1
|
Kiaheyrati N, Babaei A, Ranji R, Bahadoran E, Taheri S, Farokhpour Z. Cancer therapy with the viral and bacterial pathogens: The past enemies can be considered the present allies. Life Sci 2024; 349:122734. [PMID: 38788973 DOI: 10.1016/j.lfs.2024.122734] [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: 03/02/2024] [Revised: 05/14/2024] [Accepted: 05/15/2024] [Indexed: 05/26/2024]
Abstract
Cancer continues to be one of the leading causes of mortality worldwide despite significant advancements in cancer treatment. Many difficulties have arisen as a result of the detrimental consequences of chemotherapy and radiotherapy as a common cancer therapy, such as drug inability to penetrate deep tumor tissue, and also the drug resistance in tumor cells continues to be a major concern. These obstacles have increased the need for the development of new techniques that are more selective and effective against cancer cells. Bacterial-based therapies and the use of oncolytic viruses can suppress cancer in comparison to other cancer medications. The tumor microenvironment is susceptible to bacterial accumulation and proliferation, which can trigger immune responses against the tumor. Oncolytic viruses (OVs) have also gained considerable attention in recent years because of their potential capability to selectively target and induce apoptosis in cancer cells. This review aims to provide a comprehensive summary of the latest literature on the role of bacteria and viruses in cancer treatment, discusses the limitations and challenges, outlines various strategies, summarizes recent preclinical and clinical trials, and emphasizes the importance of optimizing current strategies for better clinical outcomes.
Collapse
Affiliation(s)
- Niloofar Kiaheyrati
- Medical Microbiology Research Center, Qazvin University of Medical Sciences, Qazvin, Iran; Department of Microbiology and Immunology, School of Medicine, Qazvin University of Medical Science, Qazvin, Iran
| | - Abouzar Babaei
- Medical Microbiology Research Center, Qazvin University of Medical Sciences, Qazvin, Iran; Department of Microbiology and Immunology, School of Medicine, Qazvin University of Medical Science, Qazvin, Iran.
| | - Reza Ranji
- Department of Genetics, Faculty of Sciences, Tarbiat Modares University, Tehran, Iran
| | - Ensiyeh Bahadoran
- School of Medicine, Qazvin University of Medical Science, Qazvin, Iran
| | - Shiva Taheri
- Medical Microbiology Research Center, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Zahra Farokhpour
- Medical Microbiology Research Center, Qazvin University of Medical Sciences, Qazvin, Iran
| |
Collapse
|
2
|
Yu Q, Ding J, Li S, Li Y. Autophagy in cancer immunotherapy: Perspective on immune evasion and cell death interactions. Cancer Lett 2024; 590:216856. [PMID: 38583651 DOI: 10.1016/j.canlet.2024.216856] [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: 03/04/2024] [Revised: 03/22/2024] [Accepted: 04/02/2024] [Indexed: 04/09/2024]
Abstract
Both the innate and adaptive immune systems work together to produce immunity. Cancer immunotherapy is a novel approach to tumor suppression that has arisen in response to the ineffectiveness of traditional treatments like radiation and chemotherapy. On the other hand, immune evasion can diminish immunotherapy's efficacy. There has been a lot of focus in recent years on autophagy and other underlying mechanisms that impact the possibility of cancer immunotherapy. The primary feature of autophagy is the synthesis of autophagosomes, which engulf cytoplasmic components and destroy them by lysosomal degradation. The planned cell death mechanism known as autophagy can have opposite effects on carcinogenesis, either increasing or decreasing it. It is autophagy's job to maintain the balance and proper functioning of immune cells like B cells, T cells, and others. In addition, autophagy controls whether macrophages adopt the immunomodulatory M1 or M2 phenotype. The ability of autophagy to control the innate and adaptive immune systems is noteworthy. Interleukins and chemokines are immunological checkpoint chemicals that autophagy regulates. Reducing antigen presentation to induce immunological tolerance is another mechanism by which autophagy promotes cancer survival. Therefore, targeting autophagy is of importance for enhancing potential of cancer immunotherapy.
Collapse
Affiliation(s)
- Qiang Yu
- Department of Digestive Surgery, Xijing Hospital, Air Force Medical University, Xi'an, 710032, China
| | - Jiajun Ding
- Department of Thyroid, Breast and Vascular Surgery, Xijing Hospital, Air Force Medical University, Xi'an, 710032, China
| | - Shisen Li
- Department of Digestive Surgery, Xijing Hospital, Air Force Medical University, Xi'an, 710032, China
| | - Yunlong Li
- Department of Digestive Surgery, Xijing Hospital, Air Force Medical University, Xi'an, 710032, China.
| |
Collapse
|
3
|
Wang Y, Yao T, Lin Y, Ge H, Huang B, Gao Y, Wu J. Association between gut microbiota and pan-dermatological diseases: a bidirectional Mendelian randomization research. Front Cell Infect Microbiol 2024; 14:1327083. [PMID: 38562964 PMCID: PMC10982508 DOI: 10.3389/fcimb.2024.1327083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Accepted: 03/05/2024] [Indexed: 04/04/2024] Open
Abstract
Background Gut microbiota has been associated with dermatological problems in earlier observational studies. However, it is unclear whether gut microbiota has a causal function in dermatological diseases. Methods Thirteen dermatological diseases were the subject of bidirectional Mendelian randomization (MR) research aimed at identifying potential causal links between gut microbiota and these diseases. Summary statistics for the Genome-Wide Association Study (GWAS) of gut microbiota and dermatological diseases were obtained from public datasets. With the goal of evaluating the causal estimates, five acknowledged MR approaches were utilized along with multiple testing corrections, with inverse variance weighted (IVW) regression serving as the main methodology. Regarding the taxa that were causally linked with dermatological diseases in the forward MR analysis, reverse MR was performed. A series of sensitivity analyses were conducted to test the robustness of the causal estimates. Results The combined results of the five MR methods and sensitivity analysis showed 94 suggestive and five significant causal relationships. In particular, the genus Eubacterium_fissicatena_group increased the risk of developing psoriasis vulgaris (odds ratio [OR] = 1.32, pFDR = 4.36 × 10-3), family Bacteroidaceae (OR = 2.25, pFDR = 4.39 × 10-3), genus Allisonella (OR = 1.42, pFDR = 1.29 × 10-2), and genus Bacteroides (OR = 2.25, pFDR = 1.29 × 10-2) increased the risk of developing acne; and the genus Intestinibacter increased the risk of urticaria (OR = 1.30, pFDR = 9.13 × 10-3). A reverse MR study revealed insufficient evidence for a significant causal relationship. In addition, there was no discernible horizontal pleiotropy or heterogeneity. Conclusion This study provides novel insights into the causality of gut microbiota in dermatological diseases and therapeutic or preventive paradigms for cutaneous conditions.
Collapse
Affiliation(s)
- Yingwei Wang
- Department of Dermatology, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
| | - Tao Yao
- Department of Cardiology, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yunlu Lin
- Department of Cardiology, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
| | - Hongping Ge
- Department of Dermatology, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
| | - Bixin Huang
- Department of Dermatology, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yu Gao
- Department of Dermatology, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
| | - Jianming Wu
- Department of Dermatology, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
4
|
Najafi S, Mortezaee K. Advances in dendritic cell vaccination therapy of cancer. Biomed Pharmacother 2023; 164:114954. [PMID: 37257227 DOI: 10.1016/j.biopha.2023.114954] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Revised: 05/16/2023] [Accepted: 05/27/2023] [Indexed: 06/02/2023] Open
Abstract
Traditionally, vaccines have helped eradication of several infectious diseases and also saved millions of lives in the human history. Those prophylactic vaccines have acted through inducing immune responses against a live attenuated, killed organism or antigenic subunits to protect the recipient against a real infection caused by the pathogenic microorganism. Nevertheless, development of anticancer vaccines as valuable targets in human health has faced challenges and requires further optimizations. Dendritic cells (DCs) are the most potent antigen presenting cells (APCs) that play essential roles in tumor immunotherapies through induction of CD8+ T cell immunity. Accordingly, various strategies have been tested to employ DCs as therapeutic vaccines for exploiting their activity against tumor cells. Application of whole tumor cells or purified/recombinant antigen peptides are the most common approaches for pulsing DCs, which then are injected back into the patients. Although some hopeful results are reported for a number of DC vaccines tested in animal and clinical trials of cancer patients, such approaches are still inefficient and require optimization. Failure of DC vaccination is postulated due to immunosuppressive tumor microenvironment (TME), overexpression of checkpoint proteins, suboptimal avidity of tumor-associated antigen (TAA)-specific T lymphocytes, and lack of appropriate adjuvants. In this review, we have an overview of the current experiments and trials evaluated the anticancer efficacy of DC vaccination as well as focusing on strategies to improve their potential including combination therapy with immune checkpoint inhibitors (ICIs).
Collapse
Affiliation(s)
- Sajad Najafi
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Cellular and Molecular Biology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Keywan Mortezaee
- Department of Anatomy, School of Medicine, Kurdistan University of Medical Sciences, Sanandaj, Iran.
| |
Collapse
|
5
|
Asseri AH, Bakhsh T, Abuzahrah SS, Ali S, Rather IA. The gut dysbiosis-cancer axis: illuminating novel insights and implications for clinical practice. Front Pharmacol 2023; 14:1208044. [PMID: 37361202 PMCID: PMC10288883 DOI: 10.3389/fphar.2023.1208044] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Accepted: 05/31/2023] [Indexed: 06/28/2023] Open
Abstract
The human intestinal microbiota, also known as the gut microbiota, comprises more than 100 trillion organisms, mainly bacteria. This number exceeds the host body cells by a factor of ten. The gastrointestinal tract, which houses 60%-80% of the host's immune cells, is one of the largest immune organs. It maintains systemic immune homeostasis in the face of constant bacterial challenges. The gut microbiota has evolved with the host, and its symbiotic state with the host's gut epithelium is a testament to this co-evolution. However, certain microbial subpopulations may expand during pathological interventions, disrupting the delicate species-level microbial equilibrium and triggering inflammation and tumorigenesis. This review highlights the impact of gut microbiota dysbiosis on the development and progression of certain types of cancers and discusses the potential for developing new therapeutic strategies against cancer by manipulating the gut microbiota. By interacting with the host microbiota, we may be able to enhance the effectiveness of anticancer therapies and open new avenues for improving patient outcomes.
Collapse
Affiliation(s)
- Amer H. Asseri
- Biochemistry Department, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
- Center for Artificial Intelligence in Precision Medicines, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Tahani Bakhsh
- Department of Biology, College of Science, University of Jeddah, Jeddah, Saudi Arabia
| | | | - Sajad Ali
- Department of Biotechnology, Yeungnam University, Gyeongsan, Republic of Korea
| | - Irfan A. Rather
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
- Centre of Excellence in Bionanoscience Research, King Abdulaziz University, Jeddah, Saudi Arabia
| |
Collapse
|
6
|
Srivastava RM, Thounaojam M, Marincola FM, Shanker A. Editorial: Lymphocyte functional crosstalk and regulation, volume II. Front Immunol 2023; 14:1214843. [PMID: 37266417 PMCID: PMC10231030 DOI: 10.3389/fimmu.2023.1214843] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2023] [Accepted: 05/05/2023] [Indexed: 06/03/2023] Open
Affiliation(s)
- Raghvendra M. Srivastava
- Center for Immunotherapy and Precision Immuno-Oncology, Cleveland Clinic, Cleveland, OH, United States
- Lerner Research Institute, Cleveland Clinic, Cleveland, OH, United States
- Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH, United States
| | - Menaka Thounaojam
- Department of Ophthalmology, Augusta University, Augusta, GA, United States
| | | | - Anil Shanker
- Department of Biochemistry, Cancer Biology, Neuroscience and Pharmacology, School of Medicine, Meharry Medical College, Nashville, TN, United States
- Host-Tumor Interactions Research Program, Vanderbilt-Ingram Comprehensive Cancer Center, Vanderbilt University School of Medicine, Nashville, TN, United States
- Vanderbilt Center for Immunobiology, Vanderbilt University School of Medicine, Nashville, TN, United States
- Vanderbilt Institute for Infection, Immunology and Inflammation, Vanderbilt University School of Medicine, Nashville, TN, United States
| |
Collapse
|
7
|
Lepri G, Catalano M, Bellando-Randone S, Pillozzi S, Giommoni E, Giorgione R, Botteri C, Matucci-Cerinic M, Antonuzzo L, Guiducci S. Systemic Sclerosis Association with Malignancy. Clin Rev Allergy Immunol 2022; 63:398-416. [PMID: 36121543 DOI: 10.1007/s12016-022-08930-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/10/2022] [Indexed: 12/17/2022]
Abstract
The association of systemic sclerosis (SSc) and cancer is well known from several decades suggesting common genetic and environmental risk factors involved in the development of both diseases. Immunosuppressive drugs widely used in SSc may increase the risk of cancer occurrence and different SSc clinical and serological features identify patients at major risk to develop malignancy. In this context, among serological features, presence of anti-RNA polymerase III and anti-topoisomerase I autoantibodies seems to increase cancer frequency in SSc patients (particularly lung and breast cancers). Lung fibrosis and a long standing SSc pulmonary involvement have been largely proposed as lung cancer risk factors, and the exposure to cyclophosphamide and an upper gastrointestinal involvement have been traditionally linked to bladder and oesophagus cancers, respectively. Furthermore, immune checkpoint inhibitors used for cancer therapy can induce immune-related adverse events, which are more frequent and severe in patients with pre-existing autoimmune diseases such as SSc. The strong association between SSc and cancer occurrence steers clinicians to carefully survey SSc patients performing periodical malignancy screening. In the present review, the most relevant bilateral relationships between SSc and cancer will be addressed.
Collapse
Affiliation(s)
- Gemma Lepri
- Department of Experimental and Clinical Medicine, University of Florence, and Division of Rheumatology, AOUC & Scleroderma Unit, Florence, Italy.
| | - Martina Catalano
- Medical Oncology Unit, Careggi University Hospital, Florence, Italy
| | - Silvia Bellando-Randone
- Department of Experimental and Clinical Medicine, University of Florence, and Division of Rheumatology, AOUC & Scleroderma Unit, Florence, Italy
| | - Serena Pillozzi
- Medical Oncology Unit, Careggi University Hospital, Florence, Italy
| | - Elisa Giommoni
- Medical Oncology Unit, Careggi University Hospital, Florence, Italy
| | | | - Cristina Botteri
- Medical Oncology Unit, Careggi University Hospital, Florence, Italy
| | - Marco Matucci-Cerinic
- Department of Experimental and Clinical Medicine, University of Florence, and Division of Rheumatology, AOUC & Scleroderma Unit, Florence, Italy.,Unit of Immunology, Rheumatology, Allergy and Rare Diseases (UnIRAR), IRCCS San Raffaele Hospital, Milan, Italy
| | - Lorenzo Antonuzzo
- Medical Oncology, Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Serena Guiducci
- Department of Experimental and Clinical Medicine, University of Florence, and Division of Rheumatology, AOUC & Scleroderma Unit, Florence, Italy
| |
Collapse
|
8
|
A Ferroptosis-Related Gene Signature for Overall Survival Prediction and Immune Infiltration in Lung Squamous Cell Carcinoma. Biosci Rep 2022; 42:231598. [PMID: 35866375 PMCID: PMC9434561 DOI: 10.1042/bsr20212835] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 06/28/2022] [Accepted: 07/20/2022] [Indexed: 02/05/2023] Open
Abstract
Background: Ferroptosis is associated with cancer initiation and progression. However, the molecular mechanism and prognostic value of ferroptosis-related genes in lung squamous cell carcinoma (LUSC) are poorly understood. Methods: The mRNA expression profiles, methylation data, and clinical information of patients with LUSC were downloaded from TCGA and GEO database. Ferroptosis-related differentially expressed genes (DEGs) were identified between cancerous and non-cancerous tissues, and their prognostic value was systemically investigated by bioinformatic analyses. Results: A ferroptosis-related gene signature (ALOX5, TFRC, PHKG2, FADS2, NOX1) was constructed using multivariate Cox regression analysis and represented as a risk score. Overall survival (OS) probability was significantly lower in the high-risk group than in the low-risk group (P<0.001), and receiver operating characteristic curve showed a good predictive capacity (AUC = 0.739). The risk score was an independent prognostic factor for LUSC. A nomogram was constructed to predict the OS probabilities at 1, 3, and 5 years. High-risk score was associated with increased immune infiltration, lower methylation levels, higher immune checkpoint genes expression levels, and better chemotherapy response. Cell adhesion molecules, focal adhesion, and extracellular matrix receptor interaction were the main pathways in the high-risk group. The signature was validated using the TCGA test cohort, entire TCGA cohort, GSE30219, GSE157010, GSE73403, and GSE4573 datasets. The gene disorders in patients with LUSC were validated using real-time PCR and single-cell RNA sequencing analysis. Conclusions: A ferroptosis-related gene signature was constructed to predict OS probability in LUSC. This could facilitate novel therapeutic methods and guide individualized therapy.
Collapse
|
9
|
Chawda C, McMorrow R, Gaspar N, Zambito G, Mezzanotte L. Monitoring Immune Cell Function Through Optical Imaging: a Review Highlighting Transgenic Mouse Models. Mol Imaging Biol 2022; 24:250-263. [PMID: 34735680 PMCID: PMC8983637 DOI: 10.1007/s11307-021-01662-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 09/29/2021] [Accepted: 09/30/2021] [Indexed: 11/17/2022]
Abstract
Transgenic mouse models have facilitated research of human diseases and validation of therapeutic approaches. Inclusion of optical reporter genes (fluorescent or bioluminescent genes) in the targeting vectors used to develop such models makes in vivo imaging of cellular and molecular events possible, from the microscale to the macroscale. In particular, transgenic mouse models expressing optical reporter genes allowed accurately distinguishing immune cell types from trafficking in vivo using intravital microscopy or whole-body optical imaging. Besides lineage tracing and trafficking of different subsets of immune cells, the ability to monitor the function of immune cells is of pivotal importance for investigating the effects of immunotherapies against cancer. Here, we introduce the reader to state-of-the-art approaches to develop transgenics, optical imaging techniques, and several notable examples of transgenic mouse models developed for immunology research by critically highlighting the models that allow the following of immune cell function.
Collapse
Affiliation(s)
- Chintan Chawda
- Department of Radiology and Nuclear Medicine, Erasmus MC, Rotterdam, The Netherlands
- Department of Molecular Genetics, Erasmus MC, Rotterdam, The Netherlands
| | - Roisin McMorrow
- Department of Radiology and Nuclear Medicine, Erasmus MC, Rotterdam, The Netherlands
- Department of Molecular Genetics, Erasmus MC, Rotterdam, The Netherlands
- Percuros B.V, Leiden, The Netherlands
| | - Natasa Gaspar
- Department of Radiology and Nuclear Medicine, Erasmus MC, Rotterdam, The Netherlands
- Department of Molecular Genetics, Erasmus MC, Rotterdam, The Netherlands
- Percuros B.V, Leiden, The Netherlands
| | - Giorgia Zambito
- Department of Radiology and Nuclear Medicine, Erasmus MC, Rotterdam, The Netherlands
- Department of Molecular Genetics, Erasmus MC, Rotterdam, The Netherlands
| | - Laura Mezzanotte
- Department of Radiology and Nuclear Medicine, Erasmus MC, Rotterdam, The Netherlands.
- Department of Molecular Genetics, Erasmus MC, Rotterdam, The Netherlands.
| |
Collapse
|
10
|
Roviello G, Iannone LF, Bersanelli M, Mini E, Catalano M. The gut microbiome and efficacy of cancer immunotherapy. Pharmacol Ther 2022; 231:107973. [PMID: 34453999 DOI: 10.1016/j.pharmthera.2021.107973] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 07/07/2021] [Accepted: 07/27/2021] [Indexed: 12/14/2022]
Abstract
Cancer treatment has been deeply changed by immunotherapy, achieving unprecedented improvement in overall and progression-free survival in several advanced and metastatic cancers. Currently, immune checkpoint inhibitor (ICI) antibodies against cytotoxic T-lymphocyte antigen (CTLA-4) and programmed death/ligand 1 (PD-1/PD-L1) are being tested and approved for different tumors, ranging from melanoma to lung carcinoma. However, only a subgroup of patients can reach treatment benefits and long-term responses, and reliable biomarkers that can accurately predict clinical responses to immunotherapy are still unidentified. In the last decade, accumulating evidence seems to suggest the gut microbiota as one of the modulators that can alter the efficacy and toxicity of immunotherapy drugs (as well as chemotherapeutics), mainly acting through the local and systemic immune system. Herein, we reviewed the highly dynamic and complex microbiome-immune system interface, its bidirectional relationship with cancer immunotherapies, and explored the future possibilities and risks in manipulating the gut microbiome.
Collapse
Affiliation(s)
- Giandomenico Roviello
- Department of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Florence, Viale Pieraccini, 6, 50139 Florence, Italy.
| | | | - Melissa Bersanelli
- Medical Oncology, University Hospital of Parma and Medicine and Surgery Department, University of Parma, Via Gramsci 14, 43126 Parma, Italy
| | - Enrico Mini
- Department of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Florence, Viale Pieraccini, 6, 50139 Florence, Italy
| | - Martina Catalano
- Department of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Florence, Viale Pieraccini, 6, 50139 Florence, Italy
| |
Collapse
|
11
|
Marzhoseyni Z, Shojaie L, Tabatabaei SA, Movahedpour A, Safari M, Esmaeili D, Mahjoubin-Tehran M, Jalili A, Morshedi K, Khan H, Okhravi R, Hamblin MR, Mirzaei H. Streptococcal bacterial components in cancer therapy. Cancer Gene Ther 2022; 29:141-155. [PMID: 33753868 DOI: 10.1038/s41417-021-00308-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Revised: 01/25/2021] [Accepted: 02/05/2021] [Indexed: 02/01/2023]
Abstract
The incidence rate of cancer is steadily increasing all around the world, and there is an urgent need to develop novel and more effective treatment strategies. Recently, bacterial therapy has been investigated as a new approach to target cancer, and is becoming a serious option. Streptococcus strains are among the most common and well-studied virulent bacteria that cause a variety of human infections. Everyone has experienced a sore throat during their lifetime, or has been asymptomatically colonized by streptococci. The ability of Streptococcus bacteria to fight cancer was discovered more than 100 years ago, and over the years has undergone clinical trials, but the mechanism is not yet completely understood. Recently, several animal models and human clinical trials have been reported. Streptococcal strains can have an intrinsic anti-tumor activity, or can activate the host immune system to fight the tumor. Bacteria can selectively accumulate and proliferate in the hypoxic regions of solid tumors. Moreover, the bacteria can be genetically engineered to secrete toxins or enzymes that can specifically attack the tumors.
Collapse
Affiliation(s)
- Zeynab Marzhoseyni
- Department of Microbiology and Immunology, Faculty of Medicine, Kashan University of Medical Sciences, Kashan, Iran
| | - Layla Shojaie
- Research Center for Liver Diseases, Keck School of Medicine, Department of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Seyed Alireza Tabatabaei
- Department of Internal Medicine, School of Medicine, Shahid Beheshti University of Medical Science, Tehran, Iran
| | - Ahmad Movahedpour
- Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mahmood Safari
- Department of Microbiology and Immunology, Faculty of Medicine, Kashan University of Medical Sciences, Kashan, Iran
| | - Davoud Esmaeili
- Department of Microbiology and Applied Microbiology Research Center, Systems Biology and Poisonings Institute and Department of Microbiology, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Maryam Mahjoubin-Tehran
- Department of Medical Biotechnology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amin Jalili
- Department of Medical Biotechnology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Korosh Morshedi
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
| | - Haroon Khan
- Department of Pharmacy, Abdul Wali Khan University Mardan, Mardan, 23200, Pakistan
| | - Ranaa Okhravi
- Department of Medical Sciences, Shahrood Branch, Islamic Azad University, Shahrood, Iran.
| | - Michael R Hamblin
- Laser Research Centre, Faculty of Health Science, University of Johannesburg, Doornfontein, 2028, South Africa.
| | - Hamed Mirzaei
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran.
| |
Collapse
|
12
|
Guru SA, Sumi MP, Mir R, Waza AA, Bhat MA, Zuberi M, Lali P, Saxena A. Ectopic PD-L1 expression in JAK2 (V617F) myeloproliferative neoplasm patients is mediated via increased activation of STAT3 and STAT5. Hum Cell 2020; 33:1099-1111. [PMID: 32430672 DOI: 10.1007/s13577-020-00370-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Accepted: 04/24/2020] [Indexed: 02/08/2023]
Abstract
Escalated PD-L1 expression has been identified during malignant transformation in a number of cancer types and helps cancer cells escape an effective anti-tumor immune response. The mechanisms underlying escalated production of PD-L1 in many cancers, however, are still far from clear. We studied PD-L1, STAT3 and STAT5 mRNA expression using qRT-PCR in 72 BCR/ABL1 negative myeloproliferative neoplasm (MPN) patients (39 polycythemia vera and 33 essential thrombocythemia). Furthermore, phosphorylation status of STAT3 and STAT5 was studied using immunoblotting in the same patients. All MPN patients were first screened for JAK2 (V617F) mutation by tetra-primer ARMS-PCR, followed by quantification of JAK2 (V617F) mutation burden in all V617F positive MPN patients by ASO-PCR. Patients were screened for BCR/ABL1 fusion gene transcripts to rule out Ph positive status. Our findings showed that mRNA levels of PD-L1 and STAT3 were significantly higher in JAK2 (V617F) MPN patients, while as STAT5 was insignificantly upregulated. STAT3 and STAT5 phosphorylation was seen to be higher in JAK2 (V617F) MPN patients compared to the JAK2 (WT) patients. Upregulation of PD-L1, STAT3 and STAT5 was significantly associated with JAK2 (V617F) percentage in MPN patients. PD-L1, STAT3 and STAT5 expression significantly and positively correlated with JAK2 (V617F) allele burden. In addition, significant coexpression of PD-L1 with STAT3 and STAT5 was observed in MPN patients. In summary, JAK2 (V617F) mutation is accompanied by increased PD-L1 expression and this PD-L1 over expression is mediated by JAK2 (V617F) mainly through STAT3, while as STAT5 may play a minor role.
Collapse
Affiliation(s)
- Sameer Ahamd Guru
- Multidisciplinary Research Unit (MRU), Maulana Azad Medical College and Associated Hospitals, Bahadur Shah Zafar Marg, New Delhi, 110002, India
| | - Mamta P Sumi
- Department of Gastroinstestinal Surgery, Govind Balab Pant Postgraduate Institute of Medical Education and Research (GIPMER), New Delhi, India
| | - Rashid Mir
- University of Tabuk, Tabuk, Saudi Arabia
| | - Ajaz Ahmad Waza
- Centre of Research for Development (CORD), University of Kashmir, Srinagar, India
| | - Musadiq Ahmad Bhat
- Institute of Pharmacology and Toxicology-Neuropharmacology, University of Zurich Winterthurerstrasse, Zurich, Switzerland
| | - Mariyam Zuberi
- University of Illinois At Chicago College of Medicine, Chicago, USA
| | - Promod Lali
- Department of Biochemistry, Maulana Azad Medical College and Associated Hospitals, Bahadur Shah Zafar Marg, New Delhi, 110002, India
| | - Alpana Saxena
- Department of Biochemistry, Maulana Azad Medical College and Associated Hospitals, Bahadur Shah Zafar Marg, New Delhi, 110002, India.
| |
Collapse
|
13
|
Li J, Chen L, Su H, Yan L, Gu Z, Chen Z, Zhang A, Zhao F, Zhao Y. The pharmaceutical multi-activity of metallofullerenol invigorates cancer therapy. NANOSCALE 2019; 11:14528-14539. [PMID: 31364651 DOI: 10.1039/c9nr04129j] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Currently, cancer continues to afflict humanity. The direct destruction and killing of tumor cells by surgery, radiation and chemotherapy gives rise to many side effects and compromised efficacy. Encouragingly, the rapid development of nanotechnology offers attractive opportunities to revolutionize the current situation of cancer therapy. Metallofullerenol Gd@C82(OH)22, in contrast to chemotherapeutics that directly kill tumor cells, demonstrates anti-tumor behavior with high efficiency and low toxicity by modulating the tumor microenvironment. Furthermore, Gd@C82(OH)22 has been recently reported to specifically target cancer stem cells. In this review, we give a concise introduction to the development of the fullerene family and then report the anti-tumor activity of Gd@C82(OH)22 based on its unique physicochemical characteristics, followed by a comprehensive summary of the anti-tumor biological mechanisms which target different components of the tumor microenvironment as well as the biodistribution and toxicity of Gd@C82(OH)22. Finally, we describe Gd@C82(OH)22 as a "particulate medicine" to highlight its distinctions from conventional "molecular medicine", with considerable emphasis on the advantages of nanomedicine. The in-depth investigation of Gd@C82(OH)22 undoubtedly provides a constructive reference for the development of other nanomedicines, especially in the fullerene family. The application of nanotechnology in the medical field definitely provides a promising and favorable future for improving the current status of cancer therapy.
Collapse
Affiliation(s)
- Jinxia Li
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences (CAS), Beijing 100049, China.
| | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Concha-Benavente F, Srivastava R, Ferrone S, Ferris RL. Immunological and clinical significance of HLA class I antigen processing machinery component defects in malignant cells. Oral Oncol 2016; 58:52-8. [PMID: 27264839 DOI: 10.1016/j.oraloncology.2016.05.008] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2016] [Accepted: 05/11/2016] [Indexed: 12/31/2022]
Abstract
Experimental as well as clinical studies demonstrate that the immune system plays a major role in controlling generation and progression of tumors. The cancer immunoediting theory supports the notion that tumor cell immunogenicity is dynamically shaped by the immune system, as it eliminates immunogenic tumor cells in the early stage of the disease and then edits their antigenicity. The end result is the generation of a tumor cell population able to escape from immune recognition and elimination by tumor infiltrating lymphocytes. Two major mechanisms, which affect the target cells and the effector phase of the immune response, play a crucial role in the editing process. One is represented by the downregulation of tumor antigen (TA) processing and presentation because of abnormalities in the HLA class I antigen processing machinery (APM). The other one is represented by the anergy of effector immune infiltrates in the tumor microenvironment caused by aberrant inhibitory signals triggered by immune checkpoint receptor (ICR) ligands, such as programmed death ligand-1 (PD-L1). In this review, we will focus on tumor immune escape mechanisms caused by defects in HLA class I APM component expression and/or function in different types of cancer, with emphasis on head and neck cancer (HNC). We will also discuss the immunological implications and clinical relevance of these HLA class I APM abnormalities. Finally, we will describe strategies to counteract defective TA presentation with the expectation that they will enhance tumor recognition and elimination by tumor infiltrating effector T cells.
Collapse
Affiliation(s)
| | | | - Soldano Ferrone
- Department of Surgery, Massachusetts General Hospital, Boston, MA, USA
| | - Robert L Ferris
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA, USA; Department of Otolaryngology, University of Pittsburgh, Pittsburgh, PA, USA; Cancer Immunology Program, University of Pittsburgh Cancer Institute, Pittsburgh, PA, USA.
| |
Collapse
|
15
|
Yu B, Shi L, Zhang BZ, Zhang KE, Peng X, Niu HB, Qu JLE. Obligate anaerobic Salmonella typhimurium strain YB1 treatment on xenograft tumor in immunocompetent mouse model. Oncol Lett 2015; 10:1069-1074. [PMID: 26622627 DOI: 10.3892/ol.2015.3302] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2014] [Accepted: 03/27/2015] [Indexed: 12/13/2022] Open
Abstract
The present authors have previously reported a novel approach to genetically engineer Salmonella typhimurium for the medically important therapeutic strategy of using bacterial agents to target malignant tumors in a breast cancer tumor-bearing nude mouse model. However, studying an immunocompromised mouse model for cancer therapy is insufficient, as certain crucial information about the influence of the immune system may be missing. In the present study, inoculation of the Salmonella strain, YB1, into a colon cancer tumor-bearing immunocompetent mouse model was investigated. The present study determined the tumor targeting efficiency, antitumor potential, the effects of multiple treatments and the systemic toxicity. Intravenous inoculation of YB1 in BALB/c mice exhibited high antitumor effects and also greatly increased the tumor targeting ability and safety compared with the previously-reported nude mouse model. In addition, repeated administration of YB1 further enhanced this effect. Furthermore, no marked toxicity was observed with YB1 treatment, while the VNP20009 and SL7207 strains demonstrated certain adverse effects. The findings of the present study indicate that the YB1 strain is effective and safe in targeting a colon cancer tumor in an immunocompetent mouse model.
Collapse
Affiliation(s)
- Bin Yu
- Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, College of Optoelectronic Engineering, Shenzhen University, Shenzhen, Guangdong 518060, P.R. China ; Department of Biochemistry, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, SAR, P.R. China
| | - Lei Shi
- Department of Biochemistry, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, SAR, P.R. China
| | - Bao-Zhong Zhang
- Department of Biochemistry, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, SAR, P.R. China
| | - K E Zhang
- Department of Microbiology, The University of Hong Kong, Pokfulam, Hong Kong, SAR, P.R. China
| | - Xiao Peng
- Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, College of Optoelectronic Engineering, Shenzhen University, Shenzhen, Guangdong 518060, P.R. China
| | - Han-Ben Niu
- Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, College of Optoelectronic Engineering, Shenzhen University, Shenzhen, Guangdong 518060, P.R. China
| | - Jun-LE Qu
- Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, College of Optoelectronic Engineering, Shenzhen University, Shenzhen, Guangdong 518060, P.R. China
| |
Collapse
|
16
|
Peske JD, Woods AB, Engelhard VH. Control of CD8 T-Cell Infiltration into Tumors by Vasculature and Microenvironment. Adv Cancer Res 2015. [PMID: 26216636 DOI: 10.1016/bs.acr.2015.05.001] [Citation(s) in RCA: 124] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
CD8 T-cells are a critical brake on the initial development of tumors. In established tumors, the presence of CD8 T-cells is correlated with a positive patient prognosis, although immunosuppressive mechanisms limit their effectiveness and they are rarely curative without manipulation. Cancer immunotherapies aim to shift the balance back to dominant antitumor immunity through antibody blockade of immunosuppressive signaling pathways, vaccination, and adoptive transfer of activated or engineered T-cells. These approaches have yielded striking responses in small subsets of patients with solid tumors, most notably those with melanoma. Importantly, the subset of patients who respond to vaccination or immunosuppression blockade therapies are those with CD8 T-cells present in the tumor prior to initiating therapy. While current adoptive cell therapy approaches can be dramatically effective, they require infusion of extremely large numbers of T-cells, but the number that actually infiltrates the tumor is very small. Thus, poor representation of CD8 T-cells in tumors is a fundamental hurdle to successful immunotherapy, over and above the well-established barrier of immunosuppression. In this review, we discuss the factors that determine whether immune cells are present in tumors, with a focus on the representation of cytotoxic CD8 T-cells. We emphasize the critically important role of tumor-associated vasculature as a gateway that enables the active infiltration of both effector and naïve CD8 T-cells that exert antitumor activity. We also discuss strategies to enhance the gateway function and extend the effectiveness of immunotherapies to a broader set of cancer patients.
Collapse
Affiliation(s)
- J David Peske
- Department of Microbiology, Immunology, and Cancer Biology, Carter Immunology Center, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Amber B Woods
- Department of Microbiology, Immunology, and Cancer Biology, Carter Immunology Center, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Victor H Engelhard
- Department of Microbiology, Immunology, and Cancer Biology, Carter Immunology Center, University of Virginia School of Medicine, Charlottesville, Virginia, USA.
| |
Collapse
|
17
|
Baxevanis CN, Anastasopoulou EA, Voutsas IF, Papamichail M, Perez SA. Immune biomarkers: how well do they serve prognosis in human cancers? Expert Rev Mol Diagn 2014; 15:49-59. [PMID: 25345403 DOI: 10.1586/14737159.2015.965684] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
In order to be optimally efficacious, therapeutic cancer vaccines must induce robust tumor-specific CD8(+) cytotoxic T cells, which are responsible for tumor cell lysis. Unlike cytotoxic drugs, which act directly on the tumor, cancer vaccines demonstrate new kinetics involving the generation of specific cellular immune responses, which need to be translated into antitumor responses to delay tumor progression and improve survival. These delayed kinetics of action establish a new concept of benefit in the long term, which implies a slow down in tumor growth rates, than a marked reduction in tumor size. Therefore, there is a significant need to identify intermediate biomarkers so that clinical responses can be evaluated in a timely manner. Therapeutic vaccination as a modality for cancer treatment has received significant attention with multiple clinical trials demonstrating improvements in overall survival. Significant challenges to this modality remain, including increasing vaccine potency and minimizing treatment-related toxicities and identifying prognostic and predictive biomarkers of clinical benefit that may guide to select and optimize the therapeutic strategies for patients most likely to gain benefit.
Collapse
Affiliation(s)
- Constantin N Baxevanis
- Cancer Immunology and Immunotherapy Center, Saint Savas Cancer Hospital, 171 Alexandras avenue, Athens 11522, Greece
| | | | | | | | | |
Collapse
|
18
|
Yaddanapudi K, Mitchell RA, Eaton JW. Cancer vaccines: Looking to the future. Oncoimmunology 2014; 2:e23403. [PMID: 23802081 PMCID: PMC3661166 DOI: 10.4161/onci.23403] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2012] [Revised: 12/20/2012] [Accepted: 12/21/2012] [Indexed: 12/25/2022] Open
Abstract
These are exciting times for the field of cancer immunotherapy. Although the clinical efficacy of monoclonal antibodies has been demonstrated since the early 1990s, the therapeutic profile of other immunotherapeutic approaches-especially vaccines-has not yet been formally clarified. However, the recent success of several immunotherapeutic regimens in cancer patients has boosted the development of this treatment modality. These achievements stemmed from recent scientific advances demonstrating the tolerogenic nature of cancer and the fundamental role of the tumor immune microenvironment in the suppression of antitumor immunity. New immunotherapeutic strategies against cancer attempt to promote protective antitumor immunity while disrupting the immunoregulatory circuits that contribute to tumor tolerance. Cancer vaccines differ from other anticancer immunotherapeutics in that they initiate the dynamic process of activating the immune system so as to successfully re-establish a state of equilibrium between tumor cells and the host. This article reviews recent clinical trials involving several different cancer vaccines and describes some of the most promising immunotherapeutic approaches that harness antitumor T-cell responses. In addition, we describe strategies whereby cancer vaccines can be exploited in combination with other therapeutic approach to overcome-in a synergistic fashion-tumor immunoevasion. Finally, we discuss prospects for the future development of broad spectrum prophylactic anticancer vaccines.
Collapse
Affiliation(s)
- Kavitha Yaddanapudi
- Molecular Targets Group; James Graham Brown Cancer Center; University of Louisville; Louisville, KY USA
| | | | | |
Collapse
|
19
|
Sakode C, Padhi R, Kapoor S, Rallabandi V, Roy P. Multimodal therapy for complete regression of malignant melanoma using constrained nonlinear optimal dynamic inversion. Biomed Signal Process Control 2014. [DOI: 10.1016/j.bspc.2014.04.010] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
20
|
Lu J, Das M, Kanji S, Aggarwal R, Joseph M, Ray A, Shapiro CL, Pompili VJ, Das H. Induction of ATM/ATR pathway combined with Vγ2Vδ2 T cells enhances cytotoxicity of ovarian cancer cells. Biochim Biophys Acta Mol Basis Dis 2014; 1842:1071-9. [PMID: 24726882 DOI: 10.1016/j.bbadis.2014.04.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2013] [Revised: 03/10/2014] [Accepted: 04/04/2014] [Indexed: 11/16/2022]
Abstract
Many ovarian cancer cells express stress-related molecule MICA/B on their surface that is recognized by Vγ2Vδ2 T cells through their NKG2D receptor, which is transmitted to downstream stress-signaling pathway. However, it is yet to be established how Vγ2Vδ2 T cell-mediated recognition of MICA/B signal is transmitted to downstream stress-related molecules. Identifying targeted molecules would be critical to develop a better therapy for ovarian cancer cells. It is well established that ATM/ATR signal transduction pathways, which is modulated by DNA damage, replication stress, and oxidative stress play central role in stress signaling pathway regulating cell cycle checkpoint and apoptosis. We investigated whether ATM/ATR and its down stream molecules affect Vγ2Vδ2 T cell-mediated cytotoxicity. Herein, we show that ATM/ATR pathway is modulated in ovarian cancer cells in the presence of Vγ2Vδ2 T cells. Furthermore, downregulation of ATM pathway resulted downregulation of MICA, and reduced Vγ2Vδ2 T cell-mediated cytotoxicity. Alternately, stimulating ATM pathway enhanced expression of MICA, and sensitized ovarian cancer cells for cytotoxic lysis by Vγ2Vδ2 T cells. We further show that combining currently approved chemotherapeutic drugs, which induced ATM signal transduction, along with Vγ2Vδ2 T cells enhanced cytotoxicity of resistant ovarian cancer cells. These findings indicate that ATM/ATR pathway plays an important role in tumor recognition, and drugs promoting ATM signaling pathway might be considered as a combination therapy together with Vγ2Vδ2 T cells for effectively treating resistant ovarian cancer cells.
Collapse
Affiliation(s)
- Jingwei Lu
- Department of Internal Medicine, Wexner Medical Center at the Ohio State University, Columbus, OH 43210, USA
| | - Manjusri Das
- Department of Internal Medicine, Wexner Medical Center at the Ohio State University, Columbus, OH 43210, USA
| | - Suman Kanji
- Department of Internal Medicine, Wexner Medical Center at the Ohio State University, Columbus, OH 43210, USA
| | - Reeva Aggarwal
- Department of Internal Medicine, Wexner Medical Center at the Ohio State University, Columbus, OH 43210, USA
| | - Matthew Joseph
- Department of Internal Medicine, Wexner Medical Center at the Ohio State University, Columbus, OH 43210, USA
| | - Alo Ray
- Department of Internal Medicine, Wexner Medical Center at the Ohio State University, Columbus, OH 43210, USA
| | - Charles L Shapiro
- Department of Breast Program, Wexner Medical Center at the Ohio State University, Columbus, OH 43210, USA
| | - Vincent J Pompili
- Department of Internal Medicine, Wexner Medical Center at the Ohio State University, Columbus, OH 43210, USA
| | - Hiranmoy Das
- Department of Internal Medicine, Wexner Medical Center at the Ohio State University, Columbus, OH 43210, USA; Innate Immunity, Comprehensive Cancer Center - Arthur G. James Cancer Hospital and Richard J. Solove Research Institute, Wexner Medical Center at The Ohio State University, Columbus, OH 43210, USA.
| |
Collapse
|
21
|
van der Burg SH. Therapeutic vaccines in cancer: moving from immunomonitoring to immunoguiding. Expert Rev Vaccines 2014; 7:1-5. [DOI: 10.1586/14760584.7.1.1] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
22
|
Rönsberg D, Debbab A, Mándi A, Vasylyeva V, Böhler P, Stork B, Engelke L, Hamacher A, Sawadogo R, Diederich M, Wray V, Lin W, Kassack MU, Janiak C, Scheu S, Wesselborg S, Kurtán T, Aly AH, Proksch P. Pro-Apoptotic and Immunostimulatory Tetrahydroxanthone Dimers from the Endophytic Fungus Phomopsis longicolla. J Org Chem 2013; 78:12409-25. [DOI: 10.1021/jo402066b] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Affiliation(s)
- David Rönsberg
- Institut
für Pharmazeutische Biologie und Biotechnologie, Heinrich-Heine-Universität, Universitätsstrasse 1, 40225 Düsseldorf, Germany
| | - Abdessamad Debbab
- Institut
für Pharmazeutische Biologie und Biotechnologie, Heinrich-Heine-Universität, Universitätsstrasse 1, 40225 Düsseldorf, Germany
| | - Attila Mándi
- Department
of Organic Chemistry, University of Debrecen, POB 20, 4010 Debrecen, Hungary
| | - Vera Vasylyeva
- Institut
für Anorganische Chemie und Strukturchemie, Heinrich-Heine-Universität, Universitätsstrasse 1, 40225 Düsseldorf, Germany
| | - Philip Böhler
- Institut
für Molekulare Medizin, Universitätsklinikum Düsseldorf, Heinrich-Heine-Universität, Universitätsstrasse 1, 40225 Düsseldorf, Germany
| | - Björn Stork
- Institut
für Molekulare Medizin, Universitätsklinikum Düsseldorf, Heinrich-Heine-Universität, Universitätsstrasse 1, 40225 Düsseldorf, Germany
| | - Laura Engelke
- Institut
für Pharmazeutische und Medizinische Chemie, Heinrich-Heine-Universität, Universitätsstrasse 1, 40225 Düsseldorf, Germany
| | - Alexandra Hamacher
- Institut
für Pharmazeutische und Medizinische Chemie, Heinrich-Heine-Universität, Universitätsstrasse 1, 40225 Düsseldorf, Germany
| | - Richard Sawadogo
- Laboratory
of Molecular and Cellular Biology of Cancer (LBMCC), Hôpital Kirchberg, 9 rue Edward Steichen, 2540 Luxembourg, Luxembourg
| | - Marc Diederich
- Laboratory
of Molecular and Cellular Biology of Cancer (LBMCC), Hôpital Kirchberg, 9 rue Edward Steichen, 2540 Luxembourg, Luxembourg
| | - Victor Wray
- Helmholtz Centre for Infection Research, Inhoffenstraße 7, 38124 Braunschweig, Germany
| | - WenHan Lin
- National
Research Laboratories of Natural and Biomimetic Drugs, Peking University, Health Science Center, 100083 Beijing, China
| | - Matthias U. Kassack
- Institut
für Pharmazeutische und Medizinische Chemie, Heinrich-Heine-Universität, Universitätsstrasse 1, 40225 Düsseldorf, Germany
| | - Christoph Janiak
- Institut
für Anorganische Chemie und Strukturchemie, Heinrich-Heine-Universität, Universitätsstrasse 1, 40225 Düsseldorf, Germany
| | - Stefanie Scheu
- Institut
für Medizinische Mikrobiologie und Krankenhaushygiene, Heinrich-Heine-Universität, Universitätsstrasse 1, 40225 Düsseldorf, Germany
| | - Sebastian Wesselborg
- Institut
für Molekulare Medizin, Universitätsklinikum Düsseldorf, Heinrich-Heine-Universität, Universitätsstrasse 1, 40225 Düsseldorf, Germany
| | - Tibor Kurtán
- Department
of Organic Chemistry, University of Debrecen, POB 20, 4010 Debrecen, Hungary
| | - Amal H. Aly
- Institut
für Pharmazeutische Biologie und Biotechnologie, Heinrich-Heine-Universität, Universitätsstrasse 1, 40225 Düsseldorf, Germany
| | - Peter Proksch
- Institut
für Pharmazeutische Biologie und Biotechnologie, Heinrich-Heine-Universität, Universitätsstrasse 1, 40225 Düsseldorf, Germany
| |
Collapse
|
23
|
Engineered Salmonella typhimurium expressing E7 fusion protein, derived from human papillomavirus, inhibits tumor growth in cervical tumor-bearing mice. Biotechnol Lett 2013; 36:349-56. [DOI: 10.1007/s10529-013-1370-8] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2013] [Accepted: 09/30/2013] [Indexed: 02/01/2023]
|
24
|
Murtas D, Maric D, De Giorgi V, Reinboth J, Worschech A, Fetsch P, Filie A, Ascierto ML, Bedognetti D, Liu Q, Uccellini L, Chouchane L, Wang E, Marincola FM, Tomei S. IRF-1 responsiveness to IFN-γ predicts different cancer immune phenotypes. Br J Cancer 2013; 109:76-82. [PMID: 23807161 PMCID: PMC3708578 DOI: 10.1038/bjc.2013.335] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Revised: 05/12/2013] [Accepted: 05/23/2013] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Several lines of evidence suggest a dichotomy between immune active and quiescent cancers, with the former associated with a good prognostic phenotype and better responsiveness to immunotherapy. Central to such dichotomy is the master regulator of the acute inflammatory process interferon regulatory factor (IRF)-1. However, it remains unknown whether the responsiveness of IRF-1 to cytokines is able to differentiate cancer immune phenotypes. METHODS IRF-1 activation was measured in 15 melanoma cell lines at basal level and after treatment with IFN-γ, TNF-α and a combination of both. Microarray analysis was used to compare transcriptional patterns between cell lines characterised by high or low IRF-1 activation. RESULTS We observed a strong positive correlation between IRF-1 activation at basal level and after IFN-γ and TNF-α treatment. Microarray demonstrated that three cell lines with low and three with high IRF-1 inducible translocation scores differed in the expression of 597 transcripts. Functional interpretation analysis showed mTOR and Wnt/β-cathenin as the top downregulated pathways in the cell lines with low inducible IRF-1 activation, suggesting that a low IRF-1 inducibility recapitulates a cancer phenotype already described in literature characterised by poor prognosis. CONCLUSION Our findings support the central role of IRF-1 in influencing different tumour phenotypes.
Collapse
Affiliation(s)
- D Murtas
- Infectious Disease and Immunogenetics Section (IDIS), Department of Transfusion Medicine, Clinical Center and trans-NIH Center for Human Immunology (CHI), National Institutes of Health, Bethesda, MD 20892, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Mocellin S, Nitti D. CTLA-4 blockade and the renaissance of cancer immunotherapy. Biochim Biophys Acta Rev Cancer 2013; 1836:187-96. [PMID: 23748107 DOI: 10.1016/j.bbcan.2013.05.003] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2013] [Accepted: 05/27/2013] [Indexed: 12/18/2022]
Abstract
Cytotoxic T-lymphocyte associated antigen 4 (CTLA-4) plays a key role in restraining the adaptive immune response of T-cells towards a variety of antigens including tumor associated antigens (TAAs). The blockade of this immune checkpoint elicits an effective anticancer immune response in a range of preclinical models, suggesting that naturally occurring (or therapeutically induced) TAA specific lymphocytes need to be "unleashed" in order to properly fight against malignant cells. Therefore, investigators have tested this therapeutic hypothesis also in humans: the favorable results obtained with this strategy in patients with advanced cutaneous melanoma are revolutionizing the management of this highly aggressive disease and are fueling new enthusiasm on cancer immunotherapy in general. Here we summarize the biology of CTLA-4, overview the experimental data supporting the rational for targeting CTLA-4 to treat cancer and review the main clinical findings on this novel anticancer approach. Moreover, we critically discuss the current challenges and potential developments of this promising field of cancer immunotherapy.
Collapse
Affiliation(s)
- Simone Mocellin
- Department of Surgery, Oncology and Gastroenterology, University of Padova, Italy.
| | | |
Collapse
|
26
|
Mocellin S, Benna C, Pilati P. Coinhibitory molecules in cancer biology and therapy. Cytokine Growth Factor Rev 2013; 24:147-61. [PMID: 23380546 DOI: 10.1016/j.cytogfr.2013.01.003] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2012] [Accepted: 01/09/2013] [Indexed: 12/31/2022]
Abstract
The adaptive immune response is controlled by checkpoints represented by coinhibitory molecules, which are crucial for maintaining self-tolerance and minimizing collateral tissue damage under physiological conditions. A growing body of preclinical evidence supports the hypothesis that unleashing this immunological break might be therapeutically beneficial in the fight against cancer, as it would elicit an effective antitumor immune response. Remarkably, recent clinical trials have demonstrated that this novel strategy can be highly effective in the treatment of patients with cancer, as shown by the paradigmatic case of ipilimumab (a monoclonal antibody blocking the coinhibitory molecule cytotoxic T lymphocyte associated antigen-4 [CTLA4]) that is opening a new era in the therapeutic approach to a chemoresistant tumor such as cutaneous melanoma. In this review we summarize the biology of coinhibitory molecules, overview the experimental and clinical attempts to interfere with these immune checkpoints to treat cancer and critically discuss the challenges posed by such a promising antitumor modality.
Collapse
Affiliation(s)
- Simone Mocellin
- Department of Surgery, Oncology and Gastroenterology, University of Padova, via Giustiniani 2, 35128 Padova, Italy.
| | | | | |
Collapse
|
27
|
Zhang M, Sukhumalchandra P, Enyenihi AA, St John LS, Hunsucker SA, Mittendorf EA, Sergeeva A, Ruisaard K, Al-Atrache Z, Ropp PA, Jakher H, Rodriguez-Cruz T, Lizee G, Clise-Dwyer K, Lu S, Molldrem JJ, Glish GL, Armistead PM, Alatrash G. A novel HLA-A*0201 restricted peptide derived from cathepsin G is an effective immunotherapeutic target in acute myeloid leukemia. Clin Cancer Res 2012; 19:247-57. [PMID: 23147993 DOI: 10.1158/1078-0432.ccr-12-2753] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Immunotherapy targeting aberrantly expressed leukemia-associated antigens has shown promise in the management of acute myeloid leukemia (AML). However, because of the heterogeneity and clonal evolution that is a feature of myeloid leukemia, targeting single peptide epitopes has had limited success, highlighting the need for novel antigen discovery. In this study, we characterize the role of the myeloid azurophil granule protease cathepsin G (CG) as a novel target for AML immunotherapy. EXPERIMENTAL DESIGN We used Immune Epitope Database and in vitro binding assays to identify immunogenic epitopes derived from CG. Flow cytometry, immunoblotting, and confocal microscopy were used to characterize the expression and processing of CG in AML patient samples, leukemia stem cells, and normal neutrophils. Cytotoxicity assays determined the susceptibility of AML to CG-specific cytotoxic T lymphocytes (CTL). Dextramer staining and cytokine flow cytometry were conducted to characterize the immune response to CG in patients. RESULTS CG was highly expressed and ubiquitinated in AML blasts, and was localized outside granules in compartments that facilitate antigen presentation. We identified five HLA-A*0201 binding nonameric peptides (CG1-CG5) derived from CG, and showed immunogenicity of the highest HLA-A*0201 binding peptide, CG1. We showed killing of primary AML by CG1-CTL, but not normal bone marrow. Blocking HLA-A*0201 abrogated CG1-CTL-mediated cytotoxicity, further confirming HLA-A*0201-dependent killing. Finally, we showed functional CG1-CTLs in peripheral blood from AML patients following allogeneic stem cell transplantation. CONCLUSION CG is aberrantly expressed and processed in AML and is a novel immunotherapeutic target that warrants further development.
Collapse
Affiliation(s)
- Mao Zhang
- Stem Cell Transplantation and Cellular Therapy, Surgical Oncology, and Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Bolhassani A, Zahedifard F. Therapeutic live vaccines as a potential anticancer strategy. Int J Cancer 2012; 131:1733-43. [PMID: 22610886 DOI: 10.1002/ijc.27640] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2012] [Accepted: 05/10/2012] [Indexed: 01/13/2023]
Abstract
The design of efficient cancer treatments is one of the major challenges of medical science. Therapeutic vaccines of cancer have been emerged as an attractive approach for their capacity of breaking the immune tolerance and invoking long-term immune response targeting cancer cells without autoimmunity. An efficient antigen delivery system is the key issue of developing an effective cancer vaccine. In this regard, live vaccination strategies including various live bacterial and viral vectors have attracted a great attention. Several bacterial strains such as Salmonella, Listeria monocytogenes and Lactococcus lactis effectively colonize solid tumors and act as antitumor therapeutics. On the other hand, the use of viruses as vaccine vectors such as Vaccinia, Adenovirus, Herpes simplex virus, Paramyxovirus and Retroviruses utilizes mechanisms that evolved in these microbes for entering cells and capturing the cellular machinery to express viral proteins. Viral/bacterial-vectored vaccines induce systemic T-cell responses including polyfunctional cytokine-secreting CD4+ and CD8+ T-cells. However, there is an urgent need for the development of new safe live vaccine vectors that are capable of enhancing antigen presentation and eliciting potent immune responses without the risk of development of disease in humans. Recently, nonpathogenic parasites including Leishmania tarentolae, Toxoplasma gondii and Trypanosoma cruzi have emerged to be a novel candidate for gene delivery and heterologous genes expression. In this review, recent researches on cancer therapy using genetically modified bacteria and virus are summarized. In addition, live parasite-based vectors will be discussed as a novel anticancer therapeutic approach.
Collapse
Affiliation(s)
- Azam Bolhassani
- Molecular Immunology and Vaccine Research Laboratory, Pasteur Institute of Iran, Tehran, Iran.
| | | |
Collapse
|
29
|
Wang E, Tomei S, Marincola FM. Reflections upon human cancer immune responsiveness to T cell-based therapy. Cancer Immunol Immunother 2012; 61:761-70. [PMID: 22576055 PMCID: PMC3362724 DOI: 10.1007/s00262-012-1274-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2012] [Accepted: 04/24/2012] [Indexed: 01/06/2023]
Abstract
Immune-mediated rejection of human cancer is a relatively rare but well-documented phenomenon. Its rate of occurrence progressively increases from the occasional observation of spontaneous regressions to the high rate of complete remissions observed in response to effective treatments. For two decades, our group has focused its interest in understanding this phenomenon by studying humans following an inductive approach. Sticking to a sequential logic, we dissected the phenomenon by studying to the best of our capability both peripheral and tumor samples and reached the conclusion that immune-mediated cancer rejection is a facet of autoimmunity where the target tissue is the cancer itself. As we are currently defining the strategy to effectively identify the mechanisms leading in individual patients to rejection of their own tumors, we considered useful to summarize the thought process that guided us to our own interpretation of the mechanisms of immune responsiveness.
Collapse
Affiliation(s)
- Ena Wang
- Infectious Disease and Immunogenetics Section (IDIS), Department of Transfusion Medicine, Clinical Center and Trans-NIH Center for Human Immunology (CHI), National Institutes of Health, Bldg 10, Room 1C711, 9000 Rockville Pike, Bethesda, MD 20892 USA
| | - Sara Tomei
- Infectious Disease and Immunogenetics Section (IDIS), Department of Transfusion Medicine, Clinical Center and Trans-NIH Center for Human Immunology (CHI), National Institutes of Health, Bldg 10, Room 1C711, 9000 Rockville Pike, Bethesda, MD 20892 USA
| | - Francesco M. Marincola
- Infectious Disease and Immunogenetics Section (IDIS), Department of Transfusion Medicine, Clinical Center and Trans-NIH Center for Human Immunology (CHI), National Institutes of Health, Bldg 10, Room 1C711, 9000 Rockville Pike, Bethesda, MD 20892 USA
| |
Collapse
|
30
|
Spivey TL, De Giorgi V, Zhao Y, Bedognetti D, Pos Z, Liu Q, Tomei S, Ascierto ML, Uccellini L, Reinboth J, Chouchane L, Stroncek DF, Wang E, Marincola FM. The stable traits of melanoma genetics: an alternate approach to target discovery. BMC Genomics 2012; 13:156. [PMID: 22537248 PMCID: PMC3362771 DOI: 10.1186/1471-2164-13-156] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2011] [Accepted: 04/26/2012] [Indexed: 12/18/2022] Open
Abstract
Background The weight that gene copy number plays in transcription remains controversial; although in specific cases gene expression correlates with copy number, the relationship cannot be inferred at the global level. We hypothesized that genes steadily expressed by 15 melanoma cell lines (CMs) and their parental tissues (TMs) should be critical for oncogenesis and their expression most frequently influenced by their respective copy number. Results Functional interpretation of 3,030 transcripts concordantly expressed (Pearson's correlation coefficient p-value < 0.05) by CMs and TMs confirmed an enrichment of functions crucial to oncogenesis. Among them, 968 were expressed according to the transcriptional efficiency predicted by copy number analysis (Pearson's correlation coefficient p-value < 0.05). We named these genes, "genomic delegates" as they represent at the transcriptional level the genetic footprint of individual cancers. We then tested whether the genes could categorize 112 melanoma metastases. Two divergent phenotypes were observed: one with prevalent expression of cancer testis antigens, enhanced cyclin activity, WNT signaling, and a Th17 immune phenotype (Class A). This phenotype expressed, therefore, transcripts previously associated to more aggressive cancer. The second class (B) prevalently expressed genes associated with melanoma signaling including MITF, melanoma differentiation antigens, and displayed a Th1 immune phenotype associated with better prognosis and likelihood to respond to immunotherapy. An intermediate third class (C) was further identified. The three phenotypes were confirmed by unsupervised principal component analysis. Conclusions This study suggests that clinically relevant phenotypes of melanoma can be retraced to stable oncogenic properties of cancer cells linked to their genetic back bone, and offers a roadmap for uncovering novel targets for tailored anti-cancer therapy.
Collapse
Affiliation(s)
- Tara L Spivey
- Infectious Disease and Immunogenetics Section (IDIS), Department of Transfusion Medicine, Clinical Center and trans-NIH Center for Human Immunology (CHI), National Institutes of Health, Bethesda, MD 20892, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Cassandri F, Tozetti IA, Fernandes CEDS, Almeida FGD, Falcão GR, Scapulatempo IDL, Prata TTM, Padovani CTJ, Alves DB, Ferreira AT, Abdo MAGS. S100, CD68, and MHC class II molecule expression in cervical high- and low-grade HPV-induced lesions. Rev Soc Bras Med Trop 2012; 45:3-8. [PMID: 22370820 DOI: 10.1590/s0037-86822012000100002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2011] [Accepted: 08/10/2011] [Indexed: 11/21/2022] Open
Abstract
INTRODUCTION Some human papillomavirus (HPV) types are involved in malignant processes in the cervical epithelium, with 99% of cases attributed to oncogenic HPV infection. This study aimed to detect S100, CD68, and major histocompatibility complex class II (MHC-II) molecules in cervical uterine epithelial samples in patients with high- and low-grade lesions induced by HPV. METHODS Fifty-eight samples from patients who were confirmed positive or negative for high-risk oncogenic HPV DNA, had histopathological diagnosis of cervical intraepithelial neoplasia (CIN) of grades I, II, or III, or were negative for intraepithelial lesion or malignancy were subjected to immunohistochemistry reaction to S100 protein, CD68, and MHC-II (HLA-DR alpha chain). RESULTS The presence of MHC-II predominated in samples exhibiting histopathological alterations (p < 0.05). S100 detection was more numerous in carcinoma samples (CIN III) (75%). Presence of this protein correlated significantly (p < 0.05) with histopathological findings and viral load. CONCLUSIONS A small expression of CD68 was observed, which may be explained by the observation in our study having been made on random microscopic fields and not on specific areas. The findings, such as the presence of S100 protein and MHC-II expression in samples with histological alterations, could suggest that the immune system fails to control HPV replication at the early stages of infection. Further studies with larger prospective data are necessary to confirm this result.
Collapse
Affiliation(s)
- Fernanda Cassandri
- Programa de Pós-graduação em Doenças Infecciosas e Parasitárias, Faculdade de Medicina, Universidade Federal de Mato Grosso do Sul, Campo Grande, MS
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Raus S, Coin S, Monsurrò V. Adenovirus as a new agent for multiple myeloma therapies: Opportunities and restrictions. THE KOREAN JOURNAL OF HEMATOLOGY 2011; 46:229-38. [PMID: 22259628 PMCID: PMC3259514 DOI: 10.5045/kjh.2011.46.4.229] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/03/2011] [Accepted: 12/19/2011] [Indexed: 01/01/2023]
Abstract
Multiple myeloma is a malignancy of B-cells that is characterized by the clonal expansion and accumulation of malignant plasma cells in the bone marrow. This disease remains incurable, and a median survival of 3-5 years has been reported with the use of current treatments. Viral-based therapies offer promising alternatives or possible integration with current therapeutic regimens. Among several gene therapy vectors and oncolytic agents, adenovirus has emerged as a promising agent, and it is already being used for the treatment of solid tumors in humans. The main concern with the clinical use of this vector has been its high immunogenicity; adenovirus is often able to induce a strong immune response in the host. Furthermore, new limitations in the efficacy of this therapy, intrinsic to the nature of tumor cells, have been recently observed. For example, our group showed a strong antiviral phenotype in vitro and in vivo in a subset of tumors, shedding new insights that may explain the partial failure of clinical trials based on this promising new therapy. In this review, we describe novel therapeutic approaches that implement viral-based treatments in hematological malignancies and address the novelty as well as the possible limitations of these new therapies, especially in the context of the use of adenoviral vectors for treating multiple myeloma.
Collapse
Affiliation(s)
- Svjetlana Raus
- Department of Pathology and Diagnostics, University of Verona Medical School, Verona, Italy
| | | | | |
Collapse
|
33
|
Antitumor activity from antigen-specific CD8 T cells generated in vivo from genetically engineered human hematopoietic stem cells. Proc Natl Acad Sci U S A 2011; 108:E1408-16. [PMID: 22123951 DOI: 10.1073/pnas.1115050108] [Citation(s) in RCA: 88] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
The goal of cancer immunotherapy is the generation of an effective, stable, and self-renewing antitumor T-cell population. One such approach involves the use of high-affinity cancer-specific T-cell receptors in gene-therapy protocols. Here, we present the generation of functional tumor-specific human T cells in vivo from genetically modified human hematopoietic stem cells (hHSC) using a human/mouse chimera model. Transduced hHSC expressing an HLA-A*0201-restricted melanoma-specific T-cell receptor were introduced into humanized mice, resulting in the generation of a sizeable melanoma-specific naïve CD8(+) T-cell population. Following tumor challenge, these transgenic CD8(+) T cells, in the absence of additional manipulation, limited and cleared human melanoma tumors in vivo. Furthermore, the genetically enhanced T cells underwent proper thymic selection, because we did not observe any responses against non-HLA-matched tumors, and no killing of any kind occurred in the absence of a human thymus. Finally, the transduced hHSC established long-term bone marrow engraftment. These studies present a potential therapeutic approach and an important tool to understand better and to optimize the human immune response to melanoma and, potentially, to other types of cancer.
Collapse
|
34
|
Leibovici J, Itzhaki O, Huszar M, Sinai J. The tumor microenvironment: part 1. Immunotherapy 2011; 3:1367-84. [DOI: 10.2217/imt.11.111] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
For years the mutated, highly proliferating neoplastic cells were presented as the only important agent in tumors; however, during the last 3–4 decades it has become clear that the microenvironment of the cancer cells plays a determinative role in the malignant evolution of neoplasia. Cancers are in fact heterogeneous entities containing, in addition to the neoplastic cell component, cells derived of multiple lineages (fibroblasts, endothelial cells lining blood and lymphatic vessels, pericytes, adipocytes and immune system cells belonging to both innate and adaptive responses), as well as the extracellular matrix, with a large variety of soluble molecules of biological importance, constituting a complex organ-like structure. The tumor microenvironment consists in a tissue that may have a predictive significance for tumor behavior and response to therapy.
Collapse
Affiliation(s)
| | - Orit Itzhaki
- Department of Pathology, Sackler Faculty of Medicine, Tel-Aviv University, 69978, Tel-Aviv, Israel
| | - Monica Huszar
- Department of Pathology, Sackler Faculty of Medicine, Tel-Aviv University, 69978, Tel-Aviv, Israel
| | - Judith Sinai
- Department of Pathology, Sackler Faculty of Medicine, Tel-Aviv University, 69978, Tel-Aviv, Israel
| |
Collapse
|
35
|
Leibovici J, Itzhaki O, Huszar M, Sinai J. Targeting the tumor microenvironment by immunotherapy: part 2. Immunotherapy 2011; 3:1385-408. [DOI: 10.2217/imt.11.112] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Cancer therapy was traditionally centered on the neoplastic cells. This included mainly surgery, radiation, and chemotherapy, in some cases hormone therapy and to a lesser extent immunotherapy – all traditionally targeted to the highly proliferating mutated tumor cells. In view of our present understanding of the powerfull influence of the tumor microenvironment (TME) on cancer behavior and response – and lack of response – to treatment, this previously ignored constituent of cancer now has to be considered as an important, even indispensable target for therapy. The TME may be targeted both to its immune and to its nonimmune components. The various immune evasion elements of the TME should be targeted as well.
Collapse
Affiliation(s)
| | - Orit Itzhaki
- Department of Pathology, Sackler Faculty of Medicine, Tel-Aviv University, 69978 Tel-Aviv, Israel
| | - Monica Huszar
- Department of Pathology, Sackler Faculty of Medicine, Tel-Aviv University, 69978 Tel-Aviv, Israel
| | - Judith Sinai
- Department of Pathology, Sackler Faculty of Medicine, Tel-Aviv University, 69978 Tel-Aviv, Israel
| |
Collapse
|
36
|
Ascierto ML, Giorgi VD, Liu Q, Bedognetti D, Spivey TL, Murtas D, Uccellini L, Ayotte BD, Stroncek DF, Chouchane L, Manjili MH, Wang E, Marincola FM. An immunologic portrait of cancer. J Transl Med 2011; 9:146. [PMID: 21875439 PMCID: PMC3175185 DOI: 10.1186/1479-5876-9-146] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2011] [Accepted: 08/29/2011] [Indexed: 12/31/2022] Open
Abstract
The advent of high-throughput technology challenges the traditional histopathological classification of cancer, and proposes new taxonomies derived from global transcriptional patterns. Although most of these molecular re-classifications did not endure the test of time, they provided bulk of new information that can reframe our understanding of human cancer biology. Here, we focus on an immunologic interpretation of cancer that segregates oncogenic processes independent from their tissue derivation into at least two categories of which one bears the footprints of immune activation. Several observations describe a cancer phenotype where the expression of interferon stimulated genes and immune effector mechanisms reflect patterns commonly observed during the inflammatory response against pathogens, which leads to elimination of infected cells. As these signatures are observed in growing cancers, they are not sufficient to entirely clear the organism of neoplastic cells but they sustain, as in chronic infections, a self-perpetuating inflammatory process. Yet, several studies determined an association between this inflammatory status and a favorable natural history of the disease or a better responsiveness to cancer immune therapy. Moreover, these signatures overlap with those observed during immune-mediated cancer rejection and, more broadly, immune-mediated tissue-specific destruction in other immune pathologies. Thus, a discussion concerning this cancer phenotype is warranted as it remains unknown why it occurs in immune competent hosts. It also remains uncertain whether a genetically determined response of the host to its own cancer, the genetic makeup of the neoplastic process or a combination of both drives the inflammatory process. Here we reflect on commonalities and discrepancies among studies and on the genetic or somatic conditions that may cause this schism in cancer behavior.
Collapse
Affiliation(s)
- Maria Libera Ascierto
- Infectious Disease and Immunogenetics Section (IDIS), Department of Transfusion Medicine, Clinical Center and Trans-NIH Center for Human Immunology (CHI), National Institutes of Health, Bethesda, Maryland, 20892, USA
- Department of Internal Medicine, University of Genoa, Italy
- Center of Excellence for Biomedical Research (CEBR), Genoa, Italy
| | - Valeria De Giorgi
- Infectious Disease and Immunogenetics Section (IDIS), Department of Transfusion Medicine, Clinical Center and Trans-NIH Center for Human Immunology (CHI), National Institutes of Health, Bethesda, Maryland, 20892, USA
| | - Qiuzhen Liu
- Infectious Disease and Immunogenetics Section (IDIS), Department of Transfusion Medicine, Clinical Center and Trans-NIH Center for Human Immunology (CHI), National Institutes of Health, Bethesda, Maryland, 20892, USA
| | - Davide Bedognetti
- Infectious Disease and Immunogenetics Section (IDIS), Department of Transfusion Medicine, Clinical Center and Trans-NIH Center for Human Immunology (CHI), National Institutes of Health, Bethesda, Maryland, 20892, USA
- Center of Excellence for Biomedical Research (CEBR), Genoa, Italy
- Department of Oncology, Biology and Genetics and National Cancer Research Institute of Genoa, Italy
| | - Tara L Spivey
- Infectious Disease and Immunogenetics Section (IDIS), Department of Transfusion Medicine, Clinical Center and Trans-NIH Center for Human Immunology (CHI), National Institutes of Health, Bethesda, Maryland, 20892, USA
| | - Daniela Murtas
- Infectious Disease and Immunogenetics Section (IDIS), Department of Transfusion Medicine, Clinical Center and Trans-NIH Center for Human Immunology (CHI), National Institutes of Health, Bethesda, Maryland, 20892, USA
| | - Lorenzo Uccellini
- Infectious Disease and Immunogenetics Section (IDIS), Department of Transfusion Medicine, Clinical Center and Trans-NIH Center for Human Immunology (CHI), National Institutes of Health, Bethesda, Maryland, 20892, USA
| | - Ben D Ayotte
- Department of Biology, Northern Michigan University, Marquette, MI 49855,USA
| | - David F Stroncek
- Infectious Disease and Immunogenetics Section (IDIS), Department of Transfusion Medicine, Clinical Center and Trans-NIH Center for Human Immunology (CHI), National Institutes of Health, Bethesda, Maryland, 20892, USA
| | - Lotfi Chouchane
- Weill Cornell Medical College in Qatar, Education City, Doha Qatar Box 24144
| | - Masoud H Manjili
- Department of Microbiology & Immunology, Virginia Commonwealth University Massey Cancer Center, Richmond, VA 23298, USA
| | - Ena Wang
- Infectious Disease and Immunogenetics Section (IDIS), Department of Transfusion Medicine, Clinical Center and Trans-NIH Center for Human Immunology (CHI), National Institutes of Health, Bethesda, Maryland, 20892, USA
| | - Francesco M Marincola
- Infectious Disease and Immunogenetics Section (IDIS), Department of Transfusion Medicine, Clinical Center and Trans-NIH Center for Human Immunology (CHI), National Institutes of Health, Bethesda, Maryland, 20892, USA
| |
Collapse
|
37
|
Russ AJ, Xu K, Wentworth L, Alam S, Meyers JV, Macklin MD, Rakhmilevich AL, Rajamanickam V, Suresh M, Cho CS. Melanoma-induced suppression of tumor antigen-specific T cell expansion is comparable to suppression of global T cell expansion. Cell Immunol 2011; 271:104-9. [PMID: 21741629 DOI: 10.1016/j.cellimm.2011.06.011] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2011] [Revised: 06/02/2011] [Accepted: 06/10/2011] [Indexed: 01/28/2023]
Abstract
We have observed that in vivo interaction between melanoma and resting T cells promotes suppression of antigen-driven proliferative T cell expansion. We hypothesized that this suppression would affect tumor antigen-specific T cell populations more potently than tumor-unrelated T cell populations. A B16F10 cell line was stably transfected to express low levels of the lymphocytic choriomeningitis virus (LCMV) glycoprotein GP33 (B16GP33). Mice bearing B16F10 or B16GP33 tumors were infected with LCMV, and proliferative expansion of LCMV epitope-specific T cell populations was quantified. In vitro and in vivo assays confirmed low levels of antigenic GP33 expression by B16GP33 tumors. Suppressed expansion of GP33-specific T cells was equivalent between mice bearing B16F10 and B16GP33 tumors. These observations suggest that the ability of growing melanoma tumors to impair antigen-driven proliferative expansion of activated T cells is global and not antigen-specific, and provide further insight into the influence of cancer on activated T cell homeostasis.
Collapse
Affiliation(s)
- Andrew J Russ
- Section of Surgical Oncology, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792-7375, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Gilbert AE, Karagiannis P, Dodev T, Koers A, Lacy K, Josephs DH, Takhar P, Geh JLC, Healy C, Harries M, Acland KM, Rudman SM, Beavil RL, Blower PJ, Beavil AJ, Gould HJ, Spicer J, Nestle FO, Karagiannis SN. Monitoring the systemic human memory B cell compartment of melanoma patients for anti-tumor IgG antibodies. PLoS One 2011; 6:e19330. [PMID: 21559411 PMCID: PMC3084832 DOI: 10.1371/journal.pone.0019330] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2011] [Accepted: 03/26/2011] [Indexed: 11/24/2022] Open
Abstract
Melanoma, a potentially lethal skin cancer, is widely thought to be immunogenic
in nature. While there has been much focus on T cell-mediated immune responses,
limited knowledge exists on the role of mature B cells. We describe an approach,
including a cell-based ELISA, to evaluate mature IgG antibody responses to
melanoma from human peripheral blood B cells. We observed a significant increase
in antibody responses from melanoma patients (n = 10) to
primary and metastatic melanoma cells compared to healthy volunteers
(n = 10) (P<0.0001). Interestingly, we
detected a significant reduction in antibody responses to melanoma with
advancing disease stage in our patient cohort (n = 21)
(P<0.0001). Overall, 28% of
melanoma patient-derived B cell cultures (n = 1,800)
compared to 2% of cultures from healthy controls
(n = 600) produced antibodies that recognized melanoma
cells. Lastly, a patient-derived melanoma-specific monoclonal antibody was
selected for further study. This antibody effectively killed melanoma cells
in vitro via antibody-mediated cellular cytotoxicity. These
data demonstrate the presence of a mature systemic B cell response in melanoma
patients, which is reduced with disease progression, adding to previous reports
of tumor-reactive antibodies in patient sera, and suggesting the merit of future
work to elucidate the clinical relevance of activating humoral immune responses
to cancer.
Collapse
Affiliation(s)
- Amy E. Gilbert
- Cutaneous Medicine and Immunotherapy Unit, Division of Genetics and
Molecular Medicine, NIHR Biomedical Research Centre at Guy’s and St.
Thomas’s Hospitals and King’s College London, King’s College
London School of Medicine, St. John’s Institute of Dermatology,
Guy’s Hospital, King’s College London, London, United
Kingdom
| | - Panagiotis Karagiannis
- Cutaneous Medicine and Immunotherapy Unit, Division of Genetics and
Molecular Medicine, NIHR Biomedical Research Centre at Guy’s and St.
Thomas’s Hospitals and King’s College London, King’s College
London School of Medicine, St. John’s Institute of Dermatology,
Guy’s Hospital, King’s College London, London, United
Kingdom
| | - Tihomir Dodev
- Randall Division of Cell and Molecular Biophysics and Division of Asthma,
Allergy and Lung Biology, MRC and Asthma UK Centre for Allergic Mechanisms of
Asthma, King's College London, London, United Kingdom
| | - Alexander Koers
- Division of Imaging Sciences, King’s College London School of
Medicine, Rayne Institute, St. Thomas's Hospital, King’s College
London, London, United Kingdom
| | - Katie Lacy
- Cutaneous Medicine and Immunotherapy Unit, Division of Genetics and
Molecular Medicine, NIHR Biomedical Research Centre at Guy’s and St.
Thomas’s Hospitals and King’s College London, King’s College
London School of Medicine, St. John’s Institute of Dermatology,
Guy’s Hospital, King’s College London, London, United
Kingdom
| | - Debra H. Josephs
- Cutaneous Medicine and Immunotherapy Unit, Division of Genetics and
Molecular Medicine, NIHR Biomedical Research Centre at Guy’s and St.
Thomas’s Hospitals and King’s College London, King’s College
London School of Medicine, St. John’s Institute of Dermatology,
Guy’s Hospital, King’s College London, London, United
Kingdom
| | - Pooja Takhar
- Randall Division of Cell and Molecular Biophysics and Division of Asthma,
Allergy and Lung Biology, MRC and Asthma UK Centre for Allergic Mechanisms of
Asthma, King's College London, London, United Kingdom
| | - Jenny L. C. Geh
- Skin Tumour Unit, Guy's and St. Thomas's NHS Trust, St.
John’s Institute of Dermatology, Guy’s Hospital, London, United
Kingdom
| | - Ciaran Healy
- Skin Tumour Unit, Guy's and St. Thomas's NHS Trust, St.
John’s Institute of Dermatology, Guy’s Hospital, London, United
Kingdom
| | - Mark Harries
- Clinical Oncology, Guy’s and St. Thomas’s NHS Foundation
Trust, London, United Kingdom
| | - Katharine M. Acland
- Skin Tumour Unit, Guy's and St. Thomas's NHS Trust, St.
John’s Institute of Dermatology, Guy’s Hospital, London, United
Kingdom
| | - Sarah M. Rudman
- Division of Cancer Studies, Department of Academic Oncology, King’s
College London, Guy's Hospital, London, United Kingdom
| | - Rebecca L. Beavil
- Randall Division of Cell and Molecular Biophysics and Division of Asthma,
Allergy and Lung Biology, MRC and Asthma UK Centre for Allergic Mechanisms of
Asthma, King's College London, London, United Kingdom
| | - Philip J. Blower
- Division of Imaging Sciences, King’s College London School of
Medicine, Rayne Institute, St. Thomas's Hospital, King’s College
London, London, United Kingdom
| | - Andrew J. Beavil
- Randall Division of Cell and Molecular Biophysics and Division of Asthma,
Allergy and Lung Biology, MRC and Asthma UK Centre for Allergic Mechanisms of
Asthma, King's College London, London, United Kingdom
| | - Hannah J. Gould
- Randall Division of Cell and Molecular Biophysics and Division of Asthma,
Allergy and Lung Biology, MRC and Asthma UK Centre for Allergic Mechanisms of
Asthma, King's College London, London, United Kingdom
| | - James Spicer
- Division of Cancer Studies, Department of Academic Oncology, King’s
College London, Guy's Hospital, London, United Kingdom
| | - Frank O. Nestle
- Cutaneous Medicine and Immunotherapy Unit, Division of Genetics and
Molecular Medicine, NIHR Biomedical Research Centre at Guy’s and St.
Thomas’s Hospitals and King’s College London, King’s College
London School of Medicine, St. John’s Institute of Dermatology,
Guy’s Hospital, King’s College London, London, United
Kingdom
- * E-mail: (SNK); (FON)
| | - Sophia N. Karagiannis
- Cutaneous Medicine and Immunotherapy Unit, Division of Genetics and
Molecular Medicine, NIHR Biomedical Research Centre at Guy’s and St.
Thomas’s Hospitals and King’s College London, King’s College
London School of Medicine, St. John’s Institute of Dermatology,
Guy’s Hospital, King’s College London, London, United
Kingdom
- * E-mail: (SNK); (FON)
| |
Collapse
|
39
|
Russ AJ, Wentworth L, Xu K, Rakhmilevich A, Seroogy CM, Sondel PM, Suresh M, Cho CS. Suppression of T-cell expansion by melanoma is exerted on resting cells. Ann Surg Oncol 2011; 18:3848-57. [PMID: 21465311 DOI: 10.1245/s10434-011-1667-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2011] [Indexed: 01/16/2023]
Abstract
BACKGROUND Immunotherapeutic cancer protocols often rely on the ability to promote proliferative expansion of tumor-specific T-cell, but the influence of cancer on in vivo T-cell expansion remains largely undefined. METHODS The ability of control and B16F10 melanoma-bearing C57BL/6 mice to expand lymphocytic choriomeningitis virus antigen-specific T-cell populations in response to acute viral infection was compared by using flow cytometric assays of splenocytes. RESULTS The ability to expand virus-specific CD8+ and CD4+ T-cells was globally and markedly suppressed in tumor-bearing mice. Expanded cytotoxic T lymphocytes (CTLs) retained in vivo and in vitro functionality, suggesting that melanoma growth did not induce T-cell anergy. The magnitude of suppressed proliferative expansion was proportional to the extent of tumor burden. Melanoma-induced suppression of CTL expansion was correlated with upregulated apoptotic activity and hampered the induction of memory precursor effector cells. Adoptive transfer of resting LCMV antigen-specific T-cells before or after tumor establishment demonstrated that a critical period of in vivo exposure of resting T-cells to growing melanoma was responsible for the induction of suppressed expansion. This suppression was durable; surgical resection of melanoma after in vivo exposure to resting T-cells but before antigenic stimulation did not restore full expansion. CONCLUSIONS These data suggest that growing melanoma tumors exert a global, antigen-independent influence on resting T-cells that fundamentally reprograms their ability to undergo proliferative expansion in response to subsequent antigenic stimulation. This finding may have direct implications for T-cell-based immunotherapeutic strategies.
Collapse
Affiliation(s)
- Andrew J Russ
- Section of Surgical Oncology, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | | | | | | | | | | | | | | |
Collapse
|
40
|
|
41
|
Ataera H, Hyde E, Price KM, Stoitzner P, Ronchese F. Murine melanoma-infiltrating dendritic cells are defective in antigen presenting function regardless of the presence of CD4CD25 regulatory T cells. PLoS One 2011; 6:e17515. [PMID: 21390236 PMCID: PMC3048402 DOI: 10.1371/journal.pone.0017515] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2010] [Accepted: 02/04/2011] [Indexed: 01/08/2023] Open
Abstract
Tumor-infiltrating dendritic cells are often ineffective at presenting tumor-derived antigen in vivo, a defect usually ascribed to the suppressive tumor environment. We investigated the effects of depleting CD4+CD25+ “natural” regulatory T cells (Treg) on the frequency, phenotype and function of total dendritic cell populations in B16.OVA tumors and in tumor-draining lymph nodes. Intraperitoneal injection of the anti-CD25 monoclonal antibody PC61 reduced Treg frequency in blood and tumors, but did not affect the frequency of tumor-infiltrating dendritic cells, or their expression of CD40, CD86 and MHCII. Tumor-infiltrating dendritic cells from PC61-treated or untreated mice induced the proliferation of allogeneic T cells in vitro, but could not induce proliferation of OVA-specific OTI and OTII T cells unless specific peptide antigen was added in culture. Some proliferation of naïve, OVA-specific OTI T cells, but not OTII T cells, was observed in the tumor-draining LN of mice carrying B16.OVA tumors, however, this was not improved by PC61 treatment. Experiments using RAG1−/− hosts adoptively transferred with OTI and CD25-depleted OTII cells also failed to show improved OTI and OTII T cell proliferation in vivo compared to C57BL/6 hosts. We conclude that the defective presentation of B16.OVA tumor antigen by tumor-infiltrating dendritic cells and in the tumor-draining lymph node is not due to the presence of “natural” CD4+CD25+ Treg.
Collapse
Affiliation(s)
- Haley Ataera
- Malaghan Institute of Medical Research, Wellington, New Zealand
| | - Evelyn Hyde
- Malaghan Institute of Medical Research, Wellington, New Zealand
| | - Kylie M. Price
- Malaghan Institute of Medical Research, Wellington, New Zealand
| | | | - Franca Ronchese
- Malaghan Institute of Medical Research, Wellington, New Zealand
- * E-mail:
| |
Collapse
|
42
|
Kotlan B, Stroncek DF, Marincola FM. Intravenous immunoglobulin-based immunotherapy: an arsenal of possibilities for patients and science. Immunotherapy 2011; 1:995-1015. [PMID: 20635915 DOI: 10.2217/imt.09.67] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The use of intravenous immunoglobulin (IVIG) concentrated from pooled healthy donors' plasma has gained increasing popularity. IVIG therapy has become important as a replacement therapy in primary and acquired humoral immunodeficiencies, and it has been extended to autoimmune, neurodegenerative and inflammatory conditions and transplantation therapy. Recurrent pregnancy failure and cancer are rather new platforms, where IVIG has shown its beneficial effects. This manuscript is focused on these two off-labelled usages. The immunomodulatory mechanisms of IVIG therapy appear as a coordinated orchestration of different functions, resulting in a synergistic effect. Treatment monitoring and detailed molecular analyses reveal how such treatments may interfere with disease pathogenesis. These finding may foster the development of novel therapeutic and/or preventive strategies. Studying this field with bidirectional bench-to-bedside and bedside-to-bench approaches fit well into 'the two-way road' paradigm of translational medicine.
Collapse
Affiliation(s)
- Beatrix Kotlan
- Center of Surgical & Molecular Tumorpathology National Institute of Oncology, Rath Gy street 7-9, Budapest 1122, Hungary.
| | | | | |
Collapse
|
43
|
Vendrell A, Gravisaco MJ, Pasetti MF, Croci M, Colombo L, Rodríguez C, Mongini C, Waldner CI. A novel Salmonella Typhi-based immunotherapy promotes tumor killing via an antitumor Th1-type cellular immune response and neutrophil activation in a mouse model of breast cancer. Vaccine 2011; 29:728-36. [DOI: 10.1016/j.vaccine.2010.11.017] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2010] [Revised: 11/08/2010] [Accepted: 11/09/2010] [Indexed: 01/21/2023]
|
44
|
A CpG-loaded tumor cell vaccine induces antitumor CD4+ T cells that are effective in adoptive therapy for large and established tumors. Blood 2010; 117:118-27. [PMID: 20876455 DOI: 10.1182/blood-2010-06-288456] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
We designed a whole tumor cell vaccine by "loading" lymphoma tumor cells with CG-enriched oligodeoxynucleotide (CpG), a ligand for the Toll-like receptor 9 (TLR9). CpG-loaded tumor cells were phagocytosed, delivering both tumor antigen(s) and the immunostimulatory CpG molecule to antigen-presenting cells (APCs). These APCs then expressed increased levels of costimulatory molecules and induced T-cell immunity. TLR9 was required in the APCs but not in the CpG-loaded tumor cell. We demonstrate that T cells induced by this vaccine are effective in adoptive cellular therapy for lymphoma. T cells from vaccinated mice transferred into irradiated, syngeneic recipients protected against subsequent lymphoma challenge and, remarkably, led to regression of large and established tumors. This therapeutic effect could be transferred by CD4(+) but not by CD8(+) T cells. A CpG-loaded whole-cell vaccination is practical and has strong potential for translation to the clinical setting. It is currently being tested in a clinical trial of adoptive immunotherapy for mantle-cell lymphoma.
Collapse
|
45
|
Schroten C, Kraaij R, Veldhoven JL, Berrevoets CA, den Bakker MA, Ma Q, Sadelain M, Bangma CH, Willemsen RA, Debets R. T cell activation upon exposure to patient-derived tumor tissue: A functional assay to select patients for adoptive T cell therapy. J Immunol Methods 2010; 359:11-20. [DOI: 10.1016/j.jim.2010.04.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2009] [Revised: 04/26/2010] [Accepted: 04/29/2010] [Indexed: 10/19/2022]
|
46
|
Bedognetti D, Wang E, Sertoli MR, Marincola FM. Gene-expression profiling in vaccine therapy and immunotherapy for cancer. Expert Rev Vaccines 2010; 9:555-65. [PMID: 20518712 PMCID: PMC3411321 DOI: 10.1586/erv.10.55] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The identification of tumor antigens recognized by T cells led to the design of therapeutic strategies aimed at eliciting adaptive immune responses. The last decade of experience has shown that, although active immunization can induce enhancement of anticancer T-cell precursors (easily detectable in standard assays), most often they are unable to induce tumor regression and, consequently, have scarcely any impact on overall survival. Moreover, in the few occasions when tumor rejection occurs, the mechanisms determining this phenomenon remain poorly understood, and data derived from in vivo human observations are rare. The advent of high-throughput gene-expression analysis (microarrays) has cast new light on unrecognized mechanisms that are now deemed to be central for the development of efficient immune-mediated tumor rejection. The aim of this article is to review the data on the molecular signature associated with this process. We believe that the description of how the mechanism of immune-mediated tissue destruction occurs would contribute to our understanding of why it happens, thereby allowing us to develop more effective immune therapeutic strategies.
Collapse
Affiliation(s)
- Davide Bedognetti
- Infectious Disease and Immunogenetics Section (IDIS), Department of Transfusion Medicine, Clinical Center, and Trans-NIH Center for Human Immunology (CHI), National Institutes of Health, Bethesda, MD 20892, USA
- S.C. Oncologia Medica B, Department of Medical Oncology, National Cancer Research Institute, Genoa, Italy
- Department of Internal Medicine, University of Genoa, Genoa, Italy
- Department of Oncology, Biology and Genetics, University of Genoa, Genoa, Italy
| | - Ena Wang
- Infectious Disease and Immunogenetics Section (IDIS), Department of Transfusion Medicine, Clinical Center, and Trans-NIH Center for Human Immunology (CHI), National Institutes of Health, Bethesda, MD 20892, USA
| | - Mario Roberto Sertoli
- S.C. Oncologia Medica B, Department of Medical Oncology, National Cancer Research Institute, Genoa, Italy
- Department of Oncology, Biology and Genetics, University of Genoa, Genoa, Italy
| | - Francesco M Marincola
- Infectious Disease and Immunogenetics Section (IDIS), Department of Transfusion Medicine, Clinical Center, and Trans-NIH Center for Human Immunology (CHI), National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
47
|
Monsurrò V, Beghelli S, Wang R, Barbi S, Coin S, Di Pasquale G, Bersani S, Castellucci M, Sorio C, Eleuteri S, Worschech A, Chiorini JA, Pederzoli P, Alter H, Marincola FM, Scarpa A. Anti-viral state segregates two molecular phenotypes of pancreatic adenocarcinoma: potential relevance for adenoviral gene therapy. J Transl Med 2010; 8:10. [PMID: 20113473 PMCID: PMC2845551 DOI: 10.1186/1479-5876-8-10] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2009] [Accepted: 01/29/2010] [Indexed: 02/07/2023] Open
Abstract
Background Pancreatic ductal adenocarcinoma (PDAC) remains a leading cause of cancer mortality for which novel gene therapy approaches relying on tumor-tropic adenoviruses are being tested. Methods We obtained the global transcriptional profiling of primary PDAC using RNA from eight xenografted primary PDAC, three primary PDAC bulk tissues, three chronic pancreatitis and three normal pancreatic tissues. The Affymetrix GeneChip HG-U133A was used. The results of the expression profiles were validated applying immunohistochemical and western blot analysis on a set of 34 primary PDAC and 10 established PDAC cell lines. Permissivity to viral vectors used for gene therapy, Adenovirus 5 and Adeno-Associated Viruses 5 and 6, was assessed on PDAC cell lines. Results The analysis of the expression profiles allowed the identification of two clearly distinguishable phenotypes according to the expression of interferon-stimulated genes. The two phenotypes could be readily recognized by immunohistochemical detection of the Myxovirus-resistance A protein, whose expression reflects the activation of interferon dependent pathways. The two molecular phenotypes discovered in primary carcinomas were also observed among established pancreatic adenocarcinoma cell lines, suggesting that these phenotypes are an intrinsic characteristic of cancer cells independent of their interaction with the host's microenvironment. The two pancreatic cancer phenotypes are characterized by different permissivity to viral vectors used for gene therapy, as cell lines expressing interferon stimulated genes resisted to Adenovirus 5 mediated lysis in vitro. Similar results were observed when cells were transduced with Adeno-Associated Viruses 5 and 6. Conclusion Our study identified two molecular phenotypes of pancreatic cancer, characterized by a differential expression of interferon-stimulated genes and easily recognized by the expression of the Myxovirus-resistance A protein. We suggest that the detection of these two phenotypes might help the selection of patients enrolled in virally-mediated gene therapy trials.
Collapse
Affiliation(s)
- Vladia Monsurrò
- Department of Pathology, University of Verona Medical School, Verona, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Analysis of vaccine-induced T cells in humans with cancer. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2010; 684:178-88. [PMID: 20795549 DOI: 10.1007/978-1-4419-6451-9_14] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Over the past several years, progress in the field of tumor immunology has lead to advances in active immunotherapy and vaccination as a means ofeliciting tumor-specific immune responses to mediate tumor regression and clearance. Developing vaccines targeted against cancer became an important focus as a therapy following the success of viral vaccines in preventing infection and disease. In humans with cancer, similar to viral infections, the host immune system is capable of recognizing antigens expressed on tumor cells. This similarity allows the immunological framework of the viral vaccine to be adapted to the cancer setting in hopes of enhancing human T-cell reactivity against tumor. It is generally believed that a requirement for tumor destruction to occur is the induction of sufficient levels of immune cells with high avidity for recognition of tumor antigens. Moreover, the cells must be targeted to the tumor site and be capable of infiltrating tumor stroma.2 Several tumor-associated antigens (TAA) have been identified in the melanoma model which has allowed for immunization trials to evaluate therapeutic potential of tumor-specific T-cell induction. Some clinical trials reported limited success ofT-cell mediated tumor rejection, reportingpartial or complete regression in 10 to 30% of patients. Although tumor regression was not observed following active immunization in vivo, ex vivo assays evaluating TAA-specific T cells demonstrated tumor recognition and subsequent T-cell activation suggesting that tumor-specific T-cell induction indeed occurs but alone is not adequate to induce tumor regression. Recently, the usefulness and success of active-specific immunization (ASI) against TAAs as a means ofeliciting a tumor-specific immune response leading to tumor regression and clearance has been a topic of debate and discussion.
Collapse
|
49
|
Cancer associated fibroblasts promote tumor growth and metastasis by modulating the tumor immune microenvironment in a 4T1 murine breast cancer model. PLoS One 2009; 4:e7965. [PMID: 19956757 PMCID: PMC2775953 DOI: 10.1371/journal.pone.0007965] [Citation(s) in RCA: 370] [Impact Index Per Article: 23.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2009] [Accepted: 10/30/2009] [Indexed: 12/13/2022] Open
Abstract
Background Local inflammation associated with solid tumors commonly results from factors released by tumor cells and the tumor stroma, and promotes tumor progression. Cancer associated fibroblasts comprise a majority of the cells found in tumor stroma and are appealing targets for cancer therapy. Here, our aim was to determine the efficacy of targeting cancer associated fibroblasts for the treatment of metastatic breast cancer. Methodology/Principal Findings We demonstrate that cancer associated fibroblasts are key modulators of immune polarization in the tumor microenvironment of a 4T1 murine model of metastatic breast cancer. Elimination of cancer associated fibroblasts in vivo by a DNA vaccine targeted to fibroblast activation protein results in a shift of the immune microenvironment from a Th2 to Th1 polarization. This shift is characterized by increased protein expression of IL-2 and IL-7, suppressed recruitment of tumor-associated macrophages, myeloid derived suppressor cells, T regulatory cells, and decreased tumor angiogenesis and lymphangiogenesis. Additionally, the vaccine improved anti-metastatic effects of doxorubicin chemotherapy and enhanced suppression of IL-6 and IL-4 protein expression while increasing recruitment of dendritic cells and CD8+ T cells. Treatment with the combination therapy also reduced tumor-associated Vegf, Pdgfc, and GM-CSF mRNA and protein expression. Conclusions/Significance Our findings demonstrate that cancer associated fibroblasts promote tumor growth and metastasis through their role as key modulators of immune polarization in the tumor microenvironment and are valid targets for therapy of metastatic breast cancer.
Collapse
|
50
|
Iero M, Filipazzi P, Castelli C, Belli F, Valdagni R, Parmiani G, Patuzzo R, Santinami M, Rivoltini L. Modified peptides in anti-cancer vaccines: are we eventually improving anti-tumour immunity? Cancer Immunol Immunother 2009; 58:1159-67. [PMID: 18998128 PMCID: PMC11030573 DOI: 10.1007/s00262-008-0610-6] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2008] [Accepted: 10/03/2008] [Indexed: 12/22/2022]
Abstract
The discovery of tumour antigens recognized by T cells and the features of immune responses directed against them has paved the way to a multitude of clinical studies aimed at boosting anti-tumour T cell immunity as a therapeutic tool for cancer patients. One of the different strategies explored to ameliorate the immunogenicity of tumour antigens in vaccine protocols is represented by the use of optimized peptides or altered peptide ligands, whose amino acid sequence has been modified for improving HLA binding or TCR interaction with respect to native epitopes. However, despite the promising results achieved with preclinical studies, the clinical efficacy of this approach has not yet met the expectations. Although multiple reasons could explain the relative failure of altered peptide ligands as more effective cancer vaccines, the possibility that T cells primed by modified tumour peptides might may be unable to effectively cross-recognize tumour cells has not been sufficiently addressed. Indeed, the introduction of conservative amino acid substitutions may still produce diverse and unpredictable changes in the HLA/peptide interface, with consequent modifications of the TCR repertoire that can interact with the complex. This could lead to the expansion of a broad array of T cells whose TCRs may not necessarily react with equivalent affinity with the original antigenic epitope. Considering the results presently achieved with this vaccine approach, and the emerging availability of alternative strategies for boosting anti-tumour immunity, the use of modified tumour peptides could be reconsidered.
Collapse
Affiliation(s)
- Manuela Iero
- Unit of Immunotherapy of Human Tumours, Fondazione IRCCS Istituto Nazionale Tumori, Via Venezian 1, 20133 Milan, Italy
| | - Paola Filipazzi
- Unit of Immunotherapy of Human Tumours, Fondazione IRCCS Istituto Nazionale Tumori, Via Venezian 1, 20133 Milan, Italy
| | - Chiara Castelli
- Unit of Immunotherapy of Human Tumours, Fondazione IRCCS Istituto Nazionale Tumori, Via Venezian 1, 20133 Milan, Italy
| | - Filiberto Belli
- Unit of Colo-rectal Surgery, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy
| | - Riccardo Valdagni
- Unit of Prostate Cancer Program, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy
| | - Giorgio Parmiani
- Unit of Immunobiotherapy of Solid Tumours, San Raffaele Scientific Institute, Milan, Italy
| | - Roberto Patuzzo
- Unit of Melanoma and Sarcoma Surgery, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy
| | - Mario Santinami
- Unit of Melanoma and Sarcoma Surgery, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy
| | - Licia Rivoltini
- Unit of Immunotherapy of Human Tumours, Fondazione IRCCS Istituto Nazionale Tumori, Via Venezian 1, 20133 Milan, Italy
| |
Collapse
|