% and drug-likeness index ≥0.18 were obtained from the traditional Chinese medicine systems pharmacology database and analysis platform. The potential target genes of the active compounds and bone were identified by PharmMapper and GeneCards, respectively. The compound-target network and protein-protein interaction network were built by Cytoscape software and Search Tool for the Retrieval of Interacting Genes webserver, respectively. GO analysis and pathway enrichment analysis were performed using R software. Results Our study demonstrated that DR had 6 active compounds, including gentisin, sitosterol, Sylvestroside III, 3,5-Di-O-caffeoylquinic acid, cauloside A, and japonine. There were 254 target genes related to these active compounds as well as to bone. SRC, AKT1, and GRB2 were the top 3 hub genes. Metabolisms and signaling pathways associated with these hub genes were significantly enriched. Conclusions This study indicated that DR could exhibit the anti-lung cancer effect by affecting multiple targets and multiple pathways. It reflects the traditional Chinese medicine characterized by multicomponents and multitargets. DR could be considered as a candidate for clinical anticancer therapy by regulating bone physiological functions.
Collapse
|
126
|
Davidson S, Efremova M, Riedel A, Mahata B, Pramanik J, Huuhtanen J, Kar G, Vento-Tormo R, Hagai T, Chen X, Haniffa MA, Shields JD, Teichmann SA. Single-Cell RNA Sequencing Reveals a Dynamic Stromal Niche That Supports Tumor Growth. Cell Rep 2020; 31:107628. [PMID: 32433953 PMCID: PMC7242909 DOI: 10.1016/j.celrep.2020.107628] [Citation(s) in RCA: 187] [Impact Index Per Article: 37.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Revised: 01/22/2020] [Accepted: 04/18/2020] [Indexed: 12/20/2022] Open
Abstract
Here, using single-cell RNA sequencing, we examine the stromal compartment in murine melanoma and draining lymph nodes (LNs) at points across tumor development, providing data at http://www.teichlab.org/data/. Naive lymphocytes from LNs undergo activation and clonal expansion within the tumor, before PD1 and Lag3 expression, while tumor-associated myeloid cells promote the formation of a suppressive niche. We identify three temporally distinct stromal populations displaying unique functional signatures, conserved across mouse and human tumors. Whereas "immune" stromal cells are observed in early tumors, "contractile" cells become more prevalent at later time points. Complement component C3 is specifically expressed in the immune population. Its cleavage product C3a supports the recruitment of C3aR+ macrophages, and perturbation of C3a and C3aR disrupts immune infiltration, slowing tumor growth. Our results highlight the power of scRNA-seq to identify complex interplays and increase stromal diversity as a tumor develops, revealing that stromal cells acquire the capacity to modulate immune landscapes from early disease.
Collapse
Affiliation(s)
- Sarah Davidson
- Medical Research Council Cancer Unit, University of Cambridge, Hutchison/Medical Research Council Research Centre, Box 197 Cambridge Biomedical Campus, Cambridge, CB2 0XZ, UK
| | - Mirjana Efremova
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge CB10 1SA, UK
| | - Angela Riedel
- Medical Research Council Cancer Unit, University of Cambridge, Hutchison/Medical Research Council Research Centre, Box 197 Cambridge Biomedical Campus, Cambridge, CB2 0XZ, UK
| | - Bidesh Mahata
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge CB10 1SA, UK; Department of Pathology, University of Cambridge, Cambridge, UK
| | - Jhuma Pramanik
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge CB10 1SA, UK
| | - Jani Huuhtanen
- Hematology Research Unit Helsinki, Department of Clinical Chemistry and Hematology, University of Helsinki and Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland
| | - Gozde Kar
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge CB10 1SA, UK
| | - Roser Vento-Tormo
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge CB10 1SA, UK
| | - Tzachi Hagai
- School of Molecular Cell Biology and Biotechnology, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978, Israel
| | - Xi Chen
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge CB10 1SA, UK
| | - Muzlifah A Haniffa
- Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
| | - Jacqueline D Shields
- Medical Research Council Cancer Unit, University of Cambridge, Hutchison/Medical Research Council Research Centre, Box 197 Cambridge Biomedical Campus, Cambridge, CB2 0XZ, UK.
| | - Sarah A Teichmann
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge CB10 1SA, UK; Cavendish Laboratory, University of Cambridge, JJ Thomson Ave, Cambridge CB3 0HE, UK.
| |
Collapse
|
127
|
Xu S, Gong Y, Yin Y, Xing H, Zhang N. The multiple function of long noncoding RNAs in osteosarcoma progression, drug resistance and prognosis. Biomed Pharmacother 2020; 127:110141. [PMID: 32334375 DOI: 10.1016/j.biopha.2020.110141] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2020] [Revised: 03/22/2020] [Accepted: 03/30/2020] [Indexed: 12/16/2022] Open
Abstract
Osteosarcoma is a bone tumor prevalent in children and young adults. LncRNAs are a family of non-protein-coding transcripts longer than 200 nucleotides. The tumor-related pathological functions of lncRNAs include proliferation, migration, and chemotherapy resistance, all of which have been widely acknowledged in research on osteosarcoma. In addition, compelling evidence suggests that lncRNAs could serve as diagnostic indicators, prognostic biomarkers, and targets for disease treatment. In this review, we systematically summarize how lncRNAs regulate tumorigenesis, invasion and therapeutic resistance. By deepening our knowledge of the relationship between lncRNAs and osteosarcoma, we hope to translate research findings into clinical applications as soon as possible.
Collapse
Affiliation(s)
- Shengjie Xu
- Department of Cardiology, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Yingchao Gong
- Department of Cardiology, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Ying Yin
- Department of Gastroenterology, Affiliated Zhongda Hospital of Southeast University, Nanjing, China
| | - Hongyuan Xing
- Department of Orthopedics, 2nd Affiliated Hospital, School of Medicine, Zhejiang University, Zhejiang, China
| | - Ning Zhang
- Department of Orthopedics, 2nd Affiliated Hospital, School of Medicine, Zhejiang University, Zhejiang, China.
| |
Collapse
|
128
|
Liu M, Tong Z, Ding C, Luo F, Wu S, Wu C, Albeituni S, He L, Hu X, Tieri D, Rouchka EC, Hamada M, Takahashi S, Gibb AA, Kloecker G, Zhang HG, Bousamra M, Hill BG, Zhang X, Yan J. Transcription factor c-Maf is a checkpoint that programs macrophages in lung cancer. J Clin Invest 2020; 130:2081-2096. [PMID: 31945018 PMCID: PMC7108920 DOI: 10.1172/jci131335] [Citation(s) in RCA: 118] [Impact Index Per Article: 23.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Accepted: 01/14/2020] [Indexed: 12/24/2022] Open
Abstract
Macrophages have been linked to tumor initiation, progression, metastasis, and treatment resistance. However, the transcriptional regulation of macrophages driving the protumor function remains elusive. Here, we demonstrate that the transcription factor c-Maf is a critical controller for immunosuppressive macrophage polarization and function in cancer. c-Maf controls many M2-related genes and has direct binding sites within a conserved noncoding sequence of the Csf-1r gene and promotes M2-like macrophage-mediated T cell suppression and tumor progression. c-Maf also serves as a metabolic checkpoint regulating the TCA cycle and UDP-GlcNAc biosynthesis, thus promoting M2-like macrophage polarization and activation. Additionally, c-Maf is highly expressed in tumor-associated macrophages (TAMs) and regulates TAM immunosuppressive function. Deletion of c-Maf specifically in myeloid cells results in reduced tumor burden with enhanced antitumor T cell immunity. Inhibition of c-Maf partly overcomes resistance to anti-PD-1 therapy in a subcutaneous LLC tumor model. Similarly, c-Maf is expressed in human M2 and tumor-infiltrating macrophages/monocytes as well as circulating monocytes of human non-small cell lung carcinoma (NSCLC) patients and critically regulates their immunosuppressive activity. The natural compound β-glucan downregulates c-Maf expression on macrophages, leading to enhanced antitumor immunity in mice. These findings establish a paradigm for immunosuppressive macrophage polarization and transcriptional regulation by c-Maf and suggest that c-Maf is a potential target for effective tumor immunotherapy.
Collapse
MESH Headings
- Animals
- Carcinoma, Non-Small-Cell Lung/genetics
- Carcinoma, Non-Small-Cell Lung/immunology
- Carcinoma, Non-Small-Cell Lung/pathology
- Carcinoma, Non-Small-Cell Lung/therapy
- Cell Line, Tumor
- Female
- Gene Expression Regulation, Neoplastic/immunology
- Humans
- Immunity, Cellular
- Lung Neoplasms/immunology
- Lung Neoplasms/pathology
- Lung Neoplasms/therapy
- Macrophage Activation
- Macrophages/immunology
- Macrophages/pathology
- Male
- Mice
- Mice, Knockout
- Monocytes/immunology
- Monocytes/pathology
- Neoplasms, Experimental/genetics
- Neoplasms, Experimental/immunology
- Neoplasms, Experimental/pathology
- Neoplasms, Experimental/therapy
- Proto-Oncogene Proteins c-maf/genetics
- Proto-Oncogene Proteins c-maf/immunology
- T-Lymphocytes/immunology
- T-Lymphocytes/pathology
Collapse
Affiliation(s)
- Min Liu
- Division of Immunotherapy, Hiram C. Polk, Jr., MD Department of Surgery, Immuno-Oncology Program, James Graham Brown Cancer Center, University of Louisville, Louisville, Kentucky, USA
| | - Zan Tong
- Division of Immunotherapy, Hiram C. Polk, Jr., MD Department of Surgery, Immuno-Oncology Program, James Graham Brown Cancer Center, University of Louisville, Louisville, Kentucky, USA
| | - Chuanlin Ding
- Division of Immunotherapy, Hiram C. Polk, Jr., MD Department of Surgery, Immuno-Oncology Program, James Graham Brown Cancer Center, University of Louisville, Louisville, Kentucky, USA
| | - Fengling Luo
- Division of Immunotherapy, Hiram C. Polk, Jr., MD Department of Surgery, Immuno-Oncology Program, James Graham Brown Cancer Center, University of Louisville, Louisville, Kentucky, USA
| | - Shouzhen Wu
- Division of Immunotherapy, Hiram C. Polk, Jr., MD Department of Surgery, Immuno-Oncology Program, James Graham Brown Cancer Center, University of Louisville, Louisville, Kentucky, USA
| | - Caijun Wu
- Division of Immunotherapy, Hiram C. Polk, Jr., MD Department of Surgery, Immuno-Oncology Program, James Graham Brown Cancer Center, University of Louisville, Louisville, Kentucky, USA
| | | | | | - Xiaoling Hu
- Division of Immunotherapy, Hiram C. Polk, Jr., MD Department of Surgery, Immuno-Oncology Program, James Graham Brown Cancer Center, University of Louisville, Louisville, Kentucky, USA
| | - David Tieri
- Department of Anatomical Sciences and Neurobiology, and
| | - Eric C. Rouchka
- Department of Computer Engineering and Computer Science, University of Louisville, Louisville, Kentucky, USA
| | - Michito Hamada
- Department of Anatomy and Embryology, University of Tsukuba, Ibaraki, Japan
| | - Satoru Takahashi
- Department of Anatomy and Embryology, University of Tsukuba, Ibaraki, Japan
| | | | - Goetz Kloecker
- Division of Immunotherapy, Hiram C. Polk, Jr., MD Department of Surgery, Immuno-Oncology Program, James Graham Brown Cancer Center, University of Louisville, Louisville, Kentucky, USA
| | - Huang-ge Zhang
- Department of Microbiology and Immunology, School of Medicine
| | - Michael Bousamra
- Department of Cardiovascular Thoracic Surgery, University of Louisville, Louisville, Kentucky, USA
| | | | | | - Jun Yan
- Division of Immunotherapy, Hiram C. Polk, Jr., MD Department of Surgery, Immuno-Oncology Program, James Graham Brown Cancer Center, University of Louisville, Louisville, Kentucky, USA
- Department of Microbiology and Immunology, School of Medicine
| |
Collapse
|
129
|
Lo MW, Woodruff TM. Complement: Bridging the innate and adaptive immune systems in sterile inflammation. J Leukoc Biol 2020; 108:339-351. [PMID: 32182389 DOI: 10.1002/jlb.3mir0220-270r] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Revised: 02/07/2020] [Accepted: 02/19/2020] [Indexed: 12/24/2022] Open
Abstract
The complement system is a collection of soluble and membrane-bound proteins that together act as a powerful amplifier of the innate and adaptive immune systems. Although its role in infection is well established, complement is becoming increasingly recognized as a key contributor to sterile inflammation, a chronic inflammatory process often associated with noncommunicable diseases. In this context, damaged tissues release danger signals and trigger complement, which acts on a range of leukocytes to augment and bridge the innate and adaptive immune systems. Given the detrimental effect of chronic inflammation, the complement system is therefore well placed as an anti-inflammatory drug target. In this review, we provide a general outline of the sterile activators, effectors, and targets of the complement system and a series of examples (i.e., hypertension, cancer, allograft transplant rejection, and neuroinflammation) that highlight complement's ability to bridge the 2 arms of the immune system.
Collapse
Affiliation(s)
- Martin W Lo
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, St Lucia, Brisbane, Queensland, Australia
| | - Trent M Woodruff
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, St Lucia, Brisbane, Queensland, Australia
| |
Collapse
|
130
|
Abstract
The recognition of microbial or danger-associated molecular patterns by complement proteins initiates a cascade of events that culminates in the activation of surface complement receptors on immune cells. Such signalling pathways converge with those activated downstream of pattern recognition receptors to determine the type and magnitude of the immune response. Intensive investigation in the field has uncovered novel pathways that link complement-mediated signalling with homeostatic and pathological T cell responses. More recently, the observation that complement proteins also act in the intracellular space to shape T cell fates has added a new layer of complexity. Here, we consider fundamental mechanisms and novel concepts at the interface of complement biology and immunity and discuss how these affect the maintenance of homeostasis and the development of human pathology.
Collapse
|
131
|
Guan B, Zhang X. Aptamers as Versatile Ligands for Biomedical and Pharmaceutical Applications. Int J Nanomedicine 2020; 15:1059-1071. [PMID: 32110008 PMCID: PMC7035142 DOI: 10.2147/ijn.s237544] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Accepted: 01/10/2020] [Indexed: 12/14/2022] Open
Abstract
Aptamers are a class of targeting ligands that bind exclusively to biomarkers of interest. Aptamers have been identified as candidates for the construction of various smart systems for therapy, diagnosis, bioimaging, and drug delivery due to their high target affinity and specificity. Aptamers are accounted as chemical antibodies that can be readily linked to drugs, sensors, signal enhancers, or nanocarriers for functionalization. Use of aptamer-guided medications, especially nanomedicines, has resulted in encouraging outcomes compared to those use of aptamer-free counterparts. This article reviews recent advances in the use of aptamers as targeting ligands for various biomedical and pharmaceutical purposes. Special interests focus on aptamer-based theranostics, biosensing, bioimaging, drug potentiation, and targeted drug delivery.
Collapse
Affiliation(s)
- Baozhang Guan
- Department of Nephrology, The First Affiliated Hospital of Jinan University, Guangzhou 510630, People's Republic of China
| | - Xingwang Zhang
- Department of Pharmaceutics, College of Pharmacy, Jinan University, Guangzhou 510632, People's Republic of China
| |
Collapse
|
132
|
Short-term starvation reduces IGF-1 levels to sensitize lung tumors to PD-1 immune checkpoint blockade. ACTA ACUST UNITED AC 2020; 1:75-85. [DOI: 10.1038/s43018-019-0007-9] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Accepted: 11/14/2019] [Indexed: 01/31/2023]
|
133
|
Grigorieva J, Asmellash S, Oliveira C, Roder H, Net L, Roder J. Application of protein set enrichment analysis to correlation of protein functional sets with mass spectral features and multivariate proteomic tests. CLINICAL MASS SPECTROMETRY 2020. [DOI: 10.1016/j.clinms.2019.09.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
|
134
|
Roumenina LT, Daugan MV, Petitprez F, Sautès-Fridman C, Fridman WH. Context-dependent roles of complement in cancer. Nat Rev Cancer 2019; 19:698-715. [PMID: 31666715 DOI: 10.1038/s41568-019-0210-0] [Citation(s) in RCA: 230] [Impact Index Per Article: 38.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/14/2019] [Indexed: 12/16/2022]
Abstract
The tumour microenvironment (TME) highly influences the growth and spread of tumours, thus impacting the patient's clinical outcome. In this context, the complement system plays a major and complex role. It may either act to kill antibody-coated tumour cells, support local chronic inflammation or hamper antitumour T cell responses favouring tumour progression. Recent studies demonstrate that these opposing effects are dependent upon the sites of complement activation, the composition of the TME and the tumour cell sensitivity to complement attack. In this Review, we present the evidence that has so far accrued showing a role for complement activation and its effects on cancer control and clinical outcome under different TME contexts. We also include a new analysis of the publicly available transcriptomic data to provide an overview of the prognostic value of complement gene expression in 30 cancer types. We argue that the interplay of complement components within each cancer type is unique, governed by the properties of the tumour cells and the TME. This concept is of critical importance for the design of efficient therapeutic strategies aimed at targeting complement components and their signalling.
Collapse
Affiliation(s)
- Lubka T Roumenina
- INSERM, UMR_S 1138, Centre de Recherche des Cordeliers, Sorbonne Universités, Université de Paris, Paris, France.
| | - Marie V Daugan
- INSERM, UMR_S 1138, Centre de Recherche des Cordeliers, Sorbonne Universités, Université de Paris, Paris, France
| | - Florent Petitprez
- INSERM, UMR_S 1138, Centre de Recherche des Cordeliers, Sorbonne Universités, Université de Paris, Paris, France
- Programme Cartes d'Identité des Tumeurs, Ligue Nationale Contre le Cancer, Paris, France
| | - Catherine Sautès-Fridman
- INSERM, UMR_S 1138, Centre de Recherche des Cordeliers, Sorbonne Universités, Université de Paris, Paris, France
| | - Wolf Herman Fridman
- INSERM, UMR_S 1138, Centre de Recherche des Cordeliers, Sorbonne Universités, Université de Paris, Paris, France.
| |
Collapse
|
135
|
Abstract
New therapies that promote antitumour immunity have been recently developed. Most of these immunomodulatory approaches have focused on enhancing T-cell responses, either by targeting inhibitory pathways with immune checkpoint inhibitors, or by targeting activating pathways, as with chimeric antigen receptor T cells or bispecific antibodies. Although these therapies have led to unprecedented successes, only a minority of patients with cancer benefit from these treatments, highlighting the need to identify new cells and molecules that could be exploited in the next generation of immunotherapy. Given the crucial role of innate immune responses in immunity, harnessing these responses opens up new possibilities for long-lasting, multilayered tumour control.
Collapse
|
136
|
Lecot P, Sarabi M, Pereira Abrantes M, Mussard J, Koenderman L, Caux C, Bendriss-Vermare N, Michallet MC. Neutrophil Heterogeneity in Cancer: From Biology to Therapies. Front Immunol 2019; 10:2155. [PMID: 31616408 PMCID: PMC6764113 DOI: 10.3389/fimmu.2019.02155] [Citation(s) in RCA: 86] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Accepted: 08/28/2019] [Indexed: 12/12/2022] Open
Abstract
Neutrophils have been extensively described in the pathophysiology of autoimmune and infectious diseases. Accumulating evidence also suggests the important role of neutrophils in cancer progression through their interaction with cancer and immune cells in blood and in the tumor microenvironment (TME). Most studies have described neutrophils as key drivers of cancer progression, due to their involvement in various tumor promoting functions including proliferation, aggressiveness, and dissemination, as well as in immune suppression. However, such studies were focusing on late-stages of tumorigenesis, in which chronic inflammation had already developed. The role of tumor-associated neutrophils (TANs) at early stages of tumor development remains poorly described, though recent findings indicate that early-stage TANs may display anti-tumor properties. Beyond their role at tumor site, evidence supported by NLR retrospective studies and functional analyses suggest that blood neutrophils could also actively contribute to tumorigenesis. Hence, it appears that the phenotype and functions of neutrophils vary greatly during tumor progression, highlighting their heterogeneity. The origin of pro- or anti-tumor neutrophils is generally believed to arise following a change in cell state, from resting to activated. Moreover, the fate of neutrophils may also involve distinct differentiation programs yielding various subsets of pro or anti-tumor neutrophils. In this review, we will discuss the current knowledge on neutrophils heterogeneity across different tissues and their impact on tumorigenesis, as well as neutrophil-based therapeutic strategies that have shown promising results in pre-clinical studies, paving the way for the design of neutrophil-based next generation immunotherapy.
Collapse
Affiliation(s)
- Pacôme Lecot
- Department of Immunity, Virus, and Inflammation (IVI), Centre de Recherche en Cancérologie de Lyon, Centre Léon Bérard, University of Lyon, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Lyon, France
| | - Matthieu Sarabi
- Department of Immunity, Virus, and Inflammation (IVI), Centre de Recherche en Cancérologie de Lyon, Centre Léon Bérard, University of Lyon, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Lyon, France
| | - Manuela Pereira Abrantes
- Department of Immunity, Virus, and Inflammation (IVI), Centre de Recherche en Cancérologie de Lyon, Centre Léon Bérard, University of Lyon, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Lyon, France
| | - Julie Mussard
- Department of Immunity, Virus, and Inflammation (IVI), Centre de Recherche en Cancérologie de Lyon, Centre Léon Bérard, University of Lyon, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Lyon, France
| | - Leo Koenderman
- Department of Respiratory Medicine and Center of Translational Immunology, University Medical Center Utrecht, Utrecht, Netherlands
| | - Christophe Caux
- Department of Immunity, Virus, and Inflammation (IVI), Centre de Recherche en Cancérologie de Lyon, Centre Léon Bérard, University of Lyon, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Lyon, France
| | - Nathalie Bendriss-Vermare
- Department of Immunity, Virus, and Inflammation (IVI), Centre de Recherche en Cancérologie de Lyon, Centre Léon Bérard, University of Lyon, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Lyon, France
| | - Marie-Cécile Michallet
- Department of Immunity, Virus, and Inflammation (IVI), Centre de Recherche en Cancérologie de Lyon, Centre Léon Bérard, University of Lyon, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Lyon, France
| |
Collapse
|
137
|
Ajona D, Zandueta C, Corrales L, Moreno H, Pajares MJ, Ortiz-Espinosa S, Martínez-Terroba E, Perurena N, de Miguel FJ, Jantus-Lewintre E, Camps C, Vicent S, Agorreta J, Montuenga LM, Pio R, Lecanda F. Blockade of the Complement C5a/C5aR1 Axis Impairs Lung Cancer Bone Metastasis by CXCL16-mediated Effects. Am J Respir Crit Care Med 2019; 197:1164-1176. [PMID: 29327939 DOI: 10.1164/rccm.201703-0660oc] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
RATIONALE C5aR1 (CD88), a receptor for complement anaphylatoxin C5a, is a potent immune mediator. Its impact on malignant growth and dissemination of non-small cell lung cancer cells is poorly understood. OBJECTIVES To investigate the contribution of the C5a/C5aR1 axis to the malignant phenotype of non-small cell lung cancer cells, particularly in skeletal colonization, a preferential lung metastasis site. METHODS Association between C5aR1 expression and clinical outcome was assessed in silico and validated by immunohistochemistry. Functional significance was evaluated by lentiviral gene silencing and ligand l-aptamer inhibition in in vivo models of lung cancer bone metastasis. In vitro functional assays for signaling, migration, invasion, metalloprotease activity, and osteoclastogenesis were also performed. MEASUREMENTS AND MAIN RESULTS High levels of C5aR1 in human lung tumors were significantly associated with shorter recurrence-free survival, overall survival, and bone metastasis. Silencing of C5aR1 in lung cancer cells led to a substantial reduction in skeletal metastatic burden and osteolysis in in vivo models. Furthermore, metalloproteolytic, migratory, and invasive tumor cell activities were modulated in vitro by C5aR1 stimulation or gene silencing. l-Aptamer blockade or C5aR1 silencing significantly reduced the osseous metastatic activity of lung cancer cells in vivo. This effect was associated with decreased osteoclastogenic activity in vitro and was rescued by the exogenous addition of the chemokine CXCL16. CONCLUSIONS Disruption of C5aR1 signaling in lung cancer cells abrogates their tumor-associated osteoclastogenic activity, impairing osseous colonization. This study unveils the role played by the C5a/C5aR1 axis in lung cancer dissemination and supports its potential use as a novel therapeutic target.
Collapse
Affiliation(s)
- Daniel Ajona
- 1 Center for Applied Medical Research, Program in Solid Tumors and Biomarkers, Pamplona, Spain.,2 IdiSNA (Navarra Institute for Health Research), Pamplona, Spain.,3 CIBERONC (Centro de Investigación Biomédica en Red de Cáncer), Spain.,4 Department of Biochemistry and Genetics, School of Sciences, and
| | - Carolina Zandueta
- 1 Center for Applied Medical Research, Program in Solid Tumors and Biomarkers, Pamplona, Spain.,2 IdiSNA (Navarra Institute for Health Research), Pamplona, Spain.,3 CIBERONC (Centro de Investigación Biomédica en Red de Cáncer), Spain
| | - Leticia Corrales
- 1 Center for Applied Medical Research, Program in Solid Tumors and Biomarkers, Pamplona, Spain
| | - Haritz Moreno
- 1 Center for Applied Medical Research, Program in Solid Tumors and Biomarkers, Pamplona, Spain.,2 IdiSNA (Navarra Institute for Health Research), Pamplona, Spain
| | - María J Pajares
- 1 Center for Applied Medical Research, Program in Solid Tumors and Biomarkers, Pamplona, Spain.,2 IdiSNA (Navarra Institute for Health Research), Pamplona, Spain.,3 CIBERONC (Centro de Investigación Biomédica en Red de Cáncer), Spain.,5 Department of Histology and Pathology, School of Medicine, University of Navarra, Pamplona, Spain
| | - Sergio Ortiz-Espinosa
- 1 Center for Applied Medical Research, Program in Solid Tumors and Biomarkers, Pamplona, Spain.,3 CIBERONC (Centro de Investigación Biomédica en Red de Cáncer), Spain.,4 Department of Biochemistry and Genetics, School of Sciences, and
| | - Elena Martínez-Terroba
- 1 Center for Applied Medical Research, Program in Solid Tumors and Biomarkers, Pamplona, Spain.,5 Department of Histology and Pathology, School of Medicine, University of Navarra, Pamplona, Spain
| | - Naiara Perurena
- 1 Center for Applied Medical Research, Program in Solid Tumors and Biomarkers, Pamplona, Spain
| | - Fernando J de Miguel
- 1 Center for Applied Medical Research, Program in Solid Tumors and Biomarkers, Pamplona, Spain.,4 Department of Biochemistry and Genetics, School of Sciences, and
| | - Eloisa Jantus-Lewintre
- 3 CIBERONC (Centro de Investigación Biomédica en Red de Cáncer), Spain.,6 Molecular Oncology Laboratory, Fundación Investigación, Hospital General Universitario de Valencia, Valencia, Spain.,7 Department of Biotechnology, Universitat Politècnica de València, Valencia, Spain
| | - Carlos Camps
- 3 CIBERONC (Centro de Investigación Biomédica en Red de Cáncer), Spain.,6 Molecular Oncology Laboratory, Fundación Investigación, Hospital General Universitario de Valencia, Valencia, Spain.,8 Department of Medical Oncology, Hospital General Universitario de Valencia, Valencia, Spain; and.,9 Department of Medicine, Universitat de València, Valencia, Spain
| | - Silvestre Vicent
- 1 Center for Applied Medical Research, Program in Solid Tumors and Biomarkers, Pamplona, Spain.,2 IdiSNA (Navarra Institute for Health Research), Pamplona, Spain.,3 CIBERONC (Centro de Investigación Biomédica en Red de Cáncer), Spain.,5 Department of Histology and Pathology, School of Medicine, University of Navarra, Pamplona, Spain
| | - Jackeline Agorreta
- 1 Center for Applied Medical Research, Program in Solid Tumors and Biomarkers, Pamplona, Spain.,2 IdiSNA (Navarra Institute for Health Research), Pamplona, Spain.,3 CIBERONC (Centro de Investigación Biomédica en Red de Cáncer), Spain.,5 Department of Histology and Pathology, School of Medicine, University of Navarra, Pamplona, Spain
| | - Luis M Montuenga
- 1 Center for Applied Medical Research, Program in Solid Tumors and Biomarkers, Pamplona, Spain.,2 IdiSNA (Navarra Institute for Health Research), Pamplona, Spain.,3 CIBERONC (Centro de Investigación Biomédica en Red de Cáncer), Spain.,5 Department of Histology and Pathology, School of Medicine, University of Navarra, Pamplona, Spain
| | - Ruben Pio
- 1 Center for Applied Medical Research, Program in Solid Tumors and Biomarkers, Pamplona, Spain.,2 IdiSNA (Navarra Institute for Health Research), Pamplona, Spain.,3 CIBERONC (Centro de Investigación Biomédica en Red de Cáncer), Spain.,4 Department of Biochemistry and Genetics, School of Sciences, and
| | - Fernando Lecanda
- 1 Center for Applied Medical Research, Program in Solid Tumors and Biomarkers, Pamplona, Spain.,2 IdiSNA (Navarra Institute for Health Research), Pamplona, Spain.,3 CIBERONC (Centro de Investigación Biomédica en Red de Cáncer), Spain.,5 Department of Histology and Pathology, School of Medicine, University of Navarra, Pamplona, Spain
| |
Collapse
|
138
|
Prolonged Remission of Cancer of Unknown Primary following Initiation of Eculizumab Therapy for Paroxysmal Nocturnal Hemoglobinuria. Case Rep Hematol 2019; 2019:2587597. [PMID: 31355021 PMCID: PMC6636445 DOI: 10.1155/2019/2587597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Revised: 05/16/2019] [Accepted: 06/19/2019] [Indexed: 11/17/2022] Open
Abstract
We report the case of a 64-year-old woman who presented with cancer of unknown primary treated with carboplatin and paclitaxel, followed by maintenance erlotinib. Her chemotherapy regimen was discontinued due to the development of profound hemolysis that was later identified to be due to paroxysmal nocturnal hemoglobinuria (PNH). She was started on a complement inhibitory antibody, eculizumab 900 mg every 2 weeks, with marked suppression of hemolysis. Eight years after diagnosis of cancer, the patient remains on eculizumab with no signs of cancer recurrence on regular imaging. Regardless of whether the co-occurrence of cancer and PNH was any more than coincidental in this patient, the uniqueness of the case is emphasized by the remarkable and sustained response of not only PNH but also possibly the associated cancer to eculizumab.
Collapse
|
139
|
Clinical promise of next-generation complement therapeutics. Nat Rev Drug Discov 2019; 18:707-729. [PMID: 31324874 DOI: 10.1038/s41573-019-0031-6] [Citation(s) in RCA: 240] [Impact Index Per Article: 40.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/13/2019] [Indexed: 02/07/2023]
Abstract
The complement system plays a key role in pathogen immunosurveillance and tissue homeostasis. However, subversion of its tight regulatory control can fuel a vicious cycle of inflammatory damage that exacerbates pathology. The clinical merit of targeting the complement system has been established for rare clinical disorders such as paroxysmal nocturnal haemoglobinuria and atypical haemolytic uraemic syndrome. Evidence from preclinical studies and human genome-wide analyses, supported by new molecular and structural insights, has revealed new pathomechanisms and unmet clinical needs that have thrust a new generation of complement inhibitors into clinical development for a variety of indications. This review critically discusses recent clinical milestones in complement drug discovery, providing an updated translational perspective that may guide optimal target selection and disease-tailored complement intervention.
Collapse
|
140
|
Riihilä P, Nissinen L, Knuutila J, Rahmati Nezhad P, Viiklepp K, Kähäri VM. Complement System in Cutaneous Squamous Cell Carcinoma. Int J Mol Sci 2019; 20:ijms20143550. [PMID: 31331124 PMCID: PMC6678994 DOI: 10.3390/ijms20143550] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Revised: 07/16/2019] [Accepted: 07/17/2019] [Indexed: 12/16/2022] Open
Abstract
Epidermal keratinocyte-derived cutaneous squamous cell carcinoma (cSCC) is the most common metastatic skin cancer with high mortality rates in the advanced stage. Chronic inflammation is a recognized risk factor for cSCC progression and the complement system, as a part of innate immunity, belongs to the microenvironment of tumors. The complement system is a double-edged sword in cancer, since complement activation is involved in anti-tumor cytotoxicity and immune responses, but it also promotes cancer progression directly and indirectly. Recently, the role of several complement components and inhibitors in the regulation of progression of cSCC has been shown. In this review, we will discuss the role of complement system components and inhibitors as biomarkers and potential new targets for therapeutic intervention in cSCC.
Collapse
Affiliation(s)
- Pilvi Riihilä
- Department of Dermatology, University of Turku and Turku University Hospital, Hämeentie 11 TE6, FI-20520 Turku, Finland
- The Western Cancer Centre of the Cancer Center Finland (FICAN West), University of Turku and Turku University Hospital, Kiinamyllynkatu 10, FI-20520 Turku, Finland
| | - Liisa Nissinen
- Department of Dermatology, University of Turku and Turku University Hospital, Hämeentie 11 TE6, FI-20520 Turku, Finland
- The Western Cancer Centre of the Cancer Center Finland (FICAN West), University of Turku and Turku University Hospital, Kiinamyllynkatu 10, FI-20520 Turku, Finland
| | - Jaakko Knuutila
- Department of Dermatology, University of Turku and Turku University Hospital, Hämeentie 11 TE6, FI-20520 Turku, Finland
- The Western Cancer Centre of the Cancer Center Finland (FICAN West), University of Turku and Turku University Hospital, Kiinamyllynkatu 10, FI-20520 Turku, Finland
| | - Pegah Rahmati Nezhad
- Department of Dermatology, University of Turku and Turku University Hospital, Hämeentie 11 TE6, FI-20520 Turku, Finland
- The Western Cancer Centre of the Cancer Center Finland (FICAN West), University of Turku and Turku University Hospital, Kiinamyllynkatu 10, FI-20520 Turku, Finland
| | - Kristina Viiklepp
- Department of Dermatology, University of Turku and Turku University Hospital, Hämeentie 11 TE6, FI-20520 Turku, Finland
- The Western Cancer Centre of the Cancer Center Finland (FICAN West), University of Turku and Turku University Hospital, Kiinamyllynkatu 10, FI-20520 Turku, Finland
| | - Veli-Matti Kähäri
- Department of Dermatology, University of Turku and Turku University Hospital, Hämeentie 11 TE6, FI-20520 Turku, Finland.
- The Western Cancer Centre of the Cancer Center Finland (FICAN West), University of Turku and Turku University Hospital, Kiinamyllynkatu 10, FI-20520 Turku, Finland.
| |
Collapse
|
141
|
Wang Y, Zhang H, He YW. The Complement Receptors C3aR and C5aR Are a New Class of Immune Checkpoint Receptor in Cancer Immunotherapy. Front Immunol 2019; 10:1574. [PMID: 31379815 PMCID: PMC6658873 DOI: 10.3389/fimmu.2019.01574] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Accepted: 06/24/2019] [Indexed: 01/11/2023] Open
Abstract
Cancer immunotherapy has made remarkable clinical advances in recent years. Antibodies targeting the immune checkpoint receptors PD-1 and CTLA-4 and adoptive cell therapy (ACT) based on ex vivo expanded peripheral CTLs, tumor infiltrating lymphocytes (TILs), gene-engineered TCR- and chimeric antigen receptor (CAR)-T cells have all shown durable clinical efficacies in multiple types of cancers. However, these immunotherapeutic approaches only benefit a small fraction of cancer patients as various immune resistance mechanisms and limitations make their effective use a challenge in the majority of cancer patients. For example, adaptive resistance to therapeutic PD-1 blockade is associated with an upregulation of some additional immune checkpoint receptors. The efficacy of transferred tumor-specific T cells under the current clinical ACT protocol is often limited by their inefficient engraftment, poor persistence, and weak capability to attack tumor cells. Recent studies demonstrate that the complement receptor C3aR and C5aR function as a new class of immune checkpoint receptors. Complement signaling through C3aR and C5aR expressed on effector T lymphocytes prevent the production of the cytokine interleukin-10 (IL-10). Removing C3aR/C5aR-mediated transcriptional suppression of IL-10 expression results in endogenous IL-10 production by antitumor effector T cells, which drives T cell expansion and enhances T cell-mediated antitumor immunity. Importantly, preclinical, and clinical data suggest that a signaling axis consisting of complement/C3aR/C5aR/IL-10 critically regulates T cell mediated antitumor immunity and manipulation of the pathway ex vivo and in vivo is an effective strategy for cancer immunotherapy. Furthermore, a combination of treatment strategies targeting the complement/C3aR/C5aR/IL-10 pathway with other treatment modalities may improve cancer therapeutic efficacy.
Collapse
Affiliation(s)
- Yu Wang
- Life Science Institute, Jinzhou Medical University, Jinzhou, China
| | - Hui Zhang
- First Affiliated Hospital, China Medical University, Shenyang, China
| | - You-Wen He
- Department of Immunology, Duke University Medical Center, Durham, NC, United States
| |
Collapse
|
142
|
Nabizadeh JA, Manthey HD, Panagides N, Steyn FJ, Lee JD, Li XX, Akhir FNM, Chen W, Boyle GM, Taylor SM, Woodruff TM, Rolfe BE. C5a receptors C5aR1 and C5aR2 mediate opposing pathologies in a mouse model of melanoma. FASEB J 2019; 33:11060-11071. [PMID: 31298935 DOI: 10.1096/fj.201800980rr] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The canonical complement component 5a (C5a) receptor (C5aR) 1 has well-described roles in tumorigenesis but the contribution of the second receptor, C5aR2, is unclear. The present study demonstrates that B16.F0 melanoma cells express mRNA for both C5aR1 and C5aR2 and signal through ERK and p38 MAPKs in response to C5a. Despite this, C5a had no impact on melanoma cell proliferation or migration in vitro. In vivo studies demonstrated that the growth of B16.F0 melanoma tumors was increased in C5aR2-/- mice but reduced in C5aR1-/- mice and wild-type mice treated with a C5aR1 antagonist. Analysis of tumor-infiltrating leukocyte populations showed no significant differences between wild-type and C5aR2-/- mice. Conversely, percentages of myeloid-derived suppressor cells, macrophages, and regulatory T lymphocytes were lower in tumors from C5aR1-/- mice, whereas total (CD3+) T lymphocytes and CD4+ subsets were higher. Analysis of cytokine and chemokine levels also showed plasma IFN-γ was higher and tumor C-C motif chemokine ligand 2 was lower in the absence of C5aR1. The results suggest that C5aR1 signaling supports melanoma growth by promoting infiltration of immunosuppressive leukocyte populations into the tumor microenvironment, whereas C5aR2 has a more restricted but beneficial role in limiting tumor growth. Overall, these data support the potential of C5aR1-inhibitory therapies for melanoma.-Nabizadeh, J. A., Manthey, H. D., Panagides, N., Steyn, F. J., Lee, J. D., Li, X. X., Akhir, F. N. M., Chen, W., Boyle, G. M., Taylor, S. M., Woodruff, T. M., Rolfe, B. E. C5a receptors C5aR1 and C5aR2 mediate opposing pathologies in a mouse model of melanoma.
Collapse
Affiliation(s)
- Jamileh A Nabizadeh
- Australian Institute of Bioengineering and Nanotechnology, The University of Queensland, St. Lucia, Brisbane, Queensland, Australia
| | - Helga D Manthey
- Australian Institute of Bioengineering and Nanotechnology, The University of Queensland, St. Lucia, Brisbane, Queensland, Australia
| | - Nadya Panagides
- Australian Institute of Bioengineering and Nanotechnology, The University of Queensland, St. Lucia, Brisbane, Queensland, Australia
| | - Frederik J Steyn
- Centre for Clinical Research, The University of Queensland, St. Lucia, Brisbane, Queensland, Australia
| | - John D Lee
- School of Biomedical Science, The University of Queensland, St. Lucia, Brisbane, Queensland, Australia
| | - Xaria X Li
- School of Biomedical Science, The University of Queensland, St. Lucia, Brisbane, Queensland, Australia
| | - Fazrena N M Akhir
- Australian Institute of Bioengineering and Nanotechnology, The University of Queensland, St. Lucia, Brisbane, Queensland, Australia
| | - Weiyu Chen
- Australian Institute of Bioengineering and Nanotechnology, The University of Queensland, St. Lucia, Brisbane, Queensland, Australia
| | - Glen M Boyle
- Queensland Institute of Medical Research (QIMR) Berghofer Medical Research Institute, Herston, Brisbane, Queensland, Australia
| | - Stephen M Taylor
- School of Biomedical Science, The University of Queensland, St. Lucia, Brisbane, Queensland, Australia
| | - Trent M Woodruff
- School of Biomedical Science, The University of Queensland, St. Lucia, Brisbane, Queensland, Australia
| | - Barbara E Rolfe
- Australian Institute of Bioengineering and Nanotechnology, The University of Queensland, St. Lucia, Brisbane, Queensland, Australia
| |
Collapse
|
143
|
Mao Y, Xue P, Li L, Xu P, Cai Y, Chu X, Jiang P, Zhu S. Bioinformatics analysis of mRNA and miRNA microarray to identify the key miRNA‑gene pairs in small‑cell lung cancer. Mol Med Rep 2019; 20:2199-2208. [PMID: 31257520 PMCID: PMC6691276 DOI: 10.3892/mmr.2019.10441] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Accepted: 06/06/2019] [Indexed: 02/07/2023] Open
Abstract
Small-cell lung cancer (SCLC) is a type of lung cancer with early metastasis, and high recurrence and mortality rates. The molecular mechanism is still unclear and further research is required. The aim of the present study was to examine the pathogenesis and potential molecular markers of SCLC by comparing the differential expression of mRNA and microRNA (miRNA) between SCLC tissue and normal lung tissue. A transcriptome sequencing dataset (GSE6044) and a non-coding RNA sequence dataset (GSE19945) were downloaded from the Gene Expression Omnibus (GEO) database. In total, 451 differentially expressed genes (DEGs) and 134 differentially expressed miRNAs (DEMs) were identified using the R limma software package and the GEO2R tool of the GEO, respectively. The Gene Ontology function was significantly enriched for 28 terms, and the Kyoto Encyclopedia of Genes and Genomes database had 19 enrichment pathways, mainly related to ‘cell cycle’, ‘DNA replication’ and ‘oocyte meiosis mismatch repair’. The protein-protein interaction network was constructed using Cytoscape software to identify the molecular mechanisms of key signaling pathways and cellular activities in SCLC. The 1,402 miRNA-gene pairs encompassed 602 target genes of the DEMs using miRNAWalk, which is a bioinformatics platform that predicts DEM target genes and miRNA-gene pairs. There were 19 overlapping genes regulated by 32 miRNAs between target genes of the DEMs and DEGs. Bioinformatics analysis may help to better understand the role of DEGs, DEMs and miRNA-gene pairs in cell proliferation and signal transduction. The related hub genes may be used as biomarkers for the diagnosis and prognosis of SCLC, and as potential drug targets.
Collapse
Affiliation(s)
- Yun Mao
- Graduate School, Beijing University of Chinese Medicine, Beijing 100029, P.R. China
| | - Peng Xue
- Department of Oncology, Wangjing Hospital, China Academy of Chinese Medical Sciences, Beijing 100102, P.R. China
| | - Linlu Li
- Graduate School, Beijing University of Chinese Medicine, Beijing 100029, P.R. China
| | - Pengpeng Xu
- Graduate School, Beijing University of Chinese Medicine, Beijing 100029, P.R. China
| | - Yafang Cai
- Graduate School, Beijing University of Chinese Medicine, Beijing 100029, P.R. China
| | - Xuelei Chu
- Graduate School, Beijing University of Chinese Medicine, Beijing 100029, P.R. China
| | - Pengyuan Jiang
- Graduate School, Beijing University of Chinese Medicine, Beijing 100029, P.R. China
| | - Shijie Zhu
- Department of Oncology, Wangjing Hospital, China Academy of Chinese Medical Sciences, Beijing 100102, P.R. China
| |
Collapse
|
144
|
Geller A, Yan J. The Role of Membrane Bound Complement Regulatory Proteins in Tumor Development and Cancer Immunotherapy. Front Immunol 2019; 10:1074. [PMID: 31164885 PMCID: PMC6536589 DOI: 10.3389/fimmu.2019.01074] [Citation(s) in RCA: 95] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Accepted: 04/26/2019] [Indexed: 12/17/2022] Open
Abstract
It has long been understood that the control and surveillance of tumors within the body involves an intricate dance between the adaptive and innate immune systems. At the center of the interplay between the adaptive and innate immune response sits the complement system—an evolutionarily ancient response that aids in the destruction of microorganisms and damaged cells, including cancer cells. Membrane-bound complement regulatory proteins (mCRPs), such as CD46, CD55, and CD59, are expressed throughout the body in order to prevent over-activation of the complement system. These mCRPs act as a double-edged sword however, as they can also over-regulate the complement system to the extent that it is no longer effective at eliminating cancerous cells. Recent studies are now indicating that mCRPs may function as a biomarker of a malignant transformation in numerous cancer types, and further, are being shown to interfere with anti-tumor treatments. This highlights the critical roles that therapeutic blockade of mCRPs can play in cancer treatment. Furthermore, with the complement system having the ability to both directly and indirectly control adaptive T-cell responses, the use of a combinatorial approach of complement-related therapy along with other T-cell activating therapies becomes a logical approach to treatment. This review will highlight the biomarker-related role that mCRP expression may have in the classification of tumor phenotype and predicted response to different anti-cancer treatments in the context of an emerging understanding that complement activation within the Tumor Microenvironment (TME) is actually harmful for tumor control. We will discuss what is known about complement activation and mCRPs relating to cancer and immunotherapy, and will examine the potential for combinatorial approaches of anti-mCRP therapy with other anti-tumor therapies, especially checkpoint inhibitors such as anti PD-1 and PD-L1 monoclonal antibodies (mAbs). Overall, mCRPs play an essential role in the immune response to tumors, and understanding their role in the immune response, particularly in modulating currently used cancer therapeutics may lead to better clinical outcomes in patients with diverse cancer types.
Collapse
Affiliation(s)
- Anne Geller
- Department of Microbiology and Immunology, University of Louisville School of Medicine, Louisville, KY, United States
| | - Jun Yan
- Immuno-Oncology Program, Department of Medicine, The James Graham Brown Cancer Center, University of Louisville School of Medicine, Louisville, KY, United States
| |
Collapse
|
145
|
Kleczko EK, Kwak JW, Schenk EL, Nemenoff RA. Targeting the Complement Pathway as a Therapeutic Strategy in Lung Cancer. Front Immunol 2019; 10:954. [PMID: 31134065 PMCID: PMC6522855 DOI: 10.3389/fimmu.2019.00954] [Citation(s) in RCA: 81] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Accepted: 04/15/2019] [Indexed: 12/20/2022] Open
Abstract
Lung cancer is the leading cause of cancer death in men and women. Lung adenocarcinoma (LUAD), represents approximately 40% of all lung cancer cases. Advances in recent years, such as the identification of oncogenes and the use of immunotherapies, have changed the treatment of LUAD. Yet survival rates still remain low. Additionally, there is still a gap in understanding the molecular and cellular interactions between cancer cells and the immune tumor microenvironment (TME). Defining how cancer cells with distinct oncogenic drivers interact with the TME and new strategies for enhancing anti-tumor immunity are greatly needed. The complement cascade, a central part of the innate immune system, plays an important role in regulation of adaptive immunity. Initially it was proposed that complement activation on the surface of cancer cells would inhibit cancer progression via membrane attack complex (MAC)-dependent killing. However, data from several groups have shown that complement activation promotes cancer progression, probably through the actions of anaphylatoxins (C3a and C5a) on the TME and engagement of immunoevasive pathways. While originally shown to be produced in the liver, recent studies show localized complement production in numerous cell types including immune cells and tumor cells. These results suggest that complement inhibitory drugs may represent a powerful new approach for treatment of NSCLC, and numerous new anti-complement drugs are in clinical development. However, the mechanisms by which complement is activated and affects tumor progression are not well understood. Furthermore, the role of local complement production vs. systemic activation has not been carefully examined. This review will focus on our current understanding of complement action in LUAD, and describe gaps in our knowledge critical for advancing complement therapy into the clinic.
Collapse
Affiliation(s)
- Emily K Kleczko
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Jeff W Kwak
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Erin L Schenk
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Raphael A Nemenoff
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| |
Collapse
|
146
|
Qian D, Liu H, Wang X, Ge J, Luo S, Patz EF, Moorman PG, Su L, Shen S, Christiani DC, Wei Q. Potentially functional genetic variants in the complement-related immunity gene-set are associated with non-small cell lung cancer survival. Int J Cancer 2019; 144:1867-1876. [PMID: 30259978 PMCID: PMC6377316 DOI: 10.1002/ijc.31896] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Revised: 08/30/2018] [Accepted: 09/10/2018] [Indexed: 12/30/2022]
Abstract
The complement system plays an important role in the innate and adaptive immunity, complement components mediate tumor cytolysis of antibody-based immunotherapy, and complement activation in the tumor microenvironment may promote tumor progression or inhibition, depending on the mechanism of action. In the present study, we conducted a two-phase analysis of two independently published genome-wide association studies (GWASs) for associations between genetic variants in a complement-related immunity gene-set and overall survival of non-small cell lung cancer (NSCLC). The GWAS dataset from Prostate, Lung, Colorectal and Ovarian (PLCO) Cancer Screening Trial was used as the discovery, and multivariate Cox proportional hazards regression with false-positive report probability for multiple test corrections were performed to evaluate associations between 14,699 single-nucleotide polymorphisms (SNPs) in 111 genes and survival of 1,185 NSCLC patients. The identified significant SNPs in a single-locus analysis were further validated with 984 NSCLC patients in the GWAS dataset from the Harvard Lung Cancer Susceptibility (HLCS) Study. The results showed that two independent, potentially functional SNPs in two genes (VWF rs73049469 and ITGB2 rs3788142) were significantly associated with NSCLC survival, with a combined hazards ratio (HR) of 1.22 [95% confidence interval (CI) = 1.07-1.40, P = 0.002] and 1.16 (1.07-1.27, 6.45 × 10-4 ), respectively. Finally, we performed expression quantitative trait loci (eQTL) analysis and found that survival-associated genotypes of VWF rs73049469 were also significantly associated with mRNA expression levels of the gene. These results indicated that genetic variants of the complement-related immunity genes might be predictors of NSCLC survival, particularly for the short-term survival, possibly by modulating the expression of genes involved in the host immunity.
Collapse
Affiliation(s)
- Danwen Qian
- Cancer Institute, Fudan University Shanghai Cancer Center; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
- Duke Cancer Institute, Duke University Medical Center, Durham, NC 27710, USA
- Department of Population Health Sciences, Duke University School of Medicine, Durham, NC 27710, USA
| | - Hongliang Liu
- Duke Cancer Institute, Duke University Medical Center, Durham, NC 27710, USA
- Department of Population Health Sciences, Duke University School of Medicine, Durham, NC 27710, USA
| | - Xiaomeng Wang
- Duke Cancer Institute, Duke University Medical Center, Durham, NC 27710, USA
- Department of Population Health Sciences, Duke University School of Medicine, Durham, NC 27710, USA
| | - Jie Ge
- Duke Cancer Institute, Duke University Medical Center, Durham, NC 27710, USA
- Department of Population Health Sciences, Duke University School of Medicine, Durham, NC 27710, USA
| | - Sheng Luo
- Department of Biostatistics and Bioinformatics, Duke University School of Medicine, Durham, NC 27710, USA
| | - Edward F. Patz
- Duke Cancer Institute, Duke University Medical Center, Durham, NC 27710, USA
- Department of Radiology, Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC 27710, USA
| | - Patricia G. Moorman
- Duke Cancer Institute, Duke University Medical Center, Durham, NC 27710, USA
- Department of Community and Family Medicine, Duke University Medical Center, Durham, NC, USA
| | - Li Su
- Departments of Environmental Health and Department of Epidemiology, Harvard School of Public Health, Boston, MA, 02115 USA
| | - Sipeng Shen
- Departments of Environmental Health and Department of Epidemiology, Harvard School of Public Health, Boston, MA, 02115 USA
| | - David C. Christiani
- Departments of Environmental Health and Department of Epidemiology, Harvard School of Public Health, Boston, MA, 02115 USA
- Department of Medicine, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Qingyi Wei
- Cancer Institute, Fudan University Shanghai Cancer Center; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
- Duke Cancer Institute, Duke University Medical Center, Durham, NC 27710, USA
- Department of Population Health Sciences, Duke University School of Medicine, Durham, NC 27710, USA
| |
Collapse
|
147
|
Pio R, Ajona D, Ortiz-Espinosa S, Mantovani A, Lambris JD. Complementing the Cancer-Immunity Cycle. Front Immunol 2019; 10:774. [PMID: 31031765 PMCID: PMC6473060 DOI: 10.3389/fimmu.2019.00774] [Citation(s) in RCA: 144] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Accepted: 03/25/2019] [Indexed: 12/12/2022] Open
Abstract
Reactivation of cytotoxic CD8+ T-cell responses has set a new direction for cancer immunotherapy. Neutralizing antibodies targeting immune checkpoint programmed cell death protein 1 (PD-1) or its ligand (PD-L1) have been particularly successful for tumor types with limited therapeutic options such as melanoma and lung cancer. However, reactivation of T cells is only one step toward tumor elimination, and a substantial fraction of patients fails to respond to these therapies. In this context, combination therapies targeting more than one of the steps of the cancer-immune cycle may provide significant benefits. To find the best combinations, it is of upmost importance to understand the interplay between cancer cells and all the components of the immune response. This review focuses on the elements of the complement system that come into play in the cancer-immunity cycle. The complement system, an essential part of innate immunity, has emerged as a major regulator of cancer immunity. Complement effectors such as C1q, anaphylatoxins C3a and C5a, and their receptors C3aR and C5aR1, have been associated with tolerogenic cell death and inhibition of antitumor T-cell responses through the recruitment and/or activation of immunosuppressive cell subpopulations such as myeloid-derived suppressor cells (MDSCs), regulatory T cells (Tregs), or M2 tumor-associated macrophages (TAMs). Evidence is provided to support the idea that complement blocks many of the effector routes associated with the cancer-immunity cycle, providing the rationale for new therapeutic combinations aimed to enhance the antitumor efficacy of anti-PD-1/PD-L1 checkpoint inhibitors.
Collapse
Affiliation(s)
- Ruben Pio
- Program in Solid Tumors (CIMA) and Department of Biochemistry and Genetics (School of Medicine), University of Navarra, Pamplona, Spain
- Navarra Institute for Health Research (IDISNA), Pamplona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
| | - Daniel Ajona
- Program in Solid Tumors (CIMA) and Department of Biochemistry and Genetics (School of Medicine), University of Navarra, Pamplona, Spain
- Navarra Institute for Health Research (IDISNA), Pamplona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
| | - Sergio Ortiz-Espinosa
- Program in Solid Tumors (CIMA) and Department of Biochemistry and Genetics (School of Medicine), University of Navarra, Pamplona, Spain
| | - Alberto Mantovani
- Humanitas Clinical and Research Center, Humanitas University, Milan, Italy
- William Harvey Research Institute, Queen Mary University of London, London, United Kingdom
| | - John D. Lambris
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
148
|
Ajona D, Ortiz-Espinosa S, Pio R, Lecanda F. Complement in Metastasis: A Comp in the Camp. Front Immunol 2019; 10:669. [PMID: 31001273 PMCID: PMC6457318 DOI: 10.3389/fimmu.2019.00669] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Accepted: 03/12/2019] [Indexed: 12/19/2022] Open
Abstract
The complement system represents a pillar of the innate immune response. This system, critical for host defense against pathogens, encompasses more than 50 soluble, and membrane-bound proteins. Emerging evidence underscores its clinical relevance in tumor progression and its role in metastasis, one of the hallmarks of cancer. The multistep process of metastasis entails the acquisition of advantageous functions required for the formation of secondary tumors. Thus, targeting components of the complement system could impact not only on tumor initiation but also on several crucial steps along tumor dissemination. This novel vulnerability could be concomitantly exploited with current strategies overcoming tumor-mediated immunosuppression to provide a substantial clinical benefit in the treatment of metastatic disease. In this review, we offer a tour d'horizon on recent advances in this area and their prospective potential for cancer treatment.
Collapse
Affiliation(s)
- Daniel Ajona
- Program in Solid Tumors, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain.,Navarra Institute for Health Research (IdISNA), Pamplona, Spain.,Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain.,Department of Biochemistry and Genetics, School of Sciences, University of Navarra, Pamplona, Spain
| | - Sergio Ortiz-Espinosa
- Program in Solid Tumors, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain.,Department of Biochemistry and Genetics, School of Sciences, University of Navarra, Pamplona, Spain
| | - Ruben Pio
- Program in Solid Tumors, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain.,Navarra Institute for Health Research (IdISNA), Pamplona, Spain.,Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain.,Department of Biochemistry and Genetics, School of Sciences, University of Navarra, Pamplona, Spain
| | - Fernando Lecanda
- Program in Solid Tumors, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain.,Navarra Institute for Health Research (IdISNA), Pamplona, Spain.,Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain.,Department of Pathology, Anatomy and Physiology, School of Medicine, University of Navarra, Pamplona, Spain
| |
Collapse
|
149
|
Zhang Y, Lai BS, Juhas M. Recent Advances in Aptamer Discovery and Applications. Molecules 2019; 24:molecules24050941. [PMID: 30866536 PMCID: PMC6429292 DOI: 10.3390/molecules24050941] [Citation(s) in RCA: 381] [Impact Index Per Article: 63.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Revised: 03/03/2019] [Accepted: 03/04/2019] [Indexed: 12/22/2022] Open
Abstract
Aptamers are short, single-stranded DNA, RNA, or synthetic XNA molecules that can be developed with high affinity and specificity to interact with any desired targets. They have been widely used in facilitating discoveries in basic research, ensuring food safety and monitoring the environment. Furthermore, aptamers play promising roles as clinical diagnostics and therapeutic agents. This review provides update on the recent advances in this rapidly progressing field of research with particular emphasis on generation of aptamers and their applications in biosensing, biotechnology and medicine. The limitations and future directions of aptamers in target specific delivery and real-time detection are also discussed.
Collapse
Affiliation(s)
- Yang Zhang
- College of Science, Harbin Institute of Technology, Shenzhen 518055, China.
| | - Bo Shiun Lai
- School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA.
| | - Mario Juhas
- Institute of Medical Microbiology, University of Zurich, Gloriastrasse 28/30, CH-8006 Zurich, Switzerland.
| |
Collapse
|
150
|
Singel KL, Emmons TR, Khan ANMNH, Mayor PC, Shen S, Wong JT, Morrell K, Eng KH, Mark J, Bankert RB, Matsuzaki J, Koya RC, Blom AM, McLeish KR, Qu J, Ram S, Moysich KB, Abrams SI, Odunsi K, Zsiros E, Segal BH. Mature neutrophils suppress T cell immunity in ovarian cancer microenvironment. JCI Insight 2019; 4:122311. [PMID: 30730851 PMCID: PMC6483507 DOI: 10.1172/jci.insight.122311] [Citation(s) in RCA: 92] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Accepted: 01/25/2019] [Indexed: 12/25/2022] Open
Abstract
Epithelial ovarian cancer (EOC) often presents with metastases and ascites. Granulocytic myeloid-derived suppressor cells are an immature population that impairs antitumor immunity. Since suppressive granulocytes in the ascites of patients with newly diagnosed EOC were morphologically mature, we hypothesized that PMN were rendered suppressive in the tumor microenvironment (TME). Circulating PMN from patients were not suppressive but acquired a suppressor phenotype (defined as ≥1 log10 reduction of anti-CD3/CD28-stimulated T cell proliferation) after ascites supernatant exposure. Ascites supernatants (20 of 31 supernatants) recapitulated the suppressor phenotype in PMN from healthy donors. T cell proliferation was restored with ascites removal and restimulation. PMN suppressors also inhibited T cell activation and cytokine production. PMN suppressors completely suppressed proliferation in naive, central memory, and effector memory T cells and in engineered tumor antigen-specific cytotoxic T lymphocytes, while antigen-specific cell lysis was unaffected. Inhibition of complement C3 activation and PMN effector functions, including CR3 signaling, protein synthesis, and vesicular trafficking, abrogated the PMN suppressor phenotype. Moreover, malignant effusions from patients with various metastatic cancers also induced the C3-dependent PMN suppressor phenotype. These results point to PMN impairing T cell expansion and activation in the TME and the potential for complement inhibition to abrogate this barrier to antitumor immunity.
Collapse
Affiliation(s)
| | | | | | - Paul C. Mayor
- Department of Surgery, Division of Gynecologic Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
| | - Shichen Shen
- New York State Center of Excellence Bioinformatics and Life Sciences, University at Buffalo, Buffalo, New York, USA
| | | | - Kayla Morrell
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
| | - Kevin H. Eng
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
| | - Jaron Mark
- Department of Surgery, Division of Gynecologic Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
| | - Richard B. Bankert
- Department of Microbiology and Immunology, University at Buffalo Jacobs School of Medicine and Biomedical Sciences, Buffalo, New York, USA
| | - Junko Matsuzaki
- Center for Immunotherapy, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
| | - Richard C. Koya
- Center for Immunotherapy, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
| | - Anna M. Blom
- Division of Medical Protein Chemistry, Department of Translational Medicine, Lund University, Malmö, Sweden
| | - Kenneth R. McLeish
- Department of Medicine, University of Louisville School of Medicine, Louisville, Kentucky, USA
| | - Jun Qu
- New York State Center of Excellence Bioinformatics and Life Sciences, University at Buffalo, Buffalo, New York, USA
| | - Sanjay Ram
- Department of Medicine, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Kirsten B. Moysich
- Department of Cancer Prevention and Control, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
| | | | - Kunle Odunsi
- Department of Surgery, Division of Gynecologic Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
- Center for Immunotherapy, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
| | - Emese Zsiros
- Department of Surgery, Division of Gynecologic Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
- Center for Immunotherapy, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
| | - Brahm H. Segal
- Department of Immunology
- Department of Internal Medicine, and
- Department of Medicine, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, New York, USA
| |
Collapse
|