1
|
Flati I, Di Vito Nolfi M, Dall’Aglio F, Vecchiotti D, Verzella D, Alesse E, Capece D, Zazzeroni F. Molecular Mechanisms Underpinning Immunometabolic Reprogramming: How the Wind Changes during Cancer Progression. Genes (Basel) 2023; 14:1953. [PMID: 37895302 PMCID: PMC10606647 DOI: 10.3390/genes14101953] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 10/11/2023] [Accepted: 10/12/2023] [Indexed: 10/29/2023] Open
Abstract
Metabolism and the immunological state are intimately intertwined, as defense responses are bioenergetically expensive. Metabolic homeostasis is a key requirement for the proper function of immune cell subsets, and the perturbation of the immune-metabolic balance is a recurrent event in many human diseases, including cancer, due to nutrient fluctuation, hypoxia and additional metabolic changes occurring in the tumor microenvironment (TME). Although much remains to be understood in the field of immunometabolism, here, we report the current knowledge on both physiological and cancer-associated metabolic profiles of immune cells, and the main molecular circuits involved in their regulation, highlighting similarities and differences, and emphasizing immune metabolic liabilities that could be exploited in cancer therapy to overcome immune resistance.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Daria Capece
- Department of Biotechnological and Applied Clinical Sciences (DISCAB), University of L’Aquila, Via Vetoio, Coppito 2, 67100 L’Aquila, Italy; (I.F.); (M.D.V.N.); (F.D.); (D.V.); (D.V.); (E.A.); (F.Z.)
| | | |
Collapse
|
2
|
Walters AA, Ali A, Wang JTW, Al-Jamal KT. Anti-tumor antibody isotype response can be modified with locally administered immunoadjuvants. Drug Deliv Transl Res 2023; 13:2032-2040. [PMID: 36417163 PMCID: PMC10238356 DOI: 10.1007/s13346-022-01258-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/07/2022] [Indexed: 11/24/2022]
Abstract
In situ vaccination with immunostimulatory compounds is a demonstrated means to treat tumors preclinically. While these therapeutic effects have been attributed to the actions of T cells or innate immune activation, characterisation of the humoral immune response is seldom performed. This study aims to identify whether the injection of immunoadjuvants, Addavax (Adda) and cytosine-phosphorothioate-guanine oligodeoxynucleotide (CpG), intratumorally can influence the antibody response. Specifically, whether intratumoral injection of immunoadjuvants can alter the tumor-specific antibody target, titre and isotype. Following this, the study aimed to investigate whether serum obtained from in situ vaccinated mice could neutralise circulating tumor cells. Serum was obtained from mice bearing B16F10-OVA-Luc-GFP tumors treated with immunoadjuvants. Antibody targets' titre and isotype were assessed by indirect ELISA. The ability of serum to neutralise circulating cancer cells was evaluated in a B16F10 pseudo-metastatic model. It was observed that tumor-bearing mice mount a specific anti-tumor antibody response. Antibody titre and target were unaffected by in situ vaccination with immunoadjuvants; however, a higher amount of IgG2c was produced in mice receiving Adda plus CpG. Serum from in situ vaccinated mice was unable to neutralise circulating B16F10 cells. Thus, this study has demonstrated that anti-tumor antibody isotype may be modified using in situ vaccination; however, this alone is not sufficient to neutralise circulating cancer cells.
Collapse
Affiliation(s)
- Adam A Walters
- Institute of Pharmaceutical Science, Faculty of Life Sciences & Medicine, King's College London, Franklin-Wilkins Building, 150 Stamford Street, London, SE1 9NH, UK
| | - Abrar Ali
- Institute of Pharmaceutical Science, Faculty of Life Sciences & Medicine, King's College London, Franklin-Wilkins Building, 150 Stamford Street, London, SE1 9NH, UK
| | - Julie Tzu-Wen Wang
- Institute of Pharmaceutical Science, Faculty of Life Sciences & Medicine, King's College London, Franklin-Wilkins Building, 150 Stamford Street, London, SE1 9NH, UK
| | - Khuloud T Al-Jamal
- Institute of Pharmaceutical Science, Faculty of Life Sciences & Medicine, King's College London, Franklin-Wilkins Building, 150 Stamford Street, London, SE1 9NH, UK.
| |
Collapse
|
3
|
Abstract
The opposing roles of innate and adaptive immune cells in suppressing or supporting cancer initiation, progression, metastasis and response to therapy has been long debated. The mechanisms by which different monocyte and T cell subtypes affect and modulate cancer have been extensively studied. However, the role of B cells and their subtypes have remained elusive, perhaps partially due to their heterogeneity and range of actions. B cells can produce a variety of cytokines and present tumor-derived antigens to T cells in combination with co-stimulatory or inhibitory ligands based on their phenotype. Unlike most T cells, B cells can be activated by innate immune stimuli, such as endotoxin. Furthermore, the isotype and specificity of the antibodies produced by plasma cells regulate distinct immune responses, including opsonization, antibody-mediated cellular cytotoxicity (ADCC) and complement activation. B cells are shaped by the tumor environment (TME), with the capability to regulate the TME in return. In this review, we will describe the mechanisms of B cell action, including cytokine production, antigen presentation, ADCC, opsonization, complement activation and how they affect tumor development and response to immunotherapy. We will also discuss how B cell fate within the TME is affected by tumor stroma, microbiome and metabolism.
Collapse
Affiliation(s)
- Shabnam Shalapour
- Department of Cancer Biology, University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA.
| | - Michael Karin
- Department of Pharmacology, School of Medicine, University of California San Diego, CA 92093, USA; Laboratory of Gene Regulation and Signal Transduction, Department of Pharmacology, School of Medicine, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| |
Collapse
|
4
|
Maia J, Otake AH, Poças J, Carvalho AS, Beck HC, Magalhães A, Matthiesen R, Strano Moraes MC, Costa-Silva B. Transcriptome Reprogramming of CD11b + Bone Marrow Cells by Pancreatic Cancer Extracellular Vesicles. Front Cell Dev Biol 2020; 8:592518. [PMID: 33330473 PMCID: PMC7729189 DOI: 10.3389/fcell.2020.592518] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Accepted: 10/27/2020] [Indexed: 12/12/2022] Open
Abstract
Pancreatic cancers (PC) are highly metastatic with poor prognosis, mainly due to delayed detection. We previously showed that PC-derived extracellular vesicles (EVs) act on macrophages residing in the liver, eliciting extracellular matrix remodeling in this organ and marked hepatic accumulation of CD11b+ bone marrow (BM) cells, which support PC liver metastasis. We here show that PC-EVs also bind to CD11b+ BM cells and induce the expansion of this cell population. Transcriptomic characterization of these cells shows that PC-EVs upregulate IgG and IgA genes, which have been linked to the presence of monocytes/macrophages in tumor microenvironments. We also report here the transcriptional downregulation of genes linked to monocyte/macrophage activation, trafficking, and expression of inflammatory molecules. Together, these results show for the first time the existence of a PC-BM communication axis mediated by EVs with a potential role in PC tumor microenvironments.
Collapse
Affiliation(s)
- Joana Maia
- Champalimaud Centre for the Unknown, Champalimaud Foundation, Lisbon, Portugal
- Graduate Program in Areas of Basic and Applied Biology, University of Porto, Porto, Portugal
| | - Andreia Hanada Otake
- Champalimaud Centre for the Unknown, Champalimaud Foundation, Lisbon, Portugal
- Center for Translational Research in Oncology, Instituto do Câncer do Estado de São Paulo, Hospital das Clínicas, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Juliana Poças
- i3S – Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- IPATIMUP – Instituto de Patologia e Imunologia Molecular da Universidade do Porto, Porto, Portugal
- Instituto de Ciências Biomédicas Abel Salazar, University of Porto, Porto, Portugal
| | - Ana Sofia Carvalho
- Computational and Experimental Biology Group, CEDOC, Chronic Diseases Research Centre, NOVA Medical School, Faculdade de Ciencias Medicas, Universidade NOVA de Lisboa, Lisbon, Portugal
| | - Hans Christian Beck
- Centre for Clinical Proteomics, Department of Clinical Biochemistry and Pharmacology, Odense University Hospital, Odense, Denmark
| | - Ana Magalhães
- i3S – Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- IPATIMUP – Instituto de Patologia e Imunologia Molecular da Universidade do Porto, Porto, Portugal
| | - Rune Matthiesen
- Computational and Experimental Biology Group, CEDOC, Chronic Diseases Research Centre, NOVA Medical School, Faculdade de Ciencias Medicas, Universidade NOVA de Lisboa, Lisbon, Portugal
| | | | - Bruno Costa-Silva
- Champalimaud Centre for the Unknown, Champalimaud Foundation, Lisbon, Portugal
| |
Collapse
|
5
|
Abstract
Tumor immunology is undergoing a renaissance due to the recent profound clinical successes of tumor immunotherapy. These advances have coincided with an exponential growth in the development of -omics technologies. Armed with these technologies and their associated computational and modeling toolsets, systems biologists have turned their attention to tumor immunology in an effort to understand the precise nature and consequences of interactions between tumors and the immune system. Such interactions are inherently multivariate, spanning multiple time and size scales, cell types, and organ systems, rendering systems biology approaches particularly amenable to their interrogation. While in its infancy, the field of 'Cancer Systems Immunology' has already influenced our understanding of tumor immunology and immunotherapy. As the field matures, studies will move beyond descriptive characterizations toward functional investigations of the emergent behavior that govern tumor-immune responses. Thus, Cancer Systems Immunology holds incredible promise to advance our ability to fight this disease.
Collapse
Affiliation(s)
| | - Edgar G Engleman
- Department of Pathology, Stanford University School of MedicineStanfordUnited States
- Division of Immunology and Rheumatology, Department of Medicine, Stanford University School of MedicineStanfordUnited States
- Stanford Cancer Institute, Stanford UniversityStanfordUnited States
| |
Collapse
|
6
|
Li X, Larsson P, Ljuslinder I, Öhlund D, Myte R, Löfgren-Burström A, Zingmark C, Ling A, Edin S, Palmqvist R. Ex Vivo Organoid Cultures Reveal the Importance of the Tumor Microenvironment for Maintenance of Colorectal Cancer Stem Cells. Cancers (Basel) 2020; 12:E923. [PMID: 32290033 PMCID: PMC7226030 DOI: 10.3390/cancers12040923] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Accepted: 04/07/2020] [Indexed: 12/14/2022] Open
Abstract
Colorectal cancer (CRC) is a heterogeneous disease, with varying clinical presentations and patient prognosis. Different molecular subgroups of CRC should be treated differently and therefore, must be better characterized. Organoid culture has recently been suggested as a good model to reflect the heterogeneous nature of CRC. However, organoid cultures cannot be established from all CRC tumors. The study examines which CRC tumors are more likely to generate organoids and thus benefit from ex vivo organoid drug testing. Long-term organoid cultures from 22 out of 40 CRC tumor specimens were established. It was found that organoid cultures were more difficult to establish from tumors characterized as microsatellite instable (MSI), BRAF-mutated, poorly differentiated and/or of a mucinous type. This suggests that patients with such tumors are less likely to benefit from ex vivo organoid drug testing, but it may also suggest biological difference in tumor growth. RNA sequencing analysis of tumor sections revealed that the in vivo maintenance of these non-organoid-forming tumors depends on factors related to inflammation and pathogen exposure. Furthermore, using TCGA data we could show a trend towards a worse prognosis for patients with organoid-forming tumors, suggesting also clinical differences. Results suggest that organoids are more difficult to establish from tumors characterized as MSI, BRAF-mutated, poorly differentiated and/or of a mucinous type. We further suggest that the maintenance of cell growth of these tumors in vivo may be promoted by immune-related factors and other stromal components within the tumor microenvironment.
Collapse
Affiliation(s)
- Xingru Li
- Department of Medical Biosciences, Pathology, Umeå University, 90185 Umeå, Sweden; (X.L.); (P.L.); (A.L.-B.); (C.Z.); (A.L.); (S.E.)
| | - Pär Larsson
- Department of Medical Biosciences, Pathology, Umeå University, 90185 Umeå, Sweden; (X.L.); (P.L.); (A.L.-B.); (C.Z.); (A.L.); (S.E.)
| | - Ingrid Ljuslinder
- Department of Radiation Sciences, Oncology, Umeå University, 90185 Umeå, Sweden; (I.L.); (D.O.); (R.M.)
| | - Daniel Öhlund
- Department of Radiation Sciences, Oncology, Umeå University, 90185 Umeå, Sweden; (I.L.); (D.O.); (R.M.)
- Wallenberg Center for Molecular Medicine, Umeå University, 90185 Umeå, Sweden
| | - Robin Myte
- Department of Radiation Sciences, Oncology, Umeå University, 90185 Umeå, Sweden; (I.L.); (D.O.); (R.M.)
| | - Anna Löfgren-Burström
- Department of Medical Biosciences, Pathology, Umeå University, 90185 Umeå, Sweden; (X.L.); (P.L.); (A.L.-B.); (C.Z.); (A.L.); (S.E.)
| | - Carl Zingmark
- Department of Medical Biosciences, Pathology, Umeå University, 90185 Umeå, Sweden; (X.L.); (P.L.); (A.L.-B.); (C.Z.); (A.L.); (S.E.)
| | - Agnes Ling
- Department of Medical Biosciences, Pathology, Umeå University, 90185 Umeå, Sweden; (X.L.); (P.L.); (A.L.-B.); (C.Z.); (A.L.); (S.E.)
| | - Sofia Edin
- Department of Medical Biosciences, Pathology, Umeå University, 90185 Umeå, Sweden; (X.L.); (P.L.); (A.L.-B.); (C.Z.); (A.L.); (S.E.)
| | - Richard Palmqvist
- Department of Medical Biosciences, Pathology, Umeå University, 90185 Umeå, Sweden; (X.L.); (P.L.); (A.L.-B.); (C.Z.); (A.L.); (S.E.)
| |
Collapse
|
7
|
Sharonov GV, Serebrovskaya EO, Yuzhakova DV, Britanova OV, Chudakov DM. B cells, plasma cells and antibody repertoires in the tumour microenvironment. Nat Rev Immunol 2020; 20:294-307. [DOI: 10.1038/s41577-019-0257-x] [Citation(s) in RCA: 201] [Impact Index Per Article: 50.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/27/2019] [Indexed: 02/07/2023]
|
8
|
Immune biological rationales for the design of combined radio- and immunotherapies. Cancer Immunol Immunother 2020; 69:293-306. [PMID: 31953578 PMCID: PMC7000501 DOI: 10.1007/s00262-019-02460-3] [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: 07/30/2019] [Accepted: 12/22/2019] [Indexed: 02/07/2023]
Abstract
Cancer immunotherapies are promising treatments for many forms of cancer. Nevertheless, the response rates to, e.g., immune checkpoint inhibitors (ICI), are still in low double-digit percentage. This calls for further therapy optimization that should take into account combination of immunotherapies with classical tumor therapies such as radiotherapy. By designing multimodal approaches, immune modulatory properties of certain radiation schemes, additional immune modulation by immunotherapy with ICI and hyperthermia, as well as patient stratification based on genetic and immune constitutions have to be considered. In this context, both the tumor and its microenvironment including cells of the innate and adaptive immune system have to be viewed in synopsis. Knowledge of immune activation and immune suppression by radiation is the basis for well-elaborated addition of certain immunotherapies. In this review, the focus is set on additional immune stimulation by hyperthermia and restoration of an immune response by ICI. The impact of radiation dose and fractionation on immune modulation in multimodal settings has to be considered, as the dynamics of the immune response and the timing between radiotherapy and immunotherapy. Another big challenge is the patient stratification that should be based on matrices of biomarkers, taking into account genetics, proteomics, radiomics, and “immunomics”. One key aim is to turn immunological “cold” tumors into “hot” tumors, and to eliminate barriers of immune-suppressed or immune-excluded tumors. Comprehensive knowledge of immune alterations induced by radiation and immunotherapy when being applied together should be utilized for patient-adapted treatment planning and testing of innovative tumor therapies within clinical trials.
Collapse
|
9
|
Isaeva OI, Sharonov GV, Serebrovskaya EO, Turchaninova MA, Zaretsky AR, Shugay M, Chudakov DM. Intratumoral immunoglobulin isotypes predict survival in lung adenocarcinoma subtypes. J Immunother Cancer 2019; 7:279. [PMID: 31665076 PMCID: PMC6819482 DOI: 10.1186/s40425-019-0747-1] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Accepted: 09/20/2019] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND The role of tumor-infiltrating B-cells (TIBs) and intratumorally-produced antibodies in cancer-immunity interactions essentially remains terra incognita. In particular, it remains unexplored how driver mutations could be associated with distinct TIBs signatures and their role in tumor microenvironment. METHODS Here we analyzed associations of immunoglobulin isotypes and clonality with survival in TCGA RNA-Seq data for lung adenocarcinoma (LUAD), stratifying patients into 12 driver mutation and phenotypic tumor subgroups. RESULTS We revealed several unexpected associations between TIBs behavior and prognosis. Abundance and high proportion of IgG1 isotype, and low proportion of IgA among all intratumorally produced immunoglobulins were specifically associated with improved overall survival for KRASmut but not KRASwt LUAD, revealing the first link between a driver mutation and B-cell response. We found specific IgG1 signature associated with long survival, which suggests that particular specificities of IgG1+ TIBs could be beneficial in KRASmut LUAD. In contrast to our previous observations for melanoma, highly clonal IgG1 production by plasma cells had no meaningful effect on prognosis, suggesting that IgG1+ TIBs may exert a beneficial effect in KRASmut cases in an alternative way, such as efficient presentation of cognate antigens or direct B cell attack on tumor cells. Notably, a high proportion of the IgG1 isotype is positively correlated with the non-silent mutation burden both in the general LUAD cohort and in most patient subgroups, supporting a role for IgG1+ TIBs in antigen presentation. Complementing the recent finding that the presence of stromal IgG4-producing cells is associated with a favorable prognosis for patients with stage I squamous cell carcinoma, we show that the abundance of IgG4-producing TIBs likewise has a strong positive effect on overall survival in STK11mut and proximal proliferative subgroups of LUAD patients. We hypothesize that the positive role of IgG4 antibodies in some of the lung cancer subtypes could be associated with reported inability of IgG4 isotype to form immune complexes, thus preventing immunosuppression via activation of the myeloid-derived suppressor cell (MDSC) phenotype. CONCLUSIONS We discover prominent and distinct associations between TIBs antibody isotypes and survival in lung adenocarcinoma carrying specific driver mutations. These findings indicate that particular types of tumor-immunity relations could be beneficial in particular driver mutation context, which should be taken into account in developing strategies of cancer immunotherapy and combination therapies. Specificity of protective B cell populations in specific cancer subgroups could become a clue to efficient targeted immunotherapies for appropriate cohorts of patients.
Collapse
Affiliation(s)
- O I Isaeva
- Center of Life Sciences, Skolkovo Institute of Science and Technology, Moscow, Russia.,BostonGene LLC, Lincoln, MA, USA
| | - G V Sharonov
- Laboratory of Genomics of Antitumor Adaptive Immunity, Privolzhsky Research Medical University, Nizhny Novgorod, Russia.,Genomics of Adaptive Immunity Department, Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry, Moscow, Russia
| | - E O Serebrovskaya
- Genomics of Adaptive Immunity Department, Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry, Moscow, Russia.,Institute of Translational Medicine, Pirogov Russian National Research Medical University, Moscow, Russia
| | - M A Turchaninova
- Laboratory of Genomics of Antitumor Adaptive Immunity, Privolzhsky Research Medical University, Nizhny Novgorod, Russia.,Genomics of Adaptive Immunity Department, Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry, Moscow, Russia
| | - A R Zaretsky
- Genomics of Adaptive Immunity Department, Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry, Moscow, Russia.,Institute of Translational Medicine, Pirogov Russian National Research Medical University, Moscow, Russia.,Evrogen JSC, Moscow, Russia
| | - M Shugay
- Center of Life Sciences, Skolkovo Institute of Science and Technology, Moscow, Russia.,Laboratory of Genomics of Antitumor Adaptive Immunity, Privolzhsky Research Medical University, Nizhny Novgorod, Russia.,Genomics of Adaptive Immunity Department, Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry, Moscow, Russia.,Institute of Translational Medicine, Pirogov Russian National Research Medical University, Moscow, Russia
| | - D M Chudakov
- Center of Life Sciences, Skolkovo Institute of Science and Technology, Moscow, Russia. .,Laboratory of Genomics of Antitumor Adaptive Immunity, Privolzhsky Research Medical University, Nizhny Novgorod, Russia. .,Genomics of Adaptive Immunity Department, Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry, Moscow, Russia. .,Institute of Translational Medicine, Pirogov Russian National Research Medical University, Moscow, Russia.
| |
Collapse
|
10
|
de Leve S, Wirsdörfer F, Jendrossek V. Targeting the Immunomodulatory CD73/Adenosine System to Improve the Therapeutic Gain of Radiotherapy. Front Immunol 2019; 10:698. [PMID: 31024543 PMCID: PMC6460721 DOI: 10.3389/fimmu.2019.00698] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Accepted: 03/14/2019] [Indexed: 12/23/2022] Open
Abstract
Extracellular adenosine is a potent endogenous immunosuppressive mediator critical to the maintenance of homeostasis in various normal tissues including the lung. Adenosine is either released from stressed or injured cells or generated from extracellular adenine nucleotides by the concerted action of the ectoenzymes ectoapyrase (CD39) and 5′ ectonucleotidase (CD73) that catabolize ATP to adenosine. An acute CD73-dependent increase of adenosine in normal tissues mostly exerts tissue protective functions whereas chronically increased adenosine-levels in tissues exposed to DNA damaging chemotherapy or radiotherapy promote pathologic remodeling processes and fibrosis for example in the skin and the lung. Importantly, cancer cells also express CD73 and high CD73 expression in the tumor tissue has been linked to poor overall survival and recurrence free survival in patients suffering from breast and ovarian cancer. CD73 and adenosine support growth-promoting neovascularization, metastasis, and survival in cancer cells. In addition, adenosine can promote tumor intrinsic or therapy-induced immune escape by various mechanisms that dampen the immune system. Consequently, modulating CD73 or cancer-derived adenosine in the tumor microenvironment emerges as an attractive novel therapeutic strategy to limit tumor progression, improve antitumor immune responses, avoid therapy-induced immune deviation, and potentially limit normal tissue toxicity. However, the role of CD73/adenosine signaling in the tumor and normal tissue responses to radiotherapy and its use as therapeutic target to improve the outcome of radiotherapy approaches is less understood. The present review will highlight the dual role of CD73 and adenosine in tumor and tissue responses to radiotherapy with a special focus to the lung. It will also discuss the potential benefits and risks of pharmacologic modulation of the CD73/adenosine system to increase the therapeutic gain of radiotherapy or combined radioimmunotherapy in cancer treatment.
Collapse
Affiliation(s)
- Simone de Leve
- Institute of Cell Biology (Cancer Research), University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Florian Wirsdörfer
- Institute of Cell Biology (Cancer Research), University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Verena Jendrossek
- Institute of Cell Biology (Cancer Research), University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| |
Collapse
|
11
|
Wirsdörfer F, de Leve S, Jendrossek V. Combining Radiotherapy and Immunotherapy in Lung Cancer: Can We Expect Limitations Due to Altered Normal Tissue Toxicity? Int J Mol Sci 2018; 20:ijms20010024. [PMID: 30577587 PMCID: PMC6337556 DOI: 10.3390/ijms20010024] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Revised: 12/18/2018] [Accepted: 12/19/2018] [Indexed: 02/08/2023] Open
Abstract
In recent decades, technical advances in surgery and radiotherapy, as well as breakthroughs in the knowledge on cancer biology, have helped to substantially improve the standard of cancer care with respect to overall response rates, progression-free survival, and the quality of life of cancer patients. In this context, immunotherapy is thought to have revolutionized the standard of care for cancer patients in the long term. For example, immunotherapy approaches such as immune checkpoint blockade are currently increasingly being used in cancer treatment, either alone or in combination with chemotherapy or radiotherapy, and there is hope from the first clinical trials that the appropriate integration of immunotherapy into standard care will raise the success rates of cancer therapy to a new level. Nevertheless, successful cancer therapy remains a major challenge, particularly in tumors with either pronounced resistance to chemotherapy and radiation treatment, a high risk of normal tissue complications, or both, as in lung cancer. Chemotherapy, radiotherapy and immunotherapy have the capacity to evoke adverse effects in normal tissues when administered alone. However, therapy concepts are usually highly complex, and it is still not clear if combining immunotherapy with radio(chemo)therapy will increase the risk of normal tissue complications, in particular since normal tissue toxicity induced by chemotherapy and radiotherapy can involve immunologic processes. Unfortunately, no reliable biomarkers are available so far that are suited to predict the unique normal tissue sensitivity of a given patient to a given treatment. Consequently, clinical trials combining radiotherapy and immunotherapy are attracting major attention, not only regarding efficacy, but also with regard to safety. In the present review, we summarize the current knowledge of radiation-induced and immunotherapy-induced effects in tumor and normal tissue of the lung, and discuss the potential limitations of combined radio-immunotherapy in lung cancer with a focus on the suspected risk for enhanced acute and chronic normal tissue toxicity.
Collapse
Affiliation(s)
- Florian Wirsdörfer
- Institute of Cell Biology (Cancer Research), University Hospital Essen, 45147 Essen, Germany.
| | - Simone de Leve
- Institute of Cell Biology (Cancer Research), University Hospital Essen, 45147 Essen, Germany.
| | - Verena Jendrossek
- Institute of Cell Biology (Cancer Research), University Hospital Essen, 45147 Essen, Germany.
| |
Collapse
|
12
|
Christodoulou I, Goulielmaki M, Devetzi M, Panagiotidis M, Koliakos G, Zoumpourlis V. Mesenchymal stem cells in preclinical cancer cytotherapy: a systematic review. Stem Cell Res Ther 2018; 9:336. [PMID: 30526687 PMCID: PMC6286545 DOI: 10.1186/s13287-018-1078-8] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Mesenchymal stem cells (MSC) comprise a heterogeneous population of rapidly proliferating cells that can be isolated from adult (e.g., bone marrow, adipose tissue) as well as fetal (e.g., umbilical cord) tissues (termed bone marrow (BM)-, adipose tissue (AT)-, and umbilical cord (UC)-MSC, respectively) and are capable of differentiation into a wide range of non-hematopoietic cell types. An additional, unique attribute of MSC is their ability to home to tumor sites and to interact with the local supportive microenvironment which rapidly conceptualized into MSC-based experimental cancer cytotherapy at the turn of the century. Towards this purpose, both naïve (unmodified) and genetically modified MSC (GM-MSC; used as delivery vehicles for the controlled expression and release of antitumorigenic molecules) have been employed using well-established in vitro and in vivo cancer models, albeit with variable success. The first approach is hampered by contradictory findings regarding the effects of naïve MSC of different origins on tumor growth and metastasis, largely attributed to inherent biological heterogeneity of MSC as well as experimental discrepancies. In the second case, although the anti-cancer effect of GM-MSC is markedly improved over that of naïve cells, it is yet apparent that some protocols are more efficient against some types of cancer than others. Regardless, in order to maximize therapeutic consistency and efficacy, a deeper understanding of the complex interaction between MSC and the tumor microenvironment is required, as well as examination of the role of key experimental parameters in shaping the final cytotherapy outcome. This systematic review represents, to the best of our knowledge, the first thorough evaluation of the impact of experimental anti-cancer therapies based on MSC of human origin (with special focus on human BM-/AT-/UC-MSC). Importantly, we dissect the commonalities and differences as well as address the shortcomings of work accumulated over the last two decades and discuss how this information can serve as a guide map for optimal experimental design implementation ultimately aiding the effective transition into clinical trials.
Collapse
Affiliation(s)
- Ioannis Christodoulou
- Institute of Biological Research and Biotechnology, National Hellenic Research Foundation (NHRF), Konstantinou 48 Av., 116 35, Athens, Greece
| | - Maria Goulielmaki
- Institute of Biological Research and Biotechnology, National Hellenic Research Foundation (NHRF), Konstantinou 48 Av., 116 35, Athens, Greece
| | - Marina Devetzi
- Institute of Biological Research and Biotechnology, National Hellenic Research Foundation (NHRF), Konstantinou 48 Av., 116 35, Athens, Greece
| | | | | | - Vassilis Zoumpourlis
- Institute of Biological Research and Biotechnology, National Hellenic Research Foundation (NHRF), Konstantinou 48 Av., 116 35, Athens, Greece.
| |
Collapse
|
13
|
|
14
|
Melis MHM, Nevedomskaya E, van Burgsteden J, Cioni B, van Zeeburg HJT, Song JY, Zevenhoven J, Hawinkels LJAC, de Visser KE, Bergman AM. The adaptive immune system promotes initiation of prostate carcinogenesis in a human c-Myc transgenic mouse model. Oncotarget 2017; 8:93867-93877. [PMID: 29212195 PMCID: PMC5706841 DOI: 10.18632/oncotarget.21305] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Accepted: 08/26/2017] [Indexed: 12/14/2022] Open
Abstract
Increasing evidence from epidemiological and pathological studies suggests a role of the immune system in the initiation and progression of multiple cancers, including prostate cancer. Reports on the contribution of the adaptive immune system are contradictive, since both suppression and acceleration of disease development have been reported. This study addresses the functional role of lymphocytes in prostate cancer development using a genetically engineered mouse model (GEMM) of human c-Myc driven prostate cancer (Hi-Myc mice) combined with B and T cell deficiency (RAG1-/- mice). From a pre-cancerous stage on, Hi-Myc mice showed higher accumulation of immune cells in their prostates then wild-type mice, of which macrophages were the most abundant. The onset of invasive adenocarcinoma was delayed in Hi-MycRAG1-/- compared to Hi-Myc mice and associated with decreased infiltration of leukocytes into the prostate. In addition, lower levels of the cytokines CXCL2, CCL5 and TGF-β1 were detected in Hi-MycRAG1-/- compared to Hi-Myc mouse prostates. These results from a GEMM of prostate cancer provide new insights into the promoting role of the adaptive immune system in prostate cancer development. Our findings indicate that the endogenous adaptive immune system does not protect against de novo prostate carcinogenesis in Hi-Myc transgenic mice, but rather accelerates the formation of invasive adenocarcinomas. This may have implications for the development of novel treatment strategies.
Collapse
Affiliation(s)
- Monique H M Melis
- Division of Molecular Genetics, Netherlands Cancer Institute, The Netherlands
| | | | | | - Bianca Cioni
- Division of Molecular Genetics, Netherlands Cancer Institute, The Netherlands
| | | | - Ji-Ying Song
- Division of Experimental Animal Pathology, Netherlands Cancer Institute, The Netherlands
| | - John Zevenhoven
- Division of Molecular Genetics, Netherlands Cancer Institute, The Netherlands
| | - Lukas J A C Hawinkels
- Division of Gastroenterology-Hepatology and Molecular Cell biology, Leiden university medical center, (LUMC), Netherlands
| | - Karin E de Visser
- Division of Immunology, Netherlands Cancer Institute, The Netherlands
| | - Andries M Bergman
- Division of Molecular Genetics, Netherlands Cancer Institute, The Netherlands.,Division of Medical Oncology, Netherlands Cancer Institute, The Netherlands
| |
Collapse
|
15
|
Campa MJ, Moody MA, Zhang R, Liao HX, Gottlin EB, Patz EF. Interrogation of individual intratumoral B lymphocytes from lung cancer patients for molecular target discovery. Cancer Immunol Immunother 2016; 65:171-80. [PMID: 26739486 PMCID: PMC11028467 DOI: 10.1007/s00262-015-1787-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Accepted: 12/17/2015] [Indexed: 12/18/2022]
Abstract
Intratumoral B lymphocytes are an integral part of the lung tumor microenvironment. Interrogation of the antibodies they express may improve our understanding of the host response to cancer and could be useful in elucidating novel molecular targets. We used two strategies to explore the repertoire of intratumoral B cell antibodies. First, we cloned VH and VL genes from single intratumoral B lymphocytes isolated from one lung tumor, expressed the genes as recombinant mAbs, and used the mAbs to identify the cognate tumor antigens. The Igs derived from intratumoral B cells demonstrated class switching, with a mean VH mutation frequency of 4%. Although there was no evidence for clonal expansion, these data are consistent with antigen-driven somatic hypermutation. Individual recombinant antibodies were polyreactive, although one clone demonstrated preferential immunoreactivity with tropomyosin 4 (TPM4). We found that higher levels of TPM4 antibodies were more common in cancer patients, but measurement of TPM4 antibody levels was not a sensitive test for detecting cancer. Second, in an effort to focus our recombinant antibody expression efforts on those B cells that displayed evidence of clonal expansion driven by antigen stimulation, we performed deep sequencing of the Ig genes of B cells collected from seven different tumors. Deep sequencing demonstrated somatic hypermutation but no dominant clones. These strategies may be useful for the study of B cell antibody expression, although identification of a dominant clone and unique therapeutic targets may require extensive investigation.
Collapse
Affiliation(s)
- Michael J Campa
- Department of Radiology, Duke University Medical Center, Box 3808, Durham, NC, 27710, USA
| | - M Anthony Moody
- Department of Pediatrics, Duke University Medical Center, Durham, NC, 27710, USA
- Department of Duke Human Vaccine Institute, Duke University Medical Center, Durham, NC, 27710, USA
| | - Ruijun Zhang
- Department of Duke Human Vaccine Institute, Duke University Medical Center, Durham, NC, 27710, USA
| | - Hua-Xin Liao
- Department of Duke Human Vaccine Institute, Duke University Medical Center, Durham, NC, 27710, USA
- Department of Medicine, Duke University Medical Center, Durham, NC, 27710, USA
| | - Elizabeth B Gottlin
- Department of Radiology, Duke University Medical Center, Box 3808, Durham, NC, 27710, USA
| | - Edward F Patz
- Department of Radiology, Duke University Medical Center, Box 3808, Durham, NC, 27710, USA.
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC, 27710, USA.
| |
Collapse
|
16
|
Becht E, Giraldo NA, Germain C, de Reyniès A, Laurent-Puig P, Zucman-Rossi J, Dieu-Nosjean MC, Sautès-Fridman C, Fridman WH. Immune Contexture, Immunoscore, and Malignant Cell Molecular Subgroups for Prognostic and Theranostic Classifications of Cancers. Adv Immunol 2016; 130:95-190. [DOI: 10.1016/bs.ai.2015.12.002] [Citation(s) in RCA: 126] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
|
17
|
Germain C, Gnjatic S, Dieu-Nosjean MC. Tertiary Lymphoid Structure-Associated B Cells are Key Players in Anti-Tumor Immunity. Front Immunol 2015; 6:67. [PMID: 25755654 PMCID: PMC4337382 DOI: 10.3389/fimmu.2015.00067] [Citation(s) in RCA: 111] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2014] [Accepted: 02/02/2015] [Indexed: 12/25/2022] Open
Abstract
It is now admitted that the immune system plays a major role in tumor control. Besides the existence of tumor-specific T cells and B cells, many studies have demonstrated that high numbers of tumor-infiltrating lymphocytes are associated with good clinical outcome. In addition, not only the density but also the organization of tumor-infiltrating immune cells has been shown to determine patient survival. Indeed, more and more studies describe the development within the tumor microenvironment of tertiary lymphoid structures (TLS), whose presence has a positive impact on tumor prognosis. TLS are transient ectopic lymphoid aggregates displaying the same organization and functionality as canonical secondary lymphoid organs, with T-cell-rich and B-cell-rich areas that are sites for the differentiation of effector and memory T cells and B cells. However, factors favoring the emergence of such structures within tumors still need to be fully characterized. In this review, we survey the state of the art of what is known about the general organization, induction, and functionality of TLS during chronic inflammation, and more especially in cancer, with a particular focus on the B-cell compartment. We detail the role played by TLS B cells in anti-tumor immunity, both as antigen-presenting cells and tumor antigen-specific antibody-secreting cells, and raise the question of the capacity of chemotherapeutic and immunotherapeutic agents to induce the development of TLS within tumors. Finally, we explore how to take advantage of our knowledge on TLS B cells to develop new therapeutic tools.
Collapse
Affiliation(s)
- Claire Germain
- Laboratory Cancer, Immune Control and Escape, Cordeliers Research Center, INSERM UMRS1138 , Paris , France ; UMRS1138, University Pierre and Marie Curie , Paris , France ; UMRS1138, University Paris Descartes , Paris , France
| | - Sacha Gnjatic
- Division of Hematology, Oncology and Immunology, The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai , New York, NY , USA
| | - Marie-Caroline Dieu-Nosjean
- Laboratory Cancer, Immune Control and Escape, Cordeliers Research Center, INSERM UMRS1138 , Paris , France ; UMRS1138, University Pierre and Marie Curie , Paris , France ; UMRS1138, University Paris Descartes , Paris , France
| |
Collapse
|
18
|
Guilleminault L, Carmier D, Heuzé-Vourc'h N, Diot P, Pichon E. [Immunotherapy in non-small cell lung cancer: inhibition of PD1/PDL1 pathway]. REVUE DE PNEUMOLOGIE CLINIQUE 2015; 71:44-56. [PMID: 25687821 DOI: 10.1016/j.pneumo.2014.11.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/11/2014] [Revised: 10/13/2014] [Accepted: 11/01/2014] [Indexed: 06/04/2023]
Abstract
Despite recent advances in targeted therapy of non-small cell lung cancer (NSCLC), many patients do not benefit from these therapies. Inhibition of PD1/PDL1 is an interesting therapeutic target which restores the immune system against tumor cells. PD1 is located on lymphocytes and PDL1 on the antigen presenting cells. PD1 and PDL1 are co-inhibition molecules and their interaction results in immune tolerance against tumor cells. Anti-PD1 and anti-PDL1 antibodies have been developed to restore immune system in solid cancer including NSCLC. In phase I, studies assessing nivolumab, an anti-PD1 antibody, objective responses were observed in 13 to 18% of NSCLC patients failing previous treatment. The data obtained with anti-PDL1 antibodies is similar with objective responses ranging from 6 to 22%. The encouraging results of phase I/II studies must be confirmed in ongoing phase III studies. Anti-PD1 and anti-PDL1 antibodies exposed to new adverse events including auto-immune diseases whose support is not codified. Questions about treatment duration and criteria evaluation are not resolved. These treatments pave the way for immunomodulation in NSCLC treatment.
Collapse
Affiliation(s)
- L Guilleminault
- Service de pneumologie et d'explorations fonctionnelles, CHRU de Tours, 2, boulevard Tonnellé, 37044 Tours cedex, France; Centre d'étude des pathologies respiratoires, UMR 1100/EA6305, 37032 Tours, France; EA6305, université François-Rabelais de Tours, 37032 Tours, France.
| | - D Carmier
- Service de pneumologie et d'explorations fonctionnelles, CHRU de Tours, 2, boulevard Tonnellé, 37044 Tours cedex, France
| | - N Heuzé-Vourc'h
- Centre d'étude des pathologies respiratoires, UMR 1100/EA6305, 37032 Tours, France; EA6305, université François-Rabelais de Tours, 37032 Tours, France
| | - P Diot
- Service de pneumologie et d'explorations fonctionnelles, CHRU de Tours, 2, boulevard Tonnellé, 37044 Tours cedex, France; Centre d'étude des pathologies respiratoires, UMR 1100/EA6305, 37032 Tours, France; EA6305, université François-Rabelais de Tours, 37032 Tours, France
| | - E Pichon
- Service de pneumologie et d'explorations fonctionnelles, CHRU de Tours, 2, boulevard Tonnellé, 37044 Tours cedex, France
| |
Collapse
|
19
|
Germain C, Gnjatic S, Tamzalit F, Knockaert S, Remark R, Goc J, Lepelley A, Becht E, Katsahian S, Bizouard G, Validire P, Damotte D, Alifano M, Magdeleinat P, Cremer I, Teillaud JL, Fridman WH, Sautès-Fridman C, Dieu-Nosjean MC. Presence of B cells in tertiary lymphoid structures is associated with a protective immunity in patients with lung cancer. Am J Respir Crit Care Med 2014; 189:832-44. [PMID: 24484236 DOI: 10.1164/rccm.201309-1611oc] [Citation(s) in RCA: 531] [Impact Index Per Article: 53.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
RATIONALE It is now well established that immune responses can take place outside of primary and secondary lymphoid organs. We previously described the presence of tertiary lymphoid structures (TLS) in patients with non-small cell lung cancer (NSCLC) characterized by clusters of mature dendritic cells (DCs) and T cells surrounded by B-cell follicles. We demonstrated that the density of these mature DCs was associated with favorable clinical outcome. OBJECTIVES To study the role of follicular B cells in TLS and the potential link with a local humoral immune response in patients with NSCLC. METHODS The cellular composition of TLS was investigated by immunohistochemistry. Characterization of B-cell subsets was performed by flow cytometry. A retrospective study was conducted in two independent cohorts of patients. Antibody specificity was analyzed by ELISA. MEASUREMENTS AND MAIN RESULTS Consistent with TLS organization, all stages of B-cell differentiation were detectable in most tumors. Germinal center somatic hypermutation and class switch recombination machineries were activated, associated with the generation of plasma cells. Approximately half of the patients showed antibody reactivity against up to 7 out of the 33 tumor antigens tested. A high density of follicular B cells correlated with long-term survival, both in patients with early-stage NSCLC and with advanced-stage NSCLC treated with chemotherapy. The combination of follicular B cell and mature DC densities allowed the identification of patients with the best clinical outcome. CONCLUSIONS B-cell density represents a new prognostic biomarker for NSCLC patient survival, and makes the link between TLS and a protective B cell-mediated immunity.
Collapse
|
20
|
Abstract
Tumor cells exhibit striking changes in cell surface glycosylation as a consequence of dysregulated glycosyltransferases and glycosidases. In particular, an increase in the expression of certain sialylated glycans is a prominent feature of many transformed cells. Altered sialylation has long been associated with metastatic cell behaviors including invasion and enhanced cell survival; however, there is limited information regarding the molecular details of how distinct sialylated structures or sialylated carrier proteins regulate cell signaling to control responses such as adhesion/migration or resistance to specific apoptotic pathways. The goal of this review is to highlight selected examples of sialylated glycans for which there is some knowledge of molecular mechanisms linking aberrant sialylation to critical processes involved in metastasis.
Collapse
Affiliation(s)
- Matthew J Schultz
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, MCLM 982A 1918 University Boulevard, Birmingham, AL 35294-0005, USA
| | | | | |
Collapse
|
21
|
Abstract
BACKGROUND With our growing understanding of the immune system and mechanisms employed by tumors to evade destruction, the field of cancer immunotherapy has been revitalized. Concurrent inflammation has long been associated with follicular cell-derived thyroid cancer (FDTC). In the last decade, much research has focused on characterizing the tumor-associated immune response in patients with FDTC. SUMMARY Mast cells, natural killer cells, macrophages, dendritic cells, B cells, and T cells have been identified within FDTC-associated immune infiltrate. Collectively, these findings suggest that the immune response to FDTC is compromised and may even promote tumor progression. A more thorough characterization of the tumor-associated immune response in FDTC may lead to the development of immune-based adjuvant therapies for patients with aggressive disease. CONCLUSIONS Immune-based therapies could provide essential alternatives to patients that cannot be treated surgically, those with recurrent or persistent lymph node metastases, and those with anaplastic thyroid cancer.
Collapse
Affiliation(s)
- Jena D French
- Department of Medicine, Division of Endocrinology, Metabolism, and Diabetes, University of Colorado , Aurora, CO 80045, USA.
| |
Collapse
|
22
|
Gunderson AJ, Coussens LM. B cells and their mediators as targets for therapy in solid tumors. Exp Cell Res 2013; 319:1644-9. [PMID: 23499742 DOI: 10.1016/j.yexcr.2013.03.005] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2013] [Accepted: 03/02/2013] [Indexed: 11/25/2022]
Abstract
B cells have recently been appreciated as paracrine mediators of solid tumor development. Their ability to influence various hallmarks of cancer development, aside from antigen presentation, can be attributed to the diversity of soluble mediators they express, including cytokines and immunoglobulins, that can act directly and indirectly on the diversity of leukocyte subsets that infiltrate developing tumors, evolving neoplastic cells, as well as select T cell populations in secondary lymphoid organs and within tumor stroma. Herein, we review the literature supporting these interactions and discuss novel approaches to ameliorate protumoral B cell effects for anti-cancer therapy.
Collapse
Affiliation(s)
- Andrew J Gunderson
- Department of Cell and Developmental Biology, Oregon Health and Science University, 3181 SW Sam Jackson Park Rd, Mail Code L215, Rm 5508, Richard Jones Hall, Portland, OR 97239-3098, USA
| | - Lisa M Coussens
- Department of Cell and Developmental Biology, Oregon Health and Science University, 3181 SW Sam Jackson Park Rd, Mail Code L215, Rm 5508, Richard Jones Hall, Portland, OR 97239-3098, USA; Knight Cancer Institute, Oregon Health and Science University, 3181 SW Sam Jackson Park Rd, Mail Code L215, Rm 5508, Richard Jones Hall, Portland, OR 97239-3098, USA.
| |
Collapse
|
23
|
Liang PY, Li HY, Zhou ZY, Jin YX, Wang SX, Peng XH, Ou SJ. Overexpression of immunoglobulin G prompts cell proliferation and inhibits cell apoptosis in human urothelial carcinoma. Tumour Biol 2013; 34:1783-91. [PMID: 23483488 PMCID: PMC3661916 DOI: 10.1007/s13277-013-0717-z] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2012] [Accepted: 02/24/2013] [Indexed: 12/13/2022] Open
Abstract
Only B lymphocytes can express immunoglobulins according to the traditional immunological theories, and the expression of immunoglobulin G (IgG) messenger RNA (mRNA) and protein was found in certain human cancer cells recently. However, the expression pattern of IgG and its possible role in human urothelial carcinoma are still elusive. In this study, we investigated the expression of IgG in two human urothelial carcinoma cell lines, T24 and BIU-87, and in 56 cases of clinical urothelial carcinoma tissues. The mRNA of IgG was positively detected by in situ hybridization and reverse transcription PCR; furthermore, IgG protein was also positively detected by immunohistochemistry and Western blot. Moreover, blockade of tumor-derived IgG by either antihuman IgG antibody or antisense oligonucleotides increased cell apoptosis and inhibited cell growth in bladder cancer cell lines in vitro, and antihuman IgG antibody could suppress the growth of xenotransplant tumor in vivo. In addition, either antihuman IgG antibody or antisense oligonucleotides enhanced the sensitivity to mitomycin C in bladder cancer cell line T24. Furthermore, blockade of IgG in bladder cancer cell T24 resulted in upregulation of cleaved caspase-3 and cleaved poly(ADP-ribose) polymerase. Our results indicated that bladder cancer cells were capable of expressing IgG, and blockade of IgG expression induced cell apoptosis through activation of caspase-dependent pathway. A novel potential targeted therapy for bladder cancer will be possibly developed based on these data.
Collapse
Affiliation(s)
- Pei-Yu Liang
- Department of Urology, The Affiliated Hospital of Medical College of Hainan, NO.31 Longhua Road, Meilan District, Haikou, 570102, Hainan Province, China
| | | | | | | | | | | | | |
Collapse
|
24
|
Defendenti C, Atzeni F, Croce AM, Mussani E, Saibeni S, Bollani S, Grosso S, Almasio PL, Bruno S, Sarzi-Puttini P. Morphological distribution of μ chains and cd15 receptors in colorectal polyp and adenocarcinoma specimens. BMC Clin Pathol 2013; 13:8. [PMID: 23448299 PMCID: PMC3637070 DOI: 10.1186/1472-6890-13-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2012] [Accepted: 02/11/2013] [Indexed: 01/03/2023] Open
Abstract
Background We have recently investigated the localisation of immunoglobulin-producing cells (IPCs) in inflamed intestinal tissue samples from patients with inflammatory bowel disease (IBD), and identified two main patterns of B lymphocyte infiltration: one characterised by the moderate strong stromal localisation of small B1 cell-like IgM+/CD79+/CD20-/CD21-/CD23-/CD5 ± IPCs, and the other by the peri-glandular localisation of IPCs with irregular nuclei that had surface markers specific for a B cell subset (IgM and CD79), but quantitative differences in their λ and κ chains. The same patients were also tested for CD15+ receptors, which were localised on inflammatory cell surfaces or in the crypts of the intestinal epithelium. CD15+ receptor distribution in inflamed tissues was limited to the cell structures. The aim of the study was to analyse variations in IPCs and CD15+ cell morphology or distribution in bowel biopsy specimens taken from patients with pre-malignant polyps or adenocarcinomas. Methods IPCs were analysed by means of immunofluorescence using polyclonal goat anti-human μ chains. The pre-malignant polyp specimens were tested for B cell surface phenotype λ and κ chains, CD79, CD20, CD21 and CD23 using an immunoperoxidase method. CD15+ cells were evaluated using the immunoperoxidase method and monoclonal anti-CD15 IgM. Results The study involved 14 patients (four with pre-malignant polyps and 10 with colorectal adenocarcinomas). The distribution of μ chains and CD15 markers varied in all of the biopsies, but delineated normal cell structures in the pre-malignant polyp specimens. B cell surface phenotype analysis of μ chain-positive cells identified a subset of CD79+/CD20-/CD21-/CD23- IPCs. The IPCs in certain areas showed the sporadic disintegration of inflammatory cell membranes or the accumulation of fluorescence in individual cells. IPC membrane disintegration was particularly marked in all of the adenocarcinoma samples, in which the CD15 markers also showed epithelial cell involvement. Furthermore, six of the ten adenocarcinoma samples had atypical and reorganised membranes that expressed an excess of both receptors and isolated small portions of tissue within the tumour. Conclusion The findings of this preliminary morphological study suggest the presence of membrane disintegration and remodelling mechanisms in the tumours. The newly-formed membranes expressed high concentrations of inflammatory cell receptors that can confer adhesive properties.
Collapse
|
25
|
Szubert S, Michalak S, Szpurek D, Moszynski R, Krygowska-Zielinska J, Sajdak S. Anti-ovarian antibodies in sera of patients with ovarian tumors. Immunol Lett 2012; 148:133-7. [PMID: 23026238 DOI: 10.1016/j.imlet.2012.09.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2012] [Revised: 09/06/2012] [Accepted: 09/19/2012] [Indexed: 12/31/2022]
Abstract
AIM OF PAPER The purpose of this study was to assess the presence of anti-ovarian autoantibodies in sera of patients with ovarian tumors. MATERIALS AND METHODS The study group consisted of 82 patients treated at the Division of Gynecological Surgery, Poznan University of Medical Sciences, Poland in 2007-2011. 46 patients with malignant ovarian tumors and 36 patients diagnosed with benign ovarian tumor were included into the study. Age-matched healthy control groups consisted of 15 women and 19 men. Anti-ovarian autoantibodies in serum were assessed with the use of indirect immunofluorescence. The presence of anti-ovarian autoantibodies in serum was correlated with clinical and histopathological features of the disease. RESULTS Serum anti-ovarian autoantibodies were found in 59% (27/46) of patients with malignant ovarian tumors, compared to 55% (20/36) and 26% (4/15) of patients with benign ovarian tumors and healthy female controls respectively. The difference was statistically significant (P=0.04). All serum samples from the male controls were negative. Anti-ovarian antibodies occurred more often among advanced stage (III and IV stage according to FIGO, P=0.037) and grade 3 (P=0.049) ovarian cancers, however, there were no differences in median progression-free survival (P=0.388). The presence of anti-ovarian antibodies was neither influenced by histopathological type of the tumor, menopausal status, presence of ascites nor CA125 levels. Seropositivity for anti-ovarian antibodies was correlated positively with patients' age and negatively with tumor size. CONCLUSIONS Anti-ovarian autoantibodies develop with higher frequency in ovarian cancer comparing to healthy controls, however with similar proportion to benign ovarian tumors patients. The presence of antibodies against normal ovarian tissue correlates with ovarian cancer aggressiveness.
Collapse
Affiliation(s)
- Sebastian Szubert
- Division of Gynecological Surgery, Poznan University of Medical Sciences, Poland.
| | | | | | | | | | | |
Collapse
|
26
|
Evaluation of humoral immunity in oral cancer patients from a nigerian referral centre. J Maxillofac Oral Surg 2012; 12:410-3. [PMID: 24431879 DOI: 10.1007/s12663-012-0440-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2012] [Accepted: 09/03/2012] [Indexed: 10/27/2022] Open
Abstract
The immune system is central in the body's defense against non-self. Immunoglobulins and acute phase proteins have been reported to play active roles in carcinogenesis. This prospective longitudinal study was carried out to determine the state of humoral immunity in Nigerian oral cancer patients relative to controls. Twenty newly diagnosed untreated cases of oral squamous cell carcinoma recruited from our centre were included in the study. The controls included 20 apparently healthy and HIV negative volunteers. Serum immunoglobulin classes and acute phase proteins were measured using immunoplates. IgA and ceruloplasmin showed statistically significantly elevated levels in the patients compared with controls, while increases in IgM and IgG were insignificant. The raised levels suggest a role for immunoglobulin A and ceruloplasmin in the mechanisms involved in oral cancers. Findings from this study are similar to that reported elsewhere in the literature. Further work is needed to ascertain the role and usefulness of immunoglobulins and acute phase proteins in staging, disease monitoring, therapy and prognostication.
Collapse
|
27
|
Yang M, Yan Y, Fang M, Wan M, Wu X, Zhang X, Zhao T, Wei H, Song D, Wang L, Yu Y. MF59 formulated with CpG ODN as a potent adjuvant of recombinant HSP65-MUC1 for inducing anti-MUC1+ tumor immunity in mice. Int Immunopharmacol 2012; 13:408-16. [PMID: 22595192 PMCID: PMC7106219 DOI: 10.1016/j.intimp.2012.05.003] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2012] [Revised: 04/23/2012] [Accepted: 05/03/2012] [Indexed: 12/31/2022]
Abstract
MF59 is an oil-in-water emulsion adjuvant approved for influenza vaccines for human use in Europe. Due to its Th2 inducing properties, MF59 is seldom tested for cancer vaccines. In this study, MF59 formulated with a C-type CpG oligodeoxynucleotide (YW002) was tested for its Th1 adjuvant activity to induce immune responses to HSP65-MUC1, a recombinant fusion protein incorporating a mycobacterial heat shock protein (HSP65) and mucin 1, cell surface associated (MUC1) derived peptide. Combination of YW002 with MF59 (MF59-YW002) could confer a potent Th1 biasing property to the adjuvant, which enhanced the immunogenicity of HSP65-MUC1 to induce significantly higher levels of specific IgG2c, increased IFN-γ mRNA expression in splenocytes and the generation of antigen-specific cytotoxic T lymphocytes in mice. When prophylactically applied, MF59-YW002 adjuvant containing HSP65-MUC1 inhibited the growth of MUC1+ B16 melanoma and prolonged the survival of tumor-bearing mice. In contrast, adjuvant containing MF59 with HSP65-MUC1 in the absence of YW002, promoted the growth of MUC1+ B16 melanoma in mice. These results suggest that MF59 plus CpG oligodeoxynucleotide might be developed as an efficient adjuvant for tumor vaccines against melanoma, and possibly other tumors.
Collapse
Affiliation(s)
- Ming Yang
- Department of Immunology, Norman Bethune College of Medicine, Jilin University, Changchun 130021, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Abstract
Tumor-associated myeloid cells have been implicated in regulating many of the “hallmarks of cancer” and thus fostering solid tumor development and metastasis. However, the same innate leukocytes also participate in anti-tumor immunity and restraint of malignant disease. While many factors regulate the propensity of myeloid cells to promote or repress cancerous growths, polarized adaptive immune responses by B and T lymphocytes have been identified as regulators of many aspects of myeloid cell biology by specifically regulating their functional capabilities. Here, we detail the diversity of heterogeneous B and T lymphocyte populations and their impacts on solid tumor development through their abilities to regulate myeloid cell function in solid tumors.
Collapse
|
29
|
Nesslinger NJ, Ng A, Tsang KY, Ferrara T, Schlom J, Gulley JL, Nelson BH. A viral vaccine encoding prostate-specific antigen induces antigen spreading to a common set of self-proteins in prostate cancer patients. Clin Cancer Res 2010; 16:4046-56. [PMID: 20562209 DOI: 10.1158/1078-0432.ccr-10-0948] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
PURPOSE We previously reported a randomized phase II clinical trial combining a poxvirus-based vaccine encoding prostate-specific antigen (PSA) with radiotherapy in patients with localized prostate cancer. Here, we investigate whether vaccination against PSA induced immune responses to additional tumor-associated antigens and how this influenced clinical outcome. EXPERIMENTAL DESIGN Pretreatment and posttreatment serum samples from patients treated with vaccine + external beam radiation therapy (EBRT) versus EBRT alone were evaluated by Western blot and serologic screening of a prostate cancer cDNA expression library (SEREX) to assess the development of treatment-associated autoantibody responses. RESULTS Western blotting revealed treatment-associated autoantibody responses in 15 of 33 (45.5%) patients treated with vaccine + EBRT versus 1 of 8 (12.5%) treated with EBRT alone. SEREX screening identified 18 antigens, which were assembled on an antigen array with 16 previously identified antigens. Antigen array screening revealed that 7 of 33 patients (21.2%) treated with vaccine + EBRT showed a vaccine-associated autoantibody response to four ubiquitously expressed self-antigens: DIRC2, NDUFS1, MRFAP1, and MATN2. These responses were not seen in patients treated with EBRT alone, or other control groups. Patients with autoantibody responses to this panel of antigens had a trend toward decreased biochemical-free survival. CONCLUSIONS Vaccine + EBRT induced antigen spreading in a large proportion of patients. A subset of patients developed autoantibodies to a panel of four self-antigens and showed a trend toward inferior outcomes. Thus, cancer vaccines directed against tumor-specific antigens can trigger autoantibody responses to self-proteins, which may influence the efficacy of vaccination.
Collapse
Affiliation(s)
- Nancy J Nesslinger
- Trev & Joyce Deeley Research Centre, BC Cancer Agency-Vancouver Island Centre, 2410 Lee Avenue, Victoria, British Columbia, Canada
| | | | | | | | | | | | | |
Collapse
|
30
|
Gerber HP, Olazoglu E, Grewal IS. Targeting inflammatory cells to improve anti-VEGF therapies in oncology. Recent Results Cancer Res 2010; 180:185-200. [PMID: 20033384 DOI: 10.1007/978-3-540-78281-0_11] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Vascular endothelial growth factor A (VEGF-A) is a well-characterized regulator of physiological and pathological angiogenesis. Multiple therapeutic compounds interfering with VEGF-A-regulated signal transduction pathways are currently being developed for the treatment of neoplasias and other malignancies associated with pathological angiogenesis. A major challenge in developing anti-VEGF therapies are tumor intrinsic refractoriness and the emergence of treatment-induced resistance. A variety of molecular and cellular mechanisms contribute to tumor angiogenesis, including the recruitment of bone marrow (BM)-derived endothelial cell progenitors (EPCs) and inflammatory cells to the tumor mass. Among the latter, two types of tumor infiltrating, inflammatory cells were recently identified to mediate refractoriness to anti-VEGF treatment: CD11b + Gr1+ myeloid derived suppressor cells (MDSC) and tumor-associated macrophages (TAMs). In this chapter, we review some of the inflammatory components regulating tumor angiogenesis and their roles in mediating refractoriness toward anti-VEGF treatment. In addition, we discuss potential therapeutic strategies targeting angiogenic pathways regulated by inflammatory cells. A better understanding of the biological and molecular events involved in mediating refractoriness to anti-VEGF treatment may help to further improve therapeutic strategies targeting tumor angiogenesis.
Collapse
Affiliation(s)
- Hans-Peter Gerber
- Sr Dir Discovery Tumor Prog, Pharma, Research & Development, Pearl River, NY, USA.
| | | | | |
Collapse
|
31
|
Andreu P, Johansson M, Affara NI, Pucci F, Tan T, Junankar S, Korets L, Lam J, Tawfik D, DeNardo DG, Naldini L, de Visser KE, De Palma M, Coussens LM. FcRgamma activation regulates inflammation-associated squamous carcinogenesis. Cancer Cell 2010; 17:121-34. [PMID: 20138013 PMCID: PMC3082507 DOI: 10.1016/j.ccr.2009.12.019] [Citation(s) in RCA: 433] [Impact Index Per Article: 30.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2009] [Revised: 11/14/2009] [Accepted: 12/09/2009] [Indexed: 01/15/2023]
Abstract
Chronically activated leukocytes recruited to premalignant tissues functionally contribute to cancer development; however, mechanisms underlying pro- versus anti-tumor programming of neoplastic tissues by immune cells remain obscure. Using the K14-HPV16 mouse model of squamous carcinogenesis, we report that B cells and humoral immunity foster cancer development by activating Fcgamma receptors (FcgammaRs) on resident and recruited myeloid cells. Stromal accumulation of autoantibodies in premalignant skin, through their interaction with activating FcgammaRs, regulate recruitment, composition, and bioeffector functions of leukocytes in neoplastic tissue, which in turn promote neoplastic progression and subsequent carcinoma development. These findings support a model in which B cells, humoral immunity, and activating FcgammaRs are required for establishing chronic inflammatory programs that promote de novo carcinogenesis.
Collapse
MESH Headings
- Animals
- B-Lymphocytes/immunology
- B-Lymphocytes/metabolism
- CD11b Antigen/metabolism
- Carcinoma, Squamous Cell/blood supply
- Carcinoma, Squamous Cell/immunology
- Carcinoma, Squamous Cell/metabolism
- Carcinoma, Squamous Cell/pathology
- Immunity, Humoral/physiology
- Mast Cells/immunology
- Mast Cells/metabolism
- Mast Cells/pathology
- Mice
- Mice, Transgenic
- Models, Biological
- Myeloid Cells/immunology
- Myeloid Cells/metabolism
- Neoplasms, Glandular and Epithelial/blood supply
- Neoplasms, Glandular and Epithelial/immunology
- Neoplasms, Glandular and Epithelial/metabolism
- Neoplasms, Glandular and Epithelial/pathology
- Neovascularization, Pathologic
- Receptors, IgG/metabolism
- Receptors, IgG/physiology
Collapse
Affiliation(s)
- Pauline Andreu
- Department of Pathology University of California, San Francisco, San Francisco, CA 94143, USA
| | - Magnus Johansson
- Department of Pathology University of California, San Francisco, San Francisco, CA 94143, USA
| | - Nesrine I. Affara
- Department of Pathology University of California, San Francisco, San Francisco, CA 94143, USA
| | - Ferdinando Pucci
- Angiogenesis and Tumor Targeting Research Unit, San Raffaele-Telethon Institute for Gene Therapy and Vita-Salute San Raffaele University, San Raffaele Institute, Milan, 20132, Italy
| | - Tingting Tan
- Department of Pathology University of California, San Francisco, San Francisco, CA 94143, USA
| | - Simon Junankar
- Department of Pathology University of California, San Francisco, San Francisco, CA 94143, USA
| | - Lidiya Korets
- Department of Pathology University of California, San Francisco, San Francisco, CA 94143, USA
| | - Julia Lam
- Department of Pathology University of California, San Francisco, San Francisco, CA 94143, USA
| | - David Tawfik
- Department of Pathology University of California, San Francisco, San Francisco, CA 94143, USA
| | - David G. DeNardo
- Department of Pathology University of California, San Francisco, San Francisco, CA 94143, USA
| | - Luigi Naldini
- Angiogenesis and Tumor Targeting Research Unit, San Raffaele-Telethon Institute for Gene Therapy and Vita-Salute San Raffaele University, San Raffaele Institute, Milan, 20132, Italy
| | - Karin E. de Visser
- Division of Molecular Biology, The Netherlands Cancer Institute, Amsterdam, 1066 CX, the Netherlands
| | - Michele De Palma
- Angiogenesis and Tumor Targeting Research Unit, San Raffaele-Telethon Institute for Gene Therapy and Vita-Salute San Raffaele University, San Raffaele Institute, Milan, 20132, Italy
| | - Lisa M. Coussens
- Department of Pathology University of California, San Francisco, San Francisco, CA 94143, USA
- Helen Diller Family Comprehensive Cancer Center University of California, San Francisco, San Francisco, CA 94143, USA
- Correspondence:
| |
Collapse
|
32
|
Tumor-associated macrophages: effectors of angiogenesis and tumor progression. Biochim Biophys Acta Rev Cancer 2009; 1796:11-8. [PMID: 19269310 DOI: 10.1016/j.bbcan.2009.02.004] [Citation(s) in RCA: 174] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2008] [Revised: 02/06/2009] [Accepted: 02/18/2009] [Indexed: 02/08/2023]
Abstract
Tumor-associated macrophages (TAMs) are a prominent inflammatory cell population in many tumor types residing in both perivascular and avascular, hypoxic regions of these tissues. Analysis of TAMs in human tumor biopsies has shown that they express a variety of tumor-promoting factors and evidence from transgenic murine tumor models has provided unequivocal evidence for the importance of these cells in driving angiogenesis, lymphangiogenesis, immunosuppression, and metastasis. This review will summarize the mechanisms by which monocytes are recruited into tumors, their myriad, tumor-promoting functions within tumors, and the influence of the tumor microenvironment in driving these activities. We also discuss recent attempts to both target/destroy TAMs and exploit them as delivery vehicles for anti-cancer gene therapy.
Collapse
|
33
|
Johansson M, Denardo DG, Coussens LM. Polarized immune responses differentially regulate cancer development. Immunol Rev 2009; 222:145-54. [PMID: 18363999 DOI: 10.1111/j.1600-065x.2008.00600.x] [Citation(s) in RCA: 153] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Tumor-associated immune responses assert varied responses toward developing neoplasms that can either act to eradicate malignant cells via engagement of potent cytotoxic programs or alternatively enhance tumor growth through release of multifunctional pro-tumor mediators. Seemingly paradoxical, these disparate activities reflect a continuum of polarization (or activation) states possible for distinct leukocyte subsets that demonstrate tissue, organ, and tumor selectivity. Herein, we review clinical and experimental studies investigating cellular and molecular mechanisms utilized by neoplastic tissues to alternatively polarize immune responses that favor either pro- or anti-tumor immunity.
Collapse
Affiliation(s)
- Magnus Johansson
- Department of Pathology, University of California, San Francisco, San Francisco, CA 94143, USA
| | | | | |
Collapse
|
34
|
DeNardo DG, Coussens LM. Inflammation and breast cancer. Balancing immune response: crosstalk between adaptive and innate immune cells during breast cancer progression. Breast Cancer Res 2008; 9:212. [PMID: 17705880 PMCID: PMC2206719 DOI: 10.1186/bcr1746] [Citation(s) in RCA: 498] [Impact Index Per Article: 31.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Recent insights into the molecular and cellular mechanisms underlying cancer development have revealed that immune cells functionally regulate epithelial cancer development and progression. Moreover, accumulated clinical and experimental data indicate that the outcome of an immune response toward an evolving breast neoplasm is largely determined by the type of immune response elicited. Acute tumor-directed immune responses involving cytolytic T lymphocytes appear to protect against tumor development, whereas immune responses involving chronic activation of humoral immunity, infiltration by Th2 cells, and protumor-polarized innate inflammatory cells result in the promotion of tumor development and disease progression. Herein we review this body of literature and summarize important new findings revealing the paradoxical role of innate and adaptive leukocytes as regulators of breast carcinogenesis.
Collapse
Affiliation(s)
- David G DeNardo
- Department of Pathology, University of California, San Francisco, 2340 Sutter Street, San Francisco, CA 94143, USA
| | - Lisa M Coussens
- Department of Pathology, University of California, San Francisco, 2340 Sutter Street, San Francisco, CA 94143, USA
- Cancer Research Institute, University of California, San Francisco, 2340 Sutter Street, San Francisco, CA 94143, USA
- Comprehensive Cancer Center, University of California, San Francisco, 2340 Sutter Street, San Francisco, CA 94143, USA
| |
Collapse
|
35
|
Abstract
The concept that the immune system can recognise tumour cells and either eliminate them (tumour immune surveillance) or select for immunologically resistant variants (immunoediting) is gaining general acceptance by immunologists. In terms of an adaptive immune response to cancer, however, much of the research has focused on the response of cytotoxic CD8+ T lymphocytes to tumour-specific antigens and the production of Th1 cytokines by CD4+ and CD8+ T cells. In contrast, Th2-mediated immunity has traditionally been viewed as favouring tumour growth, both by promoting angiogenesis and by inhibiting cell-mediated immunity and subsequent tumour cell killing. While there is evidence that components of type 2 inflammation, such as B cells and interleukin-10, do promote tumour growth, there are also many studies demonstrating the anti-tumour activity of CD4+ Th2 cells, particularly in collaboration with tumour-infiltrating granulocytes, such as eosinophils. In this review, we examine all the components of type 2 immunity and their effects on tumour growth. Collectively, from this analysis, we conclude that there is a great potential for the development of Th2-mediated immunotherapies that harness the cytotoxic activity of eosinophils.
Collapse
Affiliation(s)
- J I Ellyard
- Cancer and Vascular Biology Group, Division of Immunology and Genetics, John Curtin School of Medical Research, Australian National University, Canberra City, ACT 2601, Australia
| | | | | |
Collapse
|
36
|
Johansson M, Tan T, de Visser KE, Coussens LM. Immune cells as anti-cancer therapeutic targets and tools. J Cell Biochem 2007; 101:918-26. [PMID: 17265430 DOI: 10.1002/jcb.21230] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Chronic inflammation is a contributing factor to overall cancer risk as well as cancer promotion and progression; however, pathways regulating onset of cancer-promoting inflammatory responses are still poorly understood. Clinical data suggest that deficient anti-tumor cell-mediated immunity, in combination with enhanced pro-tumor humoral and/or innate immunity (inflammation), are significant factors influencing malignant outcome. Here, we discuss therapeutic implications from clinical data and experimental studies using de novo immune-competent mouse models of cancer development that together are revealing molecular and cellular mechanisms underlying interactions between immune cells and evolving neoplastic cells that regulate cancer outcome. Understanding the functionally significant links between adaptive and innate immunity that regulate cancer development will open new therapeutic opportunities to manipulate aspects of immunobiology and minimize lethal effects of cancer development.
Collapse
Affiliation(s)
- Magnus Johansson
- Department of Pathology, University of California, San Francisco, San Francisco, CA 94143, USA
| | | | | | | |
Collapse
|
37
|
Tan TT, Coussens LM. Humoral immunity, inflammation and cancer. Curr Opin Immunol 2007; 19:209-16. [PMID: 17276050 DOI: 10.1016/j.coi.2007.01.001] [Citation(s) in RCA: 299] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2006] [Accepted: 01/25/2007] [Indexed: 01/28/2023]
Abstract
Clinical and experimental data now clearly indicate that chronic inflammation significantly contributes to cancer development. Emerging out of these studies is an appreciation that persistent humoral immune responses exacerbate recruitment and activation of innate immune cells in neoplastic microenvironments where they regulate tissue remodeling, pro-angiogenic and pro-survival pathways that together potentiate cancer development. Population-based studies examining individuals with chronic inflammatory disorders have revealed that states of suppressed cellular immunity, in combination with enhanced humoral immunity and humoral immunity-associated cytokines, cooperate and effectively suppress anti-tumor immune responses while simultaneously enhancing angiogenesis and presumably overall cancer risk in afflicted tissue. In addition, studies in transgenic mouse models of de novo organ-specific cancer development have revealed that inflammation mediated by immunoglobulins and immune complexes might be functionally significant parameters of tumor promotion and progression. These recent advances support the hypothesis that enhanced states of local humoral and innate immune activation, in combination with suppressed cellular immunity and failed cytotoxic T cell anti-tumor immunity, alter cancer risk and therefore represent powerful targets for anti-cancer immunotherapeutics.
Collapse
Affiliation(s)
- Ting-Ting Tan
- Department of Pathology, University of California, San Francisco 2340 Sutter St, San Francisco, CA 94143, USA
| | | |
Collapse
|
38
|
Abstract
The main function of the mammalian immune system is to monitor tissue homeostasis, to protect against invading or infectious pathogens and to eliminate damaged cells. Therefore, it is surprising that cancer occurs with such a high frequency in humans. Recent insights that have been gained from clinical studies and experimental mouse models of carcinogenesis expand our understanding of the complex relationship between immune cells and developing tumours. Here, we examine the paradoxical role of adaptive and innate leukocytes as crucial regulators of cancer development and highlight recent insights that have been gained by manipulating immune responses in mouse models of de novo and spontaneous tumorigenesis.
Collapse
Affiliation(s)
- Karin E de Visser
- Department of Molecular Biology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands
| | | | | |
Collapse
|
39
|
Abstract
Chemoprevention of cancer is a reality today. Prevention of breast cancer with tamoxifen, of squamous cell skin cancer with actinic keratosis by diclofenac gel and in familial polyposis with anti-inflammatory drug (COX-2) celecoxib is considered of health care clinical use. The latter has received enormous attention by cancer investigators due to the attractiveness of its action mechanism and its possibilities of future clinical use in different neoplasms. Other anti-inflammatory drugs such as aspirin and sulindac also have a proven role in chemoprevention of cancer by cycloosygenase inhibition or of related substances. The review of the mechanisms by which these substances act gives us a clear idea of what it is and what the chemoprevention will be.
Collapse
Affiliation(s)
- J J Grau de Castro
- Universidad de Barcelona, Servicio de Oncología, Hospital Clínic, Barcelona.
| |
Collapse
|
40
|
de Visser KE, Coussens LM. The interplay between innate and adaptive immunity regulates cancer development. Cancer Immunol Immunother 2005; 54:1143-52. [PMID: 15889249 PMCID: PMC11032791 DOI: 10.1007/s00262-005-0702-5] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2005] [Accepted: 03/09/2005] [Indexed: 12/14/2022]
Abstract
There is increasing clinical and experimental evidence that inflammation and cancer are causally linked. Much progress has been made in understanding how inflammatory cells contribute to cancer development; however, it is still largely unknown which molecular mechanisms are responsible for initiation and maintenance of chronic inflammation associated with developing neoplasms. This review will discuss how the adaptive and innate immune systems interact during physiological and chronic inflammation, with a focus on studies revealing new insights into the role of adaptive immune cells as important regulators of chronic inflammation-associated carcinogenesis. We will speculate on whether current knowledge about the dysregulated interplay between adaptive and innate immunity during chronic inflammatory disorders might be useful in understanding and targeting the underlying mechanisms of chronic inflammation-associated neoplastic progression.
Collapse
Affiliation(s)
- K E de Visser
- Cancer Research Institute, University of California, San Francisco, 2340 Sutter St., N-261, San Francisco, CA 94143, USA.
| | | |
Collapse
|
41
|
de Visser KE, Korets LV, Coussens LM. De novo carcinogenesis promoted by chronic inflammation is B lymphocyte dependent. Cancer Cell 2005; 7:411-23. [PMID: 15894262 DOI: 10.1016/j.ccr.2005.04.014] [Citation(s) in RCA: 573] [Impact Index Per Article: 30.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2004] [Revised: 02/25/2005] [Accepted: 04/13/2005] [Indexed: 12/15/2022]
Abstract
Chronic inflammation predisposes tissue to cancer development; however, regulatory mechanisms underlying recruitment of innate leukocytes toward developing neoplasms are obscure. We report that genetic elimination of mature T and B lymphocytes in a transgenic mouse model of inflammation-associated de novo epithelial carcinogenesis, e.g., K14-HPV16 mice, limits neoplastic progression to development of epithelial hyperplasias that fail to recruit innate immune cells. Adoptive transfer of B lymphocytes or serum from HPV16 mice into T and B cell-deficient/HPV16 mice restores innate immune cell infiltration into premalignant tissue and reinstates necessary parameters for full malignancy, e.g., chronic inflammation, angiogenic vasculature, hyperproliferative epidermis. These findings support a model in which B lymphocytes are required for establishing chronic inflammatory states that promote de novo carcinogenesis.
Collapse
Affiliation(s)
- Karin E de Visser
- Cancer Research Institute, University of California, San Francisco, California 94143, USA
| | | | | |
Collapse
|
42
|
Wen YJ, Mancino A, Pashov A, Whitehead T, Stanley J, Kieber-Emmons T. Antigen binding of human IgG Fabs mediate ERK-associated proliferation of human breast cancer cells. DNA Cell Biol 2005; 24:73-84. [PMID: 15699628 DOI: 10.1089/dna.2005.24.73] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Serum-circulating antibody can be linked to poor outcomes in some cancer patients. To investigate the role of human antibodies in regulating tumor cell growth, we constructed a recombinant cDNA expression library of human IgG Fab from a patient with breast cancer. Clones were screened from the library with breast tumor cell lysate. Sequence analysis of the clones showed somatic hypermutations when compared to their closest VH/VL germ-line genes. Initial characterizations focused on five clones. All tested clones displayed stronger binding to antigen derived from primary breast cancers and established breast cancer cell lines than to normal breast tissues. In vitro functional studies showed that four out of five tested clones could stimulate the growth of MDA-MB-231 breast cancer cell lines, and one out of five was able to promote MCF-7 cell growth as well. Involvement of ERK2 pathway was observed. By 1H-NMR spectra and Western blot analysis, it was evident that two tested antibody Fabs are capable of interacting with sialic acid. Our study suggests a possible role for human antibody in promoting tumor cell growth by direct binding of IgG Fab to breast tumor antigen. Such studies prompt speculation regarding the role of serum antibodies in mediating tumor growth as well as their contribution to disease progression.
Collapse
Affiliation(s)
- Yue-Jin Wen
- Department of Pathology, Arkansas Cancer Research Center, University of Arkansas for Medical Sciences, Little Rock, Arkansas 72205, USA
| | | | | | | | | | | |
Collapse
|
43
|
Guillem EB, Sampsel JW. Immune-promoted tumor cell invasion and metastasis. New considerations in cancer therapy. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2004; 532:153-73. [PMID: 12908556 DOI: 10.1007/978-1-4615-0081-0_13] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/04/2023]
|
44
|
Moldovan NI. Role of monocytes and macrophages in adult angiogenesis: a light at the tunnel's end. JOURNAL OF HEMATOTHERAPY & STEM CELL RESEARCH 2002; 11:179-94. [PMID: 11983092 DOI: 10.1089/152581602753658394] [Citation(s) in RCA: 58] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
In spite of sustained efforts, there are still gaps in our understanding of angiogenesis as it takes place in vivo. Older observations and a number of recent developments strongly involve the blood mononuclear cell population, collectively known as monocytes (MC), in the normal and pathological adult angiogenesis. An emerging paradigm should eventually incorporate the established biochemical cross talk between MC and their descendents, tissular macrophages (Mph), and the endothelial cells (EC); additionally, it should account for both the intercellular cooperation at the morphological level and the phenotypic overlap between the two cell populations. This focused review puts together the pieces of this puzzle in such a way as to suggest an alternative angiogenic model applicable to adult animals, and particularly to pathological conditions. A working hypothesis is put forward, which is centered on the preformation of capillary lumen as a "tunnel" drilled by penetrating MC/Mph. The tunnels may be colonized in a later stage by sprouts, circulating progenitor endothelial cells (CPEC) or transdifferentiated EC. Thus, MC/Mph are suggested to be included among the targets of therapeutic manipulation of angiogenesis.
Collapse
Affiliation(s)
- Nicanor I Moldovan
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH 43210, USA.
| |
Collapse
|
45
|
Nelson MB, Nyhus JK, Oravecz-Wilson KI, Barbera-Guillem E. Tumor cells express FcgammaRI which contributes to tumor cell growth and a metastatic phenotype. Neoplasia 2001; 3:115-24. [PMID: 11420747 PMCID: PMC1505421 DOI: 10.1038/sj.neo.7900140] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2000] [Accepted: 11/28/2000] [Indexed: 11/09/2022] Open
Abstract
High levels of circulating immune complexes containing tumor-associated antigens are associated with a poor prognosis for individuals with cancer. The ability of B cells, previously exposed to tumor-associated antigens, to promote both in vitro and in vivo tumor growth formed the rationale to evaluate the mechanism by which immune complexes may promote tumor growth. In elucidating this mechanism, FcgammaRI expression by tumor cells was characterized by flow cytometry, polymerase chain reaction, and sequence analysis. Immune complexes containing shed tumor antigen and anti-shed tumor antigen Ab cross-linked FcgammaRI-expressing tumor cells, which resulted in an induction of tumor cell proliferation and of shed tumor antigen production. Use of selective tyrosine kinase inhibitors demonstrated that tumor cell proliferation induced by immune complex cross-linking of FcgammaRI is dependent on the tyrosine kinase signal transduction pathway. A selective inhibitor of phosphatidylinositol-3 kinase also inhibited this induction of tumor cell proliferation. These findings support a role for immune complexes and FcgammaRI expression by tumor cells in augmentation of tumor growth and a metastatic phenotype.
Collapse
Affiliation(s)
- M B Nelson
- BioCrystal Ltd., Westerville, OH 43082-8888, USA
| | | | | | | |
Collapse
|