1
|
Colley A, Brauns T, Sluder AE, Poznansky MC, Gemechu Y. Immunomodulatory drugs: a promising clinical ally for cancer immunotherapy. Trends Mol Med 2024; 30:765-780. [PMID: 38821771 DOI: 10.1016/j.molmed.2024.05.001] [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: 12/07/2023] [Revised: 04/27/2024] [Accepted: 05/01/2024] [Indexed: 06/02/2024]
Abstract
While immunomodulatory imide drugs (IMiDs) have been authorised for treatment of haematological cancers for over two decades, the appreciation of their ability to stimulate antitumour T cell and natural killer (NK) cell responses is relatively recent. Clinical trial data increasingly show that targeted immunotherapies, such as antibodies, T cells, and vaccines, improve outcomes when delivered in combination with the IMiD derivatives lenalidomide or pomalidomide. Here, we review these clinical data to highlight the relevance of IMiDs in combinatorial immunotherapy for both haematological and solid tumours. Further research into the molecular mechanisms of IMiDs and an increased understanding of their immunomodulatory effects may refine the specific applications of IMiDs and improve the design of future clinical trials, moving IMiDs to the forefront of combinatorial cancer immunotherapy.
Collapse
Affiliation(s)
- Abigail Colley
- Vaccine and Immunotherapy Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA; Department of Oncology, University of Cambridge, Cambridge, UK
| | - Timothy Brauns
- Vaccine and Immunotherapy Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Ann E Sluder
- Vaccine and Immunotherapy Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Mark C Poznansky
- Vaccine and Immunotherapy Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Yohannes Gemechu
- Vaccine and Immunotherapy Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
2
|
Jasim SA, Farber IM, Noraldeen SAM, Bansal P, Alsaab HO, Abdullaev B, Alkhafaji AT, Alawadi AH, Hamzah HF, Mohammed BA. Incorporation of immunotherapies and nanomedicine to better normalize angiogenesis-based cancer treatment. Microvasc Res 2024; 154:104691. [PMID: 38703993 DOI: 10.1016/j.mvr.2024.104691] [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: 02/19/2024] [Revised: 04/11/2024] [Accepted: 04/27/2024] [Indexed: 05/06/2024]
Abstract
Neoadjuvant targeting of tumor angiogenesis has been developed and approved for the treatment of malignant tumors. However, vascular disruption leads to tumor hypoxia, which exacerbates the treatment process and causes drug resistance. In addition, successful delivery of therapeutic agents and efficacy of radiotherapy require normal vascular networks and sufficient oxygen, which complete tumor vasculopathy hinders their efficacy. In view of this controversy, an optimal dose of FDA-approved anti-angiogenic agents and combination with other therapies, such as immunotherapy and the use of nanocarrier-mediated targeted therapy, could improve therapeutic regimens, reduce the need for administration of high doses of chemotherapeutic agents and subsequently reduce side effects. Here, we review the mechanism of anti-angiogenic agents, highlight the challenges of existing therapies, and present how the combination of immunotherapies and nanomedicine could improve angiogenesis-based tumor treatment.
Collapse
Affiliation(s)
| | - Irina M Farber
- Department of children's diseases of the F. Filatov clinical institute of children's health, I. M. Sechenov First Moscow State Medical University of Health of Russian Federation (Sechenov University), Moscow, Russia
| | | | - Pooja Bansal
- Department of Biotechnology and Genetics, Jain (Deemed-to-be) University, Bengaluru, Karnataka 560069, India; Department of Allied Healthcare and Sciences, Vivekananda Global University, Jaipur, Rajasthan 303012, India
| | - Hashem O Alsaab
- Department of Pharmaceutics and Pharmaceutical Technology, Taif University, Taif 21944, Saudi Arabia
| | - Bekhzod Abdullaev
- Research Department of Biotechnology, New Uzbekistan University, Mustaqillik Avenue 54, Tashkent 100007, Uzbekistan; Department of Oncology, School of Medicine, Central Asian University, Milliy Bog Street 264, Tashkent 111221, Uzbekistan..
| | | | - Ahmed Hussien Alawadi
- College of Technical Engineering, the Islamic University, Najaf, Iraq; College of Technical Engineering, the Islamic University of Al Diwaniyah, Qadisiyyah, Iraq; College of Technical Engineering, the Islamic University of Babylon, Babylon, Iraq
| | - Hamza Fadhel Hamzah
- Department of Medical Laboratories Technology, AL-Nisour University College, Baghdad, Iraq
| | | |
Collapse
|
3
|
Song MS, Nam JH, Noh KE, Lim DS. Dendritic Cell-Based Immunotherapy: The Importance of Dendritic Cell Migration. J Immunol Res 2024; 2024:7827246. [PMID: 38628676 PMCID: PMC11019573 DOI: 10.1155/2024/7827246] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 03/22/2024] [Accepted: 03/27/2024] [Indexed: 04/19/2024] Open
Abstract
Dendritic cells (DCs) are specialized antigen-presenting cells that are crucial for maintaining self-tolerance, initiating immune responses against pathogens, and patrolling body compartments. Despite promising aspects, DC-based immunotherapy faces challenges that include limited availability, immune escape in tumors, immunosuppression in the tumor microenvironment, and the need for effective combination therapies. A further limitation in DC-based immunotherapy is the low population of migratory DC (around 5%-10%) that migrate to lymph nodes (LNs) through afferent lymphatics depending on the LN draining site. By increasing the population of migratory DCs, DC-based immunotherapy could enhance immunotherapeutic effects on target diseases. This paper reviews the importance of DC migration and current research progress in the context of DC-based immunotherapy.
Collapse
Affiliation(s)
- Min-Seon Song
- Department of Bioconvergence, Graduate School and Department of Biotechnology, CHA University, 335 Pangyo-ro, Bundang-gu, Seongnam-si, Gyeonggi-do 13488, Republic of Korea
| | - Ji-Hee Nam
- Department of Bioconvergence, Graduate School and Department of Biotechnology, CHA University, 335 Pangyo-ro, Bundang-gu, Seongnam-si, Gyeonggi-do 13488, Republic of Korea
| | - Kyung-Eun Noh
- Department of Bioconvergence, Graduate School and Department of Biotechnology, CHA University, 335 Pangyo-ro, Bundang-gu, Seongnam-si, Gyeonggi-do 13488, Republic of Korea
| | - Dae-Seog Lim
- Department of Bioconvergence, Graduate School and Department of Biotechnology, CHA University, 335 Pangyo-ro, Bundang-gu, Seongnam-si, Gyeonggi-do 13488, Republic of Korea
| |
Collapse
|
4
|
Onyshchenko K, Luo R, Rao X, Zhang X, Gaedicke S, Grosu AL, Firat E, Niedermann G. Hypofractionated radiotherapy combined with lenalidomide improves systemic antitumor activity in mouse solid tumor models. Theranostics 2024; 14:2573-2588. [PMID: 38646638 PMCID: PMC11024858 DOI: 10.7150/thno.88864] [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: 08/06/2023] [Accepted: 03/02/2024] [Indexed: 04/23/2024] Open
Abstract
Background: Hypofractionated radiotherapy (hRT) can induce a T cell-mediated abscopal effect on non-irradiated tumor lesions, especially in combination with immune checkpoint blockade (ICB). However, clinically, this effect is still rare, and ICB-mediated adverse events are common. Lenalidomide (lena) is an anti-angiogenic and immunomodulatory drug used in the treatment of hematologic malignancies. We here investigated in solid tumor models whether lena can enhance the abscopal effect in double combination with hRT. Methods: In two syngeneic bilateral tumor models (B16-CD133 melanoma and MC38 colon carcinoma), the primary tumor was treated with hRT. Lena was given daily for 3 weeks. Besides tumor size and survival, the dependence of the antitumor effects on CD8+ cells, type-I IFN signaling, and T cell costimulation was determined with depleting or blocking antibodies. Tumor-specific CD8+ T cells were quantified, and their differentiation and effector status were characterized by multicolor flow cytometry using MHC-I tetramers and various antibodies. In addition, dendritic cell (DC)-mediated tumor antigen cross-presentation in vitro and directly ex vivo and the composition of tumor-associated vascular endothelial cells were investigated. Results: In both tumor models, the hRT/lena double combination induced a significant abscopal effect. Control of the non-irradiated secondary tumor and survival were considerably better than with the respective monotherapies. The abscopal effect was strongly dependent on CD8+ cells and associated with an increase in tumor-specific CD8+ T cells in the non-irradiated tumor and its draining lymph nodes. Additionally, we found more tumor-specific T cells with a stem-like (TCF1+ TIM3- PD1+) and a transitory (TCF1- TIM3+ CD101- PD1+) exhausted phenotype and more expressing effector molecules such as GzmB, IFNγ, and TNFα. Moreover, in the non-irradiated tumor, hRT/lena treatment also increased DCs cross-presenting a tumor model antigen. Blocking type-I IFN signaling, which is essential for cross-presentation, completely abrogated the abscopal effect. A gene expression analysis of bone marrow-derived DCs revealed that lena augmented the expression of IFN response genes and genes associated with differentiation, maturation (including CD70, CD83, and CD86), migration to lymph nodes, and T cell activation. Flow cytometry confirmed an increase in CD70+ CD83+ CD86+ DCs in both irradiated and abscopal tumors. Moreover, the hRT/lena-induced abscopal effect was diminished when these costimulatory molecules were blocked simultaneously using antibodies. In line with the enhanced infiltration by DCs and tumor-specific CD8+ T cells, including more stem-like cells, hRT/lena also increased tumor-associated high endothelial cells (TA-HECs) in the non-irradiated tumor. Conclusions: We demonstrate that lena can augment the hRT-induced abscopal effect in mouse solid tumor models in a CD8 T cell- and IFN-I-dependent manner, correlating with enhanced anti-tumor CD8 T cell immunity, DC cross-presentation, and TA-HEC numbers. Our findings may be helpful for the planning of clinical trials in (oligo)metastatic patients.
Collapse
Affiliation(s)
- Kateryna Onyshchenko
- Department of Radiation Oncology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Faculty of Biology, University of Freiburg, Freiburg, Germany
- Laboratory of Biosynthesis of Nucleic Acids, Institute of Molecular Biology and Genetics of NASU, Kyiv, Ukraine
- German Cancer Consortium (DKTK), Partner Site Freiburg, Freiburg, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Ren Luo
- Department of Radiation Oncology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Faculty of Biology, University of Freiburg, Freiburg, Germany
- German Cancer Consortium (DKTK), Partner Site Freiburg, Freiburg, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
- Division of Thoracic Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Department of Radiation Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Xi Rao
- Department of Radiation Oncology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- German Cancer Consortium (DKTK), Partner Site Freiburg, Freiburg, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Xuanwei Zhang
- Division of Thoracic Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Department of Radiation Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Simone Gaedicke
- Department of Radiation Oncology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Anca-Ligia Grosu
- Department of Radiation Oncology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- German Cancer Consortium (DKTK), Partner Site Freiburg, Freiburg, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Elke Firat
- Department of Radiation Oncology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Gabriele Niedermann
- Department of Radiation Oncology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- German Cancer Consortium (DKTK), Partner Site Freiburg, Freiburg, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
5
|
Darvishi M, Tosan F, Nakhaei P, Manjili DA, Kharkouei SA, Alizadeh A, Ilkhani S, Khalafi F, Zadeh FA, Shafagh SG. Recent progress in cancer immunotherapy: Overview of current status and challenges. Pathol Res Pract 2023; 241:154241. [PMID: 36543080 DOI: 10.1016/j.prp.2022.154241] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2022] [Revised: 11/20/2022] [Accepted: 11/22/2022] [Indexed: 11/25/2022]
Abstract
Cancer treatment is presently one of the most important challenges in medical science. Surgery, chemotherapy, radiotherapy, or combining these methods is used to eliminate the tumor. Hormone therapy, bone marrow transplantation, stem cell therapy as well as immunotherapy are other well-known therapeutic modalities. Immunotherapy, as the most important complementary method, uses the immune system for treating cancer followed by surgery, chemotherapy, and radiotherapy. This method is systematically used to prevent malignancies development mainly via potentiating antitumor immune cells activation and conversely compromising their exhaustion with the lowest negative effects on healthy cells. Active immunotherapy can be employed for cancer immunotherapy by directly using the ingredients of the immune system and activating immune responses. On the other hand, inactive immunotherapy is utilized by indirect induction and using immune cell-based products consisting of monoclonal antibodies. It has strongly been proved that combination therapy with immunotherapies and other therapeutic means, such as anti-angiogenic agents, could be a rational plan to treat cancer. Herein, we have focused on recent findings concerning the therapeutic merits of cancer therapy using immune checkpoint inhibitors (ICIs), adoptive cell transfer (ACT) and cancer vaccine alone or in combination with other approaches. Also, we offer a glimpse into the current challenges in this context.
Collapse
Affiliation(s)
- Mohammad Darvishi
- Infectious Diseases and Tropical Medicine Research Center (IDTMRC), Department of Aerospace and Subaquatic Medicine, AJA University of Medicinal Sciences, Tehran, Iran.
| | - Foad Tosan
- Student Research Committee, Semnan University of Medical Sciences, Semnan, Iran.
| | - Pooria Nakhaei
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| | - Danial Amiri Manjili
- Department of Infectious Disease, School of Medicine, Babol University of Medical Sciences, Babol, Iran.
| | | | - Ali Alizadeh
- Department of Digital Health, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| | - Saba Ilkhani
- Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Farima Khalafi
- Department of Pharmacology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | | | | |
Collapse
|
6
|
Sánchez-León ML, Jiménez-Cortegana C, Cabrera G, Vermeulen EM, de la Cruz-Merino L, Sánchez-Margalet V. The effects of dendritic cell-based vaccines in the tumor microenvironment: Impact on myeloid-derived suppressor cells. Front Immunol 2022; 13:1050484. [PMID: 36458011 PMCID: PMC9706090 DOI: 10.3389/fimmu.2022.1050484] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Accepted: 10/27/2022] [Indexed: 09/27/2023] Open
Abstract
Dendritic cells (DCs) are a heterogenous population of professional antigen presenting cells whose main role is diminished in a variety of malignancies, including cancer, leading to ineffective immune responses. Those mechanisms are inhibited due to the immunosuppressive conditions found in the tumor microenvironment (TME), where myeloid-derived suppressor cells (MDSCs), a heterogeneous population of immature myeloid cells known to play a key role in tumor immunoevasion by inhibiting T-cell responses, are extremely accumulated. In addition, it has been demonstrated that MDSCs not only suppress DC functions, but also their maturation and development within the myeloid linage. Considering that an increased number of DCs as well as the improvement in their functions boost antitumor immunity, DC-based vaccines were developed two decades ago, and promising results have been obtained throughout these years. Therefore, the remodeling of the TME promoted by DC vaccination has also been explored. Here, we aim to review the effectiveness of different DCs-based vaccines in murine models and cancer patients, either alone or synergistically combined with other treatments, being especially focused on their effect on the MDSC population.
Collapse
Affiliation(s)
- María Luisa Sánchez-León
- Department of Medical Biochemistry and Molecular Biology, School of Medicine, University of Seville, Seville, Spain
- Medical Oncology Service, Virgen Macarena University Hospital, Seville, Spain
| | - Carlos Jiménez-Cortegana
- Department of Medical Biochemistry and Molecular Biology, School of Medicine, University of Seville, Seville, Spain
- Department of Laboratory Medicine, Virgen Macarena University Hospital, Seville, Spain
| | - Gabriel Cabrera
- Laboratorio de Tecnología Inmunológica, Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, Santa Fe capital, Argentina
| | - Elba Mónica Vermeulen
- Laboratorio de Células Presentadoras de Antígeno y Respuesta Inflamatoria, Instituto de Medicina Experimental (IMEX) - CONICET, Academia Nacional de Medicina, Buenos Aires, Argentina
| | | | - Victor Sánchez-Margalet
- Department of Medical Biochemistry and Molecular Biology, School of Medicine, University of Seville, Seville, Spain
- Department of Laboratory Medicine, Virgen Macarena University Hospital, Seville, Spain
| |
Collapse
|
7
|
Tokhanbigli S, Alavifard H, Asadzadeh Aghdaei H, Zali MR, Baghaei K. Combination of pioglitazone and dendritic cell to optimize efficacy of immune cell therapy in CT26 tumor models. BIOIMPACTS : BI 2022; 13:333-346. [PMID: 37645031 PMCID: PMC10460770 DOI: 10.34172/bi.2022.24209] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/11/2021] [Revised: 02/28/2022] [Accepted: 06/20/2022] [Indexed: 08/31/2023]
Abstract
Introduction The maturation faith of dendritic cells is restrained by the inflammatory environment and cytokines, such as interleukin-6 and its downstream component. Therefore, introducing the suitable antigen to dendritic cells is crucial. However, reducing the severity of the suppressive tumor microenvironment is indispensable. The present study examined the combination therapy of lymphocyte antigen 6 family member E (LY6E) pulsed mature dendritic cells (LPMDCs) and pioglitazone against colorectal cancer (CRC) to elevate the effectiveness of cancer treatment through probable role of pioglitazone on inhibiting IL-6/STAT3 pathway. Methods Dendritic cells were generated from murine bone marrow and were pulsed with lymphocyte antigen 6 family member E peptide to assess antigen-specific T-cell proliferation and cytotoxicity assay with Annexin/PI. The effect of pioglitazone on interleukin (IL)-6/STAT3 was evaluated in vitro by real-time polymerase chain reaction (PCR). Afterward, the CRC model was established by subcutaneous injection of CT26, mouse colon carcinoma cell line, in female mice. After treatment, tumor, spleen, and lymph nodes samples were removed for histopathological, ELISA, and real-time PCR analysis. Results In vitro results revealed the potential of lysate-pulsed dendritic cells in the proliferation of double-positive CD3-8 splenocytes and inducing immunogenic cell death responses, whereas pioglitazone declined the expression of IL-6/STAT3 in colorectal cell lines. In animal models, the recipient of LPMDCs combined with pioglitazone demonstrated high tumor-infiltrating lymphocytes. Elevating the IL-12 and interferon-gamma (IFN-γ) levels and prolonged survival in lysate-pulsed dendritic cell and combination groups were observed. Conclusion Pioglitazone could efficiently ameliorate the immunosuppressive feature of the tumor microenvironment, mainly through IL-6. Accordingly, applying this drug combined with LPMDCs provoked substantial CD8 positive responses in tumor-challenged animal models.
Collapse
Affiliation(s)
- Samaneh Tokhanbigli
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Helia Alavifard
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hamid Asadzadeh Aghdaei
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Reza Zali
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Kaveh Baghaei
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
8
|
Zhao L, Zhang S, Kepp O, Kroemer G, Liu P. Dendritic cell transfer for cancer immunotherapy. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2022; 370:33-64. [PMID: 35798506 DOI: 10.1016/bs.ircmb.2022.03.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Dendritic cells (DCs) play a major role in cancer immunosurveillance as they bridge innate and adaptive immunity by detecting tumor-associated antigens and presenting them to T lymphocytes. The adoptive transfer of antigen loaded DCs has been proposed as an immunotherapeutic approach for the treatment of various types of cancer. Nevertheless, despite promising preclinical data, the therapeutic efficacy of DC transfer is still deceptive in cancer patients. Here we summarize recent findings in DC biology with a special focus on the development of actionable therapeutic strategies and discuss experimental and clinical approaches that aim at improving the efficacy of DC-based immunotherapies, including, but not limited to, optimized DC production and antigen loading, stimulated maturation, the co-treatment with additional immunotherapies, as well as the inhibition of DC checkpoints.
Collapse
Affiliation(s)
- Liwei Zhao
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Center, Université Paris Saclay, Villejuif, France; Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université de Paris, Sorbonne Université, Inserm U1138, Institut Universitaire de France, Paris, France
| | - Shuai Zhang
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Center, Université Paris Saclay, Villejuif, France; Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université de Paris, Sorbonne Université, Inserm U1138, Institut Universitaire de France, Paris, France
| | - Oliver Kepp
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Center, Université Paris Saclay, Villejuif, France; Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université de Paris, Sorbonne Université, Inserm U1138, Institut Universitaire de France, Paris, France.
| | - Guido Kroemer
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Center, Université Paris Saclay, Villejuif, France; Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université de Paris, Sorbonne Université, Inserm U1138, Institut Universitaire de France, Paris, France; Institut du Cancer Paris Carpem, Department of Biology, Hôpital Européen Georges Pompidou, APHP, Paris, France.
| | - Peng Liu
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Center, Université Paris Saclay, Villejuif, France; Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université de Paris, Sorbonne Université, Inserm U1138, Institut Universitaire de France, Paris, France.
| |
Collapse
|
9
|
Ansari MJ, Bokov D, Markov A, Jalil AT, Shalaby MN, Suksatan W, Chupradit S, AL-Ghamdi HS, Shomali N, Zamani A, Mohammadi A, Dadashpour M. Cancer combination therapies by angiogenesis inhibitors; a comprehensive review. Cell Commun Signal 2022; 20:49. [PMID: 35392964 PMCID: PMC8991477 DOI: 10.1186/s12964-022-00838-y] [Citation(s) in RCA: 65] [Impact Index Per Article: 32.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Accepted: 02/03/2022] [Indexed: 02/06/2023] Open
Abstract
Abnormal vasculature is one of the most conspicuous traits of tumor tissue, largely contributing to tumor immune evasion. The deregulation mainly arises from the potentiated pro-angiogenic factors secretion and can also target immune cells' biological events, such as migration and activation. Owing to this fact, angiogenesis blockade therapy was established to fight cancer by eliminating the nutrient and oxygen supply to the malignant cells by impairing the vascular network. Given the dominant role of vascular-endothelium growth factor (VEGF) in the angiogenesis process, the well-known anti-angiogenic agents mainly depend on the targeting of its actions. However, cancer cells mainly show resistance to anti-angiogenic agents by several mechanisms, and also potentiated local invasiveness and also distant metastasis have been observed following their administration. Herein, we will focus on clinical developments of angiogenesis blockade therapy, more particular, in combination with other conventional treatments, such as immunotherapy, chemoradiotherapy, targeted therapy, and also cancer vaccines. Video abstract.
Collapse
Affiliation(s)
- Mohammad Javed Ansari
- Department of Pharmaceutics, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj, Kingdom of Saudi Arabia
| | - Dmitry Bokov
- Institute of Pharmacy, Sechenov First Moscow State Medical University, 8 Trubetskaya St., bldg. 2, Moscow, 119991 Russian Federation
- Laboratory of Food Chemistry, Federal Research Center of Nutrition, Biotechnology and Food Safety, 2/14 Ustyinsky pr., Moscow, 109240 Russian Federation
| | - Alexander Markov
- Tyumen State Medical University, Tyumen, Russian Federation
- Industrial University, Tyumen, Russian Federation
| | - Abduladheem Turki Jalil
- Faculty of Biology and Ecology, Yanka Kupala State University of Grodno, 230023 Grodno, Belarus
- College of Technical Engineering, The Islamic University, Najaf, Iraq
- Department of Dentistry, Kut University College, Kut, Wasit 52001 Iraq
| | - Mohammed Nader Shalaby
- Biological Sciences and Sports Health Department, Faculty of Physical Education, Suez Canal University, Ismailia, Egypt
| | - Wanich Suksatan
- Faculty of Nursing, HRH Princess Chulabhorn College of Medical Science, Chulabhorn Royal Academy, Bangkok, Thailand
| | - Supat Chupradit
- Department of Occupational Therapy, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, 50200 Thailand
| | - Hasan S. AL-Ghamdi
- Internal Medicine Department, Division of Dermatology, Albaha University, Al Bahah, Kingdom of Saudi Arabia
| | - Navid Shomali
- Immunology Research Center (IRC), Tabriz University of Medical Sciences, Tabriz, Iran
| | - Amir Zamani
- Shiraz Transplant Center, Abu Ali Sina Hospital, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Ali Mohammadi
- Department of Neurology, Imam Khomeini Hospital, Urmia University of Medical Sciences, Urmia, Iran
| | - Mehdi Dadashpour
- Department of Medical Biotechnology, Faculty of Medicine, Semnan University of Medical Sciences, Semnan, Iran
| |
Collapse
|
10
|
Zhang T, Yang Y, Huang L, Liu Y, Chong G, Yin W, Dong H, Li Y, Li Y. Biomimetic and Materials-Potentiated Cell Engineering for Cancer Immunotherapy. Pharmaceutics 2022; 14:pharmaceutics14040734. [PMID: 35456568 PMCID: PMC9024915 DOI: 10.3390/pharmaceutics14040734] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Revised: 03/17/2022] [Accepted: 03/25/2022] [Indexed: 02/01/2023] Open
Abstract
In cancer immunotherapy, immune cells are the main force for tumor eradication. However, they appear to be dysfunctional due to the taming of the tumor immunosuppressive microenvironment. Recently, many materials-engineered strategies are proposed to enhance the anti-tumor effect of immune cells. These strategies either utilize biomimetic materials, as building blocks to construct inanimate entities whose functions are similar to natural living cells, or engineer immune cells with functional materials, to potentiate their anti-tumor effects. In this review, we will summarize these advanced strategies in different cell types, as well as discussing the prospects of this field.
Collapse
Affiliation(s)
- Tingting Zhang
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai 200092, China; (T.Z.); (Y.Y.); (L.H.); (Y.L.); (G.C.); (W.Y.); (Y.L.)
| | - Yushan Yang
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai 200092, China; (T.Z.); (Y.Y.); (L.H.); (Y.L.); (G.C.); (W.Y.); (Y.L.)
| | - Li Huang
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai 200092, China; (T.Z.); (Y.Y.); (L.H.); (Y.L.); (G.C.); (W.Y.); (Y.L.)
| | - Ying Liu
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai 200092, China; (T.Z.); (Y.Y.); (L.H.); (Y.L.); (G.C.); (W.Y.); (Y.L.)
| | - Gaowei Chong
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai 200092, China; (T.Z.); (Y.Y.); (L.H.); (Y.L.); (G.C.); (W.Y.); (Y.L.)
| | - Weimin Yin
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai 200092, China; (T.Z.); (Y.Y.); (L.H.); (Y.L.); (G.C.); (W.Y.); (Y.L.)
| | - Haiqing Dong
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration, Ministry of Education, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200092, China
- Correspondence: (H.D.); (Y.L.); Tel.: +86-021-659-819-52 (H.D. & Y.L.)
| | - Yan Li
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai 200092, China; (T.Z.); (Y.Y.); (L.H.); (Y.L.); (G.C.); (W.Y.); (Y.L.)
- Correspondence: (H.D.); (Y.L.); Tel.: +86-021-659-819-52 (H.D. & Y.L.)
| | - Yongyong Li
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai 200092, China; (T.Z.); (Y.Y.); (L.H.); (Y.L.); (G.C.); (W.Y.); (Y.L.)
| |
Collapse
|
11
|
Inhibition of apelin/APJ axis enhances the potential of dendritic cell-based vaccination to modulate TH1 and TH2 cell-related immune responses in an animal model of metastatic breast cancer. Adv Med Sci 2022; 67:170-178. [PMID: 35290873 DOI: 10.1016/j.advms.2022.02.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 01/05/2022] [Accepted: 02/24/2022] [Indexed: 12/19/2022]
Abstract
PURPOSE The immunosuppressive microenvironment of tumors reduces the effectiveness of immunotherapies. Apelin as an immunosuppressor peptide is expressed in the microenvironment of many tumors. Thus, inhibition of apelin-related protumor activities can promote the effectiveness of cancer immunotherapy. Here, we investigated the efficacy of a dendritic cell (DC) vaccine in combination with an apelin receptor antagonist, ML221, to modulate Th1 and Th2 cell-related responses in breast cancer-bearing mice. MATERIALS AND METHODS Tumor was induced in female BALB/c mice by injecting 7 × 105 4T1 cells in the right flank. Tumor-bearing mice were then given PBS, ML221, DC vaccine and "ML221 + DC vaccine" for 21 days. On day 37, mice were sacrificed and the frequency of Th1/Th2 cells in spleen and serum levels of IFN-γ/IL-10 were determined using flow cytometry and ELISA, respectively. Lung metastasis was evaluated in lung tissues stained with hematoxylin and eosin. Finally, the obtained data were analyzed using appropriate statistical tests. RESULTS Combination therapy with ML221 + DC vaccination was more effective in reducing tumor growth (P < 0.0001), preventing lung metastasis (P < 0.0001) and increasing survival rate (P < 0.01) compared to the control group. Moreover, combination treatment substantially increased the frequency of Th1 cells while decreasing the frequency of Th2 cells in the spleen compared to the control group (P < 0.01). It also reduced serum levels of IL-10 compared with the control group (P < 0.05). CONCLUSION Our findings showed that combination therapy using ML221 + DC vaccine can be considered as an effective cancer therapeutic program to potentiate anti-tumor immune responses.
Collapse
|
12
|
Immunomodulatory drugs suppress Th1-inducing ability of dendritic cells but enhance Th2-mediated allergic responses. Blood Adv 2021; 4:3572-3585. [PMID: 32761232 DOI: 10.1182/bloodadvances.2019001410] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Accepted: 05/01/2020] [Indexed: 11/20/2022] Open
Abstract
Immunomodulatory drugs (IMiDs), lenalidomide and pomalidomide, are widely used treatments for multiple myeloma; however, they occasionally lead to episodes of itchy skin and rashes. Here, we analyzed the effects of IMiDs on human myeloid dendritic cells (mDCs) as major regulators of Th1 or Th2 responses and the role they play in allergy. We found that lenalidomide and pomalidomide used at clinical concentrations did not affect the survival or CD86 and OX40-ligand expression of blood mDCs in response to lipopolysaccharide (LPS) and thymic stromal lymphopoietin (TSLP) stimulation. Both lenalidomide and pomalidomide dose-dependently inhibited interleukin-12 (IL-12) and TNF production and STAT4 expression, and enhanced IL-10 production in response to LPS. When stimulated with TSLP, both IMiDs significantly enhanced CCL17 production and STAT6 and IRF4 expression and promoted memory Th2-cell responses. In 46 myeloma patients, serum CCL17 levels at the onset of lenalidomide-associated rash were significantly higher than those without rashes during lenalidomide treatment and those before treatment. Furthermore, serum CCL17 levels in patients who achieved a very good partial response (VGPR) were significantly higher compared with a less than VGPR during lenalidomide treatment. The median time to next treatment was significantly longer in lenalidomide-treated patients with rashes than those without. Collectively, IMiDs suppressed the Th1-inducing capacity of DCs, instead promoting a Th2 response. Thus, the lenalidomide-associated rashes might be a result of an allergic response driven by Th2-axis activation. Our findings suggest clinical efficacy and rashes as a side effect of IMiDs are inextricably linked through immunostimulation.
Collapse
|
13
|
Ujiie N, Enomoto Y, Takido N, Kawaharada Y, Zuguchi M, Kubota Y. Rapid progression of gastric cancer with liver metastasis after discontinuation of lenalidomide in a patient with concurrent multiple myeloma: A case report. Int J Surg Case Rep 2021; 81:105834. [PMID: 33887857 PMCID: PMC8050724 DOI: 10.1016/j.ijscr.2021.105834] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2021] [Accepted: 03/22/2021] [Indexed: 11/25/2022] Open
Abstract
This is the first report regarding the perioperative management of patients with concurrent multiple myeloma and malignant solid tumor. Our case highlights the need to devise a perioperative management strategy for multiple myeloma patients. Surgeons should be familiar with the risks associated with discontinuation of multiple myeloma drugs. Continuation of the multiple myeloma agent may be preferable in patients who do not have marked myelosuppression.
Introduction The synchronous incidence of multiple myeloma (MM) and other primary malignant solid tumor is rare. No detailed studies have been published regarding the perioperative management of patients with concurrent MM and malignant solid tumor. We report a patient with concurrent MM and gastric cancer who experienced rapid progression of liver metastasis after lenalidomide was discontinued. Presentation of case An 82-year-old woman with MM was diagnosed with clinical T3N2M0 gastric cancer, and MM had been maintained in remission with lenalidomide. Preoperatively, pancytopenia was found, and lenalidomide was discontinued and lenograstim was administered. Blood transfusions were also administered preoperatively due to anemia caused by tumor bleeding. Surgery was performed after her pancytopenia improved. Intraoperatively, several nodules were found on the liver, which were diagnosed as adenocarcinoma metastases. On postoperative day 13, a low density mass in the liver that was not observed before surgery was shown. The patient received best supportive care because she did not desire adjuvant chemotherapy for gastric cancer or resumption of treatment for MM. She died of progressive gastric cancer on postoperative day 80. Discussion Discontinuation of lenalidomide in our case may have promoted tumor angiogenesis and lowered antitumor immunity, causing rapid tumor growth and liver metastasis. Continuation of the MM agent may be preferable in patients who do not have marked myelosuppression. Conclusion Surgeons should be familiar with the risks associated with discontinuation of MM drugs when operating on patients with MM and concurrent malignant solid tumor.
Collapse
Affiliation(s)
- Naoto Ujiie
- Division of Surgery, Tohoku University Hospital, Sendai, Miyagi, Japan.
| | | | - Naruhito Takido
- Division of Surgery, Tohoku University Hospital, Sendai, Miyagi, Japan
| | | | - Masashi Zuguchi
- Division of Surgery, Hiraka General Hospital, Yokote, Akita, Japan
| | - Yosuke Kubota
- Division of Surgery, Hiraka General Hospital, Yokote, Akita, Japan
| |
Collapse
|
14
|
In Vivo Antitumor Effect against Murine Cells of CT26 Colon Cancer and EL4 Lymphoma by Autologous Whole Tumor Dead Cells. BIOMED RESEARCH INTERNATIONAL 2021; 2021:6626851. [PMID: 33623783 PMCID: PMC7875630 DOI: 10.1155/2021/6626851] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 01/08/2021] [Accepted: 01/19/2021] [Indexed: 11/17/2022]
Abstract
Active immunotherapy against cancer is based on immune system stimulation, triggering efficient and long-lasting antigen-specific immune responses. Immunization strategies using whole dead cells from tumor tissue, containing specific antigens inside, have become a promising approach, providing efficient lymphocyte activation through dendritic cells (DCs). In this work, we generate whole dead tumor cells from CT26, E.G7, and EL4 live tumor cells as antigen sources, which termed immunogenic cell bodies (ICBs), generated by a simple and cost-efficient starvation-protocol, in order to determine whether are capable of inducing a transversal anticancer response regardless of the tumor type, in a similar way to what we describe previously with B16 melanoma. We evaluated the anticancer effects of immunization with doses of ICBs in syngeneic murine tumor models. Our results showed that mice's immunization with ICBs-E.G7 and ICBs-CT26 generate 18% and 25% of tumor-free animals, respectively. On the other hand, all carrying tumor-animals and immunized with ICBs, including ICBs-EL4, showed a significant delay in their growth compared to not immunized animals. These effects relate to DCs maturation, cytokine production, increase in CD4+T-bet+ and CD4+ROR-γt+ population, and decrease of T regulatory lymphocytes in the spleen. Altogether, our data suggest that whole dead tumor cell-based cancer immunotherapy generated by a simple starvation protocol is a promising way to develop complementary, innovative, and affordable antitumor therapies in a broad spectrum of tumors.
Collapse
|
15
|
Yu L, Feng R, Zhu L, Hao Q, Chu J, Gu Y, Luo Y, Zhang Z, Chen G, Chen H. Promoting the activation of T cells with glycopolymer-modified dendritic cells by enhancing cell interactions. SCIENCE ADVANCES 2020; 6:eabb6595. [PMID: 33219021 PMCID: PMC7679162 DOI: 10.1126/sciadv.abb6595] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Accepted: 10/08/2020] [Indexed: 05/12/2023]
Abstract
Dendritic cell (DC) modification to enhance antigen presentation is a valuable strategy in cancer immune therapy. Other than focusing on regulating interactions between DC and antigens, we intend to promote cell interactions between DC and T cell by cell surface engineering. T cell activation is greatly improved and generates higher tumor toxicity with the aid of the synthetic glycopolymer modified on the DC surface, although the glycopolymer alone shows no effect. The great promotion of DC-T cell attraction is revealed by cell image tracking in terms of both frequency and duration of contacts. Our findings provide a new method of T cell activation by these engineered "sweet DCs." This strategy is beneficial for developing more efficient DC-based vaccines.
Collapse
Affiliation(s)
- Liyin Yu
- State and Local Joint Engineering Laboratory for Novel Functional Polymeric Materials, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, 199 Ren'ai Road, Suzhou 215123, P. R. China
| | - Ruyan Feng
- State and Local Joint Engineering Laboratory for Novel Functional Polymeric Materials, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, 199 Ren'ai Road, Suzhou 215123, P. R. China
| | - Lijuan Zhu
- State and Local Joint Engineering Laboratory for Novel Functional Polymeric Materials, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, 199 Ren'ai Road, Suzhou 215123, P. R. China
| | - Qing Hao
- State and Local Joint Engineering Laboratory for Novel Functional Polymeric Materials, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, 199 Ren'ai Road, Suzhou 215123, P. R. China
| | - Jiacheng Chu
- Institute of Functional Nano and Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou, Jiangsu 215123, P. R. China
| | - Yan Gu
- State and Local Joint Engineering Laboratory for Novel Functional Polymeric Materials, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, 199 Ren'ai Road, Suzhou 215123, P. R. China
| | - Yan Luo
- State and Local Joint Engineering Laboratory for Novel Functional Polymeric Materials, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, 199 Ren'ai Road, Suzhou 215123, P. R. China
| | - Zexin Zhang
- State and Local Joint Engineering Laboratory for Novel Functional Polymeric Materials, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, 199 Ren'ai Road, Suzhou 215123, P. R. China
| | - Gaojian Chen
- State and Local Joint Engineering Laboratory for Novel Functional Polymeric Materials, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, 199 Ren'ai Road, Suzhou 215123, P. R. China.
- Center for Soft Condensed Matter Physics and Interdisciplinary Research, Soochow University, Suzhou 215006, P. R. China
| | - Hong Chen
- State and Local Joint Engineering Laboratory for Novel Functional Polymeric Materials, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, 199 Ren'ai Road, Suzhou 215123, P. R. China.
| |
Collapse
|
16
|
Qiang W, Dai Y, Sun G, Xing X, Sun X. Development of a prognostic index of colon adenocarcinoma based on immunogenomic landscape analysis. ANNALS OF TRANSLATIONAL MEDICINE 2020; 8:284. [PMID: 32355728 PMCID: PMC7186653 DOI: 10.21037/atm.2020.03.09] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Background Colon adenocarcinoma (COAD) is one of the most commonly diagnosed cancers, and it is closely related to the immune microenvironment. Considering that immunotherapy is not effective for all COAD patients, it is necessary to identify the effective population before administering treatment. In this study, we established an independent prognostic index based on immune-related genes (IRGs), in order to evaluate the clinical outcome of COAD. Methods The gene expression profiles and IRGs taken from The Cancer Genome Atlas (TCGA) and Immunology Database and Analysis Portal (ImmPort), respectively, were integrated in order to identify the differentially expressed IRGs. Functional enrichment analysis was conducted and the prognostic value of survival-related IRGs was determined. Based on Cox regression analysis, the IRG-based prognostic index (IRGPI) was established, and the model was evaluated and applied. Results A total of 51 differentially expressed survival-related IRGs were identified. The most significant signaling pathway was "cytokine-cytokine receptor interaction". The index established herein was based on 12 survival-related IRGs, and it was highly accurate in monitoring prognosis. Moreover, the IRGPI was significantly correlated with multiple clinicopathologic factors, as well as with the infiltration of immune cells. Conclusions An independent IRGPI was established in order to assess the immune status and tumor prognosis in COAD patients. This index can serve as a robust biomarker in clinical prognosis applications, including cancer immunotherapy.
Collapse
Affiliation(s)
- Weijie Qiang
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100193, China.,Key Laboratory of New Drug Discovery Based on Classic Chinese Medicine Prescription, Chinese Academy of Medical Sciences, Beijing 100193, China
| | - Yifei Dai
- School of Medicine, Tsinghua University, Beijing 100084, China
| | - Guibo Sun
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100193, China.,Key Laboratory of New Drug Discovery Based on Classic Chinese Medicine Prescription, Chinese Academy of Medical Sciences, Beijing 100193, China
| | - Xiaoyan Xing
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100193, China.,Key Laboratory of New Drug Discovery Based on Classic Chinese Medicine Prescription, Chinese Academy of Medical Sciences, Beijing 100193, China
| | - Xiaobo Sun
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100193, China.,Key Laboratory of New Drug Discovery Based on Classic Chinese Medicine Prescription, Chinese Academy of Medical Sciences, Beijing 100193, China
| |
Collapse
|
17
|
Models for Monocytic Cells in the Tumor Microenvironment. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020. [PMID: 32036607 DOI: 10.1007/978-3-030-35723-8_7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/18/2023]
Abstract
Monocytes (Mos) are immune cells that critically regulate cancer, enabling tumor growth and modulating metastasis. Mos can give rise to tumor-associated macrophages (TAMs) and Mo-derived dendritic cells (moDCs), all of which shape the tumor microenvironment (TME). Thus, understanding their roles in the TME is key for improved immunotherapy. Concurrently, various biological and mechanical factors including changes in local cytokines, extracellular matrix production, and metabolic changes in the TME affect the roles of monocytic cells. As such, relevant TME models are critical to achieve meaningful insight on the precise functions, mechanisms, and effects of monocytic cells. Notably, murine models have yielded significant insight into human Mo biology. However, many of these results have yet to be confirmed in humans, reinforcing the need for improved in vitro human TME models for the development of cancer interventions. Thus, this chapter (1) summarizes current insight on the tumor biology of Mos, TAMs, and moDCs, (2) highlights key therapeutic applications relevant to these cells, and (3) discusses various TME models to study their TME-related activity. We conclude with a perspective on the future research trajectory of this topic.
Collapse
|
18
|
Leystra AA, Clapper ML. Gut Microbiota Influences Experimental Outcomes in Mouse Models of Colorectal Cancer. Genes (Basel) 2019; 10:genes10110900. [PMID: 31703321 PMCID: PMC6895921 DOI: 10.3390/genes10110900] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Revised: 10/31/2019] [Accepted: 11/05/2019] [Indexed: 02/06/2023] Open
Abstract
Colorectal cancer (CRC) is a leading cause of cancer-related deaths worldwide. Mouse models are a valuable resource for use throughout the development and testing of new therapeutic strategies for CRC. Tumorigenesis and response to therapy in humans and mouse models alike are influenced by the microbial communities that colonize the gut. Differences in the composition of the gut microbiota can confound experimental findings and reduce the replicability and translatability of the resulting data. Despite this, the contribution of resident microbiota to preclinical tumor models is often underappreciated. This review does the following: (1) summarizes evidence that the gut microbiota influence CRC disease phenotypes; (2) outlines factors that can influence the composition of the gut microbiota; and (3) provides strategies that can be incorporated into the experimental design, to account for the influence of the microbiota on intestinal phenotypes in mouse models of CRC. Through careful experimental design and documentation, mouse models can continue to rapidly advance efforts to prevent and treat colon cancer.
Collapse
|
19
|
Kondratova M, Czerwinska U, Sompairac N, Amigorena SD, Soumelis V, Barillot E, Zinovyev A, Kuperstein I. A multiscale signalling network map of innate immune response in cancer reveals cell heterogeneity signatures. Nat Commun 2019; 10:4808. [PMID: 31641119 PMCID: PMC6805895 DOI: 10.1038/s41467-019-12270-x] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Accepted: 08/02/2019] [Indexed: 12/14/2022] Open
Abstract
The lack of integrated resources depicting the complexity of the innate immune response in cancer represents a bottleneck for high-throughput data interpretation. To address this challenge, we perform a systematic manual literature mining of molecular mechanisms governing the innate immune response in cancer and represent it as a signalling network map. The cell-type specific signalling maps of macrophages, dendritic cells, myeloid-derived suppressor cells and natural killers are constructed and integrated into a comprehensive meta map of the innate immune response in cancer. The meta-map contains 1466 chemical species as nodes connected by 1084 biochemical reactions, and it is supported by information from 820 articles. The resource helps to interpret single cell RNA-Seq data from macrophages and natural killer cells in metastatic melanoma that reveal different anti- or pro-tumor sub-populations within each cell type. Here, we report a new open source analytic platform that supports data visualisation and interpretation of tumour microenvironment activity in cancer. The complexity of the innate immune response to cancer makes interpretation of large data sets challenging. Here, the authors provide an integrated multi-scale map of signalling networks representing the different immune cells and their interactions and show its utility for data interpretation.
Collapse
Affiliation(s)
- Maria Kondratova
- Institut Curie, PSL Research University, Mines Paris Tech, Inserm, U900, 75005, Paris, France
| | - Urszula Czerwinska
- Institut Curie, PSL Research University, Mines Paris Tech, Inserm, U900, 75005, Paris, France.,Université Paris Descartes, Centre de Recherches Interdisciplinaires, Paris, France
| | - Nicolas Sompairac
- Institut Curie, PSL Research University, Mines Paris Tech, Inserm, U900, 75005, Paris, France.,Université Paris Descartes, Centre de Recherches Interdisciplinaires, Paris, France
| | | | - Vassili Soumelis
- Institut Curie, PSL Research University, Inserm, U932, 75005, Paris, France
| | - Emmanuel Barillot
- Institut Curie, PSL Research University, Mines Paris Tech, Inserm, U900, 75005, Paris, France
| | - Andrei Zinovyev
- Institut Curie, PSL Research University, Mines Paris Tech, Inserm, U900, 75005, Paris, France
| | - Inna Kuperstein
- Institut Curie, PSL Research University, Mines Paris Tech, Inserm, U900, 75005, Paris, France.
| |
Collapse
|
20
|
Lapenta C, Donati S, Spadaro F, Lattanzi L, Urbani F, Macchia I, Sestili P, Spada M, Cox MC, Belardelli F, Santini SM. Lenalidomide improves the therapeutic effect of an interferon-α-dendritic cell-based lymphoma vaccine. Cancer Immunol Immunother 2019; 68:1791-1804. [PMID: 31620858 DOI: 10.1007/s00262-019-02411-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Accepted: 10/05/2019] [Indexed: 12/25/2022]
Abstract
The perspective of combining cancer vaccines with immunomodulatory drugs is currently regarded as a highly promising approach for boosting tumor-specific T cell immunity and eradicating residual malignant cells. The efficacy of dendritic cell (DC) vaccination in combination with lenalidomide, an anticancer drug effective in several hematologic malignancies, was investigated in a follicular lymphoma (FL) model. First, we evaluated the in vitro activity of lenalidomide in modulating the immune responses of lymphocytes co-cultured with a new DC subset differentiated with IFN-α (IFN-DC) and loaded with apoptotic lymphoma cells. We next evaluated the efficacy of lenalidomide and IFN-DC-based vaccination, either alone or in combination, in hu-PBL-NOD/SCID mice bearing established human lymphoma. We found that lenalidomide reduced Treg frequency and IL-10 production in vitro, improved the formation of immune synapses of CD8 + lymphocytes with lymphoma cells and enhanced anti-lymphoma cytotoxicity. Treatment of lymphoma-bearing mice with either IFN-DC vaccination or lenalidomide led to a significant decrease in tumor growth and lymphoma cell spread. Lenalidomide treatment was shown to substantially inhibit tumor-induced neo-angiogenesis rather than to exert a direct cytotoxic effect on lymphoma cells. Notably, the combined treatment with the vaccine plus lenalidomide was more effective than either single treatment, resulting in the significant regression of established tumors and delayed tumor regrowth upon treatment discontinuation. In conclusion, our data demonstrate that IFN-DC-based vaccination plus lenalidomide exert an additive therapeutic effect in xenochimeric mice bearing established lymphoma. These results may pave the way to evaluate this combination in the clinical ground.
Collapse
Affiliation(s)
- Caterina Lapenta
- Reparto di Immunologia dei Tumori, Dipartimento di Oncologia e Medicina Molecolare, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161, Rome, Italy.
| | - Simona Donati
- Reparto di Immunologia dei Tumori, Dipartimento di Oncologia e Medicina Molecolare, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161, Rome, Italy
| | - Francesca Spadaro
- Servizio Grandi Strumentazioni e Core Facilities, Istituto Superiore di Sanità, 00161, Rome, Italy
| | - Laura Lattanzi
- Reparto di Immunologia dei Tumori, Dipartimento di Oncologia e Medicina Molecolare, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161, Rome, Italy
| | - Francesca Urbani
- Reparto di Immunologia dei Tumori, Dipartimento di Oncologia e Medicina Molecolare, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161, Rome, Italy.,Scuola di Dottorato in Biotecnologie Mediche e Medicina Traslazionale, Tor Vergata University, 00133, Rome, Italy
| | - Iole Macchia
- Reparto di Immunologia dei Tumori, Dipartimento di Oncologia e Medicina Molecolare, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161, Rome, Italy
| | - Paola Sestili
- Servizio Grandi Strumentazioni e Core Facilities, Istituto Superiore di Sanità, 00161, Rome, Italy
| | - Massimo Spada
- Centro nazionale sperimentazione e benessere animale, Istituto Superiore di Sanità, 00161, Rome, Italy
| | - Maria Christina Cox
- Unità di Ematologia, Azienda Ospedaliera Sant'Andrea, Università La Sapienza, 00189, Rome, Italy
| | - Filippo Belardelli
- Istituto di Farmacologia Traslazionale, Consiglio Nazionale delle Ricerche (CNR), 00133, Rome, Italy
| | - Stefano M Santini
- Reparto di Immunologia dei Tumori, Dipartimento di Oncologia e Medicina Molecolare, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161, Rome, Italy.
| |
Collapse
|
21
|
Vo MC, Lakshmi TJ, Jung SH, Cho D, Park HS, Chu TH, Lee HJ, Kim HJ, Kim SK, Lee JJ. Cellular immunotherapy in multiple myeloma. Korean J Intern Med 2019; 34:954-965. [PMID: 30754964 PMCID: PMC6718748 DOI: 10.3904/kjim.2018.325] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Accepted: 12/04/2018] [Indexed: 12/11/2022] Open
Abstract
In multiple myeloma (MM), the impaired function of several types of immune cells favors the tumor's escape from immune surveillance and, therefore, its growth and survival. Tremendous improvements have been made in the treatment of MM over the past decade but cellular immunotherapy using dendritic cells, natural killer cells, and genetically engineered T-cells represent a new therapeutic era. The application of these treatments is growing rapidly, based on their capacity to eradicate MM. In this review, we summarize recent progress in cellular immunotherapy for MM and its future prospects.
Collapse
Affiliation(s)
- Manh-Cuong Vo
- Research Center for Cancer Immunotherapy, Chonnam National University Hwasun Hospital, Hwasun, Korea
- Department of Hematology-Oncology, Chonnam National University Hwasun Hospital, Hwasun, Korea
| | - Thangaraj Jaya Lakshmi
- Research Center for Cancer Immunotherapy, Chonnam National University Hwasun Hospital, Hwasun, Korea
| | - Sung-Hoon Jung
- Research Center for Cancer Immunotherapy, Chonnam National University Hwasun Hospital, Hwasun, Korea
- Department of Hematology-Oncology, Chonnam National University Hwasun Hospital, Hwasun, Korea
| | - Duck Cho
- Department of Laboratory Medicine and Genetics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Hye-Seong Park
- Research Center for Cancer Immunotherapy, Chonnam National University Hwasun Hospital, Hwasun, Korea
| | - Tan-Huy Chu
- Research Center for Cancer Immunotherapy, Chonnam National University Hwasun Hospital, Hwasun, Korea
| | - Hyun-Ju Lee
- VaxCell-Bio Therapeutics, Hwasun, College of Industrial Science, Kongju National University, Yesan, Korea
| | - Hyeoung-Joon Kim
- Department of Hematology-Oncology, Chonnam National University Hwasun Hospital, Hwasun, Korea
| | - Sang-Ki Kim
- Department of Companion and Laboratory Animal Science, College of Industrial Science, Kongju National University, Yesan, Korea
| | - Je-Jung Lee
- Research Center for Cancer Immunotherapy, Chonnam National University Hwasun Hospital, Hwasun, Korea
- Department of Hematology-Oncology, Chonnam National University Hwasun Hospital, Hwasun, Korea
- VaxCell-Bio Therapeutics, Hwasun, College of Industrial Science, Kongju National University, Yesan, Korea
- Correspondence to Je-Jung Lee, M.D. Department of Hematology-Oncology, Chonnam National University Hwasun Hospital, 322 Seoyang-ro, Hwasun 58128, Korea Tel: +82-61-379-7638, Fax: +82-61-379-7628, E-mail:
| |
Collapse
|
22
|
Sprooten J, Ceusters J, Coosemans A, Agostinis P, De Vleeschouwer S, Zitvogel L, Kroemer G, Galluzzi L, Garg AD. Trial watch: dendritic cell vaccination for cancer immunotherapy. Oncoimmunology 2019; 8:e1638212. [PMID: 31646087 PMCID: PMC6791419 DOI: 10.1080/2162402x.2019.1638212] [Citation(s) in RCA: 100] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Accepted: 06/26/2019] [Indexed: 12/12/2022] Open
Abstract
Dendritic- cells (DCs) have received considerable attention as potential targets for the development of anticancer vaccines. DC-based anticancer vaccination relies on patient-derived DCs pulsed with a source of tumor-associated antigens (TAAs) in the context of standardized maturation-cocktails, followed by their reinfusion. Extensive evidence has confirmed that DC-based vaccines can generate TAA-specific, cytotoxic T cells. Nonetheless, clinical efficacy of DC-based vaccines remains suboptimal, reflecting the widespread immunosuppression within tumors. Thus, clinical interest is being refocused on DC-based vaccines as combinatorial partners for T cell-targeting immunotherapies. Here, we summarize the most recent preclinical/clinical development of anticancer DC vaccination and discuss future perspectives for DC-based vaccines in immuno-oncology.
Collapse
Affiliation(s)
- Jenny Sprooten
- Cell Death Research & Therapy (CDRT) unit, Department of Cellular & Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Jolien Ceusters
- Department of Oncology, Laboratory of Tumor Immunology and Immunotherapy, ImmunOvar Research Group, KU Leuven, Leuven Cancer Institute, Leuven, Belgium
| | - An Coosemans
- Department of Oncology, Laboratory of Tumor Immunology and Immunotherapy, ImmunOvar Research Group, KU Leuven, Leuven Cancer Institute, Leuven, Belgium
- Department of Gynecology and Obstetrics, UZ Leuven, Leuven, Belgium
| | - Patrizia Agostinis
- Cell Death Research & Therapy (CDRT) unit, Department of Cellular & Molecular Medicine, KU Leuven, Leuven, Belgium
- Center for Cancer Biology (CCB), VIB, Leuven, Belgium
| | - Steven De Vleeschouwer
- Research Group Experimental Neurosurgery and Neuroanatomy, KU Leuven, Leuven, Belgium
- Department of Neurosurgery, UZ Leuven, Leuven, Belgium
| | - Laurence Zitvogel
- Gustave Roussy Comprehensive Cancer Institute, Villejuif, France
- INSERM, Villejuif, France
- Center of Clinical Investigations in Biotherapies of Cancer (CICBT) 1428, Villejuif, France
- Université Paris Sud/Paris XI, Le Kremlin-Bicêtre, France
| | - Guido Kroemer
- Equipe labellisée par la Ligue contre le cancer, Université de Paris, Sorbonne Université, INSERM U1138, Centre de Recherche des Cordeliers, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Comprehensive Cancer Institute, Villejuif, France
- Pôle de Biologie, Hôpital Européen Georges Pompidou, AP-HP, Paris, France
- Suzhou Institute for Systems Medicine, Chinese Academy of Sciences, Suzhou, China
- Department of Women’s and Children’s Health, Karolinska University Hospital, Stockholm, Sweden
| | - Lorenzo Galluzzi
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA
- Sandra and Edward Meyer Cancer Center, New York, NY, USA
- Department of Dermatology, Yale School of Medicine, New Haven, CT, USA
- Université de Paris Descartes, Paris, France
| | - Abhishek D. Garg
- Cell Death Research & Therapy (CDRT) unit, Department of Cellular & Molecular Medicine, KU Leuven, Leuven, Belgium
| |
Collapse
|
23
|
Shinde P, Melinkeri S, Santra MK, Kale V, Limaye L. Autologous Hematopoietic Stem Cells Are a Preferred Source to Generate Dendritic Cells for Immunotherapy in Multiple Myeloma Patients. Front Immunol 2019; 10:1079. [PMID: 31164886 PMCID: PMC6536579 DOI: 10.3389/fimmu.2019.01079] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Accepted: 04/29/2019] [Indexed: 11/13/2022] Open
Abstract
In multiple myeloma (MM), dendritic cells (DCs), and their precursors are prone to malignant cell-mediated regulation of function leading to low efficacy of DC vaccine. DCs taken directly from MM patient's body or derived from monocytes are fewer in numbers and are also dysfunctional. Here, we investigated the functionality of Hematopoietic stem cell-derived DCs (SC-DCs) from MM patients. Mature-MM-SC-DCs showed all essential functions like antigen uptake, allogenic T cells simulation and migration comparable to those derived from healthy donor (HD) samples. A comparison of Mo-DCs and SC-DCs obtained from the same MM patients' samples revealed that the expression of IL-6 was higher in the precursors of Mo-DCs leading to their impaired migration. In addition, expression of CCR7 which is responsible for DCs migration was found to be lower in MM-Mo-DCs. The chromatin permissiveness as observed by H3K4me3 histone modification at the Ccr7 promoter in MM-Mo-DCs was significantly lower than those in MM-SC-DCs. Levels of Zbtb46- a hall mark DC transcription factor mRNA was also found to be reduced in MM-Mo-DCs. Cytotoxic T cells generated from MM-SC-DCs from autologous naïve T cells exhibited reduced antitumor activity because the T cells were exhausted. Blocking of CTLA-4 on autologous T cells could partially restore T cell proliferation and activation. Thus, a combination of MM-SC-DC vaccine and anti-CTLA-4 antibody may serve as a better candidate for immunotherapy of MM. This study has implications in increasing the efficacy of cancer immunotherapy in MM.
Collapse
Affiliation(s)
- Prajakta Shinde
- National Centre for Cell Science, Savitribai Phule Pune University Campus, Pune, India
| | - Sameer Melinkeri
- Blood and Marrow Transplant Unit, Deenanath Mangeshkar Hospital, Pune, India
| | - Manas Kumar Santra
- National Centre for Cell Science, Savitribai Phule Pune University Campus, Pune, India
| | - Vaijayanti Kale
- National Centre for Cell Science, Savitribai Phule Pune University Campus, Pune, India
| | - Lalita Limaye
- National Centre for Cell Science, Savitribai Phule Pune University Campus, Pune, India
| |
Collapse
|
24
|
Unleashing Tumour-Dendritic Cells to Fight Cancer by Tackling Their Three A's: Abundance, Activation and Antigen-Delivery. Cancers (Basel) 2019; 11:cancers11050670. [PMID: 31091774 PMCID: PMC6562396 DOI: 10.3390/cancers11050670] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Revised: 04/22/2019] [Accepted: 05/10/2019] [Indexed: 12/24/2022] Open
Abstract
Recent advances in cancer immunotherapy have mainly focused on re-activating T-cell responses against cancer cells. However, both priming and activation of effector T-cell responses against cancer-specific antigens require cross-talk with dendritic cells (DCs), which are responsible for the capturing, processing and presentation of tumour-(neo)antigens to T cells. DCs consequently constitute an essential target in efforts to generate therapeutic immunity against cancer. This review will discuss recent research that is unlocking the cancer-fighting potential of tumour-infiltrating DCs. First, the complexity of DCs in the tumour microenvironment regarding the different subsets and the difficulty of translating mouse data into equivalent human data will be briefly touched upon. Mainly, possible solutions to problems currently faced in DC-based cancer treatments will be discussed, including their infiltration into tumours, activation strategies, and antigen delivery methods. In this way, we hope to put together a broad picture of potential synergistic therapies that could be implemented to harness the full capacity of tumour-infiltrating DCs to stimulate anti-tumour immune responses in patients.
Collapse
|
25
|
Huber A, Dammeijer F, Aerts JGJV, Vroman H. Current State of Dendritic Cell-Based Immunotherapy: Opportunities for in vitro Antigen Loading of Different DC Subsets? Front Immunol 2018; 9:2804. [PMID: 30559743 PMCID: PMC6287551 DOI: 10.3389/fimmu.2018.02804] [Citation(s) in RCA: 79] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Accepted: 11/14/2018] [Indexed: 12/12/2022] Open
Abstract
Dendritic cell (DC) based cancer immunotherapy aims at the activation of the immune system, and in particular tumor-specific cytotoxic T lymphocytes (CTLs) to eradicate the tumor. DCs represent a heterogeneous cell population, including conventional DCs (cDCs), consisting of cDC1s, cDC2s, plasmacytoid DCs (pDCs), and monocyte-derived DCs (moDCs). These DC subsets differ both in ontogeny and functional properties, such as the capacity to induce CD4+ and CD8+ T-cell activation. MoDCs are most frequently used for vaccination purposes, based on technical aspects such as availability and in vitro expansion. However, whether moDCs are superior over other DC subsets in inducing anti-tumor immune responses, is unknown, and likely depends on tumor type and composition of the tumor microenvironment. In this review, we discuss cellular aspects essential for DC vaccination efficacy, and the most recent findings on different DC subsets that could be used for DC-based cancer immunotherapy. This can prove valuable for the future design of more effective DC vaccines by choosing different DC subsets, and sheds light on the working mechanism of DC immunotherapy.
Collapse
Affiliation(s)
- Anne Huber
- Department of Pulmonary Medicine, Erasmus Medical Center, Rotterdam, Netherlands
| | - Floris Dammeijer
- Department of Pulmonary Medicine, Erasmus Medical Center, Rotterdam, Netherlands
- Erasmus Cancer Institute, Erasmus Medical Center, Rotterdam, Netherlands
| | - Joachim G. J. V. Aerts
- Department of Pulmonary Medicine, Erasmus Medical Center, Rotterdam, Netherlands
- Erasmus Cancer Institute, Erasmus Medical Center, Rotterdam, Netherlands
| | - Heleen Vroman
- Department of Pulmonary Medicine, Erasmus Medical Center, Rotterdam, Netherlands
- Erasmus Cancer Institute, Erasmus Medical Center, Rotterdam, Netherlands
| |
Collapse
|
26
|
Fujiwara Y, Sun Y, Torphy RJ, He J, Yanaga K, Edil BH, Schulick RD, Zhu Y. Pomalidomide Inhibits PD-L1 Induction to Promote Antitumor Immunity. Cancer Res 2018; 78:6655-6665. [PMID: 30315115 DOI: 10.1158/0008-5472.can-18-1781] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Revised: 09/11/2018] [Accepted: 10/08/2018] [Indexed: 11/16/2022]
Abstract
: Thalidomide-like drugs have been approved for the treatment of human multiple myeloma, with their direct antitumor effects and immunomodulatory functions well documented. However, the exact molecular mechanisms that govern these effects remain unclear. Here we demonstrate that pomalidomide promotes immune response by inhibiting expression of PD-L1. Pomalidomide inhibited PD-L1 expression on tumor cells to promote CTL activity in vitro and suppressed PD-L1 upregulation on antigen-presenting cells to prevent peptide-induced T-cell tolerance. Knockout of PD-L1 on tumor cells or in mice completely eliminated the immunomodulatory effect of pomalidomide. Furthermore, pomalidomide synergized with other immunotherapies to improve anticancer therapy. Taken together, this study identifies a new mechanism for the immunomodulatory functions of pomalidomide in cancer therapy. These results also offer a clinical approach for blocking PD-L1 induction and potentially promoting antitumor immunity. SIGNIFICANCE: These findings report that the immunomodulatory drug pomalidomide, widely used to treat myeloma and other cancers, enhances antitumor immunity by inhibiting PD-1/PD-L1 expression.
Collapse
Affiliation(s)
- Yuki Fujiwara
- Department of Surgery, University of Colorado Anschutz Medical Campus, Aurora, Colorado.,Department of Surgery, The Jikei University School of Medicine, Minato-ku, Tokyo, Japan
| | - Yi Sun
- Department of Surgery, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Robert J Torphy
- Department of Surgery, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Jiadai He
- Computer Science and Engineering Program, The Chinese University of Hong Kong - Shenzhen Campus, Shenzhen, Guangdong, China
| | - Katsuhiko Yanaga
- Department of Surgery, The Jikei University School of Medicine, Minato-ku, Tokyo, Japan
| | - Barish H Edil
- Department of Surgery, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Richard D Schulick
- Department of Surgery, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Yuwen Zhu
- Department of Surgery, University of Colorado Anschutz Medical Campus, Aurora, Colorado.
| |
Collapse
|
27
|
Vo MC, Yang S, Jung SH, Chu TH, Lee HJ, Lakshmi TJ, Park HS, Kim HJ, Lee JJ. Synergistic Antimyeloma Activity of Dendritic Cells and Pomalidomide in a Murine Myeloma Model. Front Immunol 2018; 9:1798. [PMID: 30123221 PMCID: PMC6085413 DOI: 10.3389/fimmu.2018.01798] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2018] [Accepted: 07/20/2018] [Indexed: 01/14/2023] Open
Abstract
We have previously shown that immunization with tumor antigen-loaded dendritic cells (DCs) and the immunomodulating drug, lenalidomide, synergistically potentiates the enhancing antitumor immunity in a myeloma mouse model. In this study, we investigated the immunogenicity of DCs combined with pomalidomide and dexamethasone in a myeloma mouse model. MOPC-315 cells were injected subcutaneously to establish myeloma-bearing mice. Four test groups were used to mimic clinical protocol: (1) PBS control, (2) DCs, (3) pomalidomide + dexamethasone, and (4) DCs + pomalidomide + dexamethasone. The combination of DCs plus pomalidomide and dexamethasone displayed greater inhibition of tumor growth compared to the other groups. This effect was closely related with reduced numbers of immune suppressor cells including myeloid-derived suppressor cells, M2 macrophages, and regulatory T cells, with the induction of immune effector cells such as CD4+ and CD8+ T cells, memory T cells, natural killer (NK) cells, and M1 macrophages, and with the activation of T lymphocytes and NK cells in the spleen. Moreover, the level of the immunosuppressive factor vascular endothelial growth factor was significantly reduced in the tumor microenvironment. The collective findings in the murine myeloma model suggest that tumor antigen-loaded DCs combined with pomalidomide and dexamethasone synergistically enhance antitumor immunity by skewing the immune-suppressive status toward an immune-supportive status.
Collapse
Affiliation(s)
- Manh-Cuong Vo
- Research Center for Cancer Immunotherapy, Chonnam National University Hwasun Hospital, Hwasun, South Korea.,Department of Hematology-Oncology, Chonnam National University Hwasun Hospital, Hwasun, South Korea
| | - Seoyun Yang
- Research Center for Cancer Immunotherapy, Chonnam National University Hwasun Hospital, Hwasun, South Korea
| | - Sung-Hoon Jung
- Research Center for Cancer Immunotherapy, Chonnam National University Hwasun Hospital, Hwasun, South Korea.,Department of Hematology-Oncology, Chonnam National University Hwasun Hospital, Hwasun, South Korea
| | - Tan-Huy Chu
- Research Center for Cancer Immunotherapy, Chonnam National University Hwasun Hospital, Hwasun, South Korea
| | - Hyun-Ju Lee
- Vaxcell-Bio Therapeutics, Hwasun, South Korea
| | - Thangaraj Jaya Lakshmi
- Research Center for Cancer Immunotherapy, Chonnam National University Hwasun Hospital, Hwasun, South Korea
| | - Hye-Seong Park
- Research Center for Cancer Immunotherapy, Chonnam National University Hwasun Hospital, Hwasun, South Korea
| | - Hyeoung-Joon Kim
- Department of Hematology-Oncology, Chonnam National University Hwasun Hospital, Hwasun, South Korea
| | - Je-Jung Lee
- Research Center for Cancer Immunotherapy, Chonnam National University Hwasun Hospital, Hwasun, South Korea.,Department of Hematology-Oncology, Chonnam National University Hwasun Hospital, Hwasun, South Korea.,Vaxcell-Bio Therapeutics, Hwasun, South Korea
| |
Collapse
|
28
|
Vo MC, Jung SH, Chu TH, Lee HJ, Lakshmi TJ, Park HS, Kim HJ, Rhee JH, Lee JJ. Lenalidomide and Programmed Death-1 Blockade Synergistically Enhances the Effects of Dendritic Cell Vaccination in a Model of Murine Myeloma. Front Immunol 2018; 9:1370. [PMID: 29967612 PMCID: PMC6015916 DOI: 10.3389/fimmu.2018.01370] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Accepted: 06/01/2018] [Indexed: 01/22/2023] Open
Abstract
The therapeutic efficacy of dendritic cell (DC)-based immunotherapy may be potentiated in combination with other anticancer therapies that enhance DC function by modulating immune responses and the tumor microenvironment. In this study, we investigated the efficacy of DC vaccination in combination with lenalidomide and programmed death (PD)-1 blockade in a model of murine myeloma. MOPC-315 cell lines were injected subcutaneously to establish myeloma-bearing mice and the following five test groups were established: PBS control, DCs, DCs + lenalidomide, DCs + PD-1 blockade, and DCs + lenalidomide + PD-1 blockade. The combination of DCs plus lenalidomide and PD-1 blockade more potently inhibited tumor growth compared to the other groups. This effect was associated with a reduction in immune suppressor cells (such as myeloid-derived suppressor cells, M2 macrophages, and regulatory T cells) and an increase in immune effector cells [such as CD4+ and CD8+ T cells, natural killer (NK) cells, and M1 macrophages] in the spleen. Functional activities of cytotoxic T lymphocytes and NK cells were also enhanced by the triple combination. Levels of immunosuppressive cytokines, such as TGF-β and IL-10, were significantly reduced in the tumor microenvironment. These findings suggest that the combination of DCs plus lenalidomide and PD-1 blockade synergistically establishes a robust anti-myeloma immunity through a two-way mechanism, which inhibits immunosuppressive cells while activating effector cells with superior polarization of the Th1/Th2 balance in favor of the tumor immune response. This result should provide an experimental ground for incorporating check point inhibitors to existing immunotherapeutic modalities against multiple myeloma.
Collapse
Affiliation(s)
- Manh-Cuong Vo
- Research Center for Cancer Immunotherapy, Chonnam National University Hwasun Hospital, Hwasun, South Korea.,Department of Hematology-Oncology, Chonnam National University Hwasun Hospital and Chonnam National University Medical School, Hwasun, South Korea
| | - Sung-Hoon Jung
- Research Center for Cancer Immunotherapy, Chonnam National University Hwasun Hospital, Hwasun, South Korea.,Department of Hematology-Oncology, Chonnam National University Hwasun Hospital and Chonnam National University Medical School, Hwasun, South Korea
| | - Tan-Huy Chu
- Research Center for Cancer Immunotherapy, Chonnam National University Hwasun Hospital, Hwasun, South Korea
| | - Hyun-Ju Lee
- Research Institute, VaxCell-Bio Therapeutics, Hwasun, South Korea
| | - Thangaraj Jaya Lakshmi
- Research Center for Cancer Immunotherapy, Chonnam National University Hwasun Hospital, Hwasun, South Korea
| | - Hye-Seong Park
- Research Center for Cancer Immunotherapy, Chonnam National University Hwasun Hospital, Hwasun, South Korea
| | - Hyeoung-Joon Kim
- Department of Hematology-Oncology, Chonnam National University Hwasun Hospital and Chonnam National University Medical School, Hwasun, South Korea
| | - Joon Haeng Rhee
- Research Institute, VaxCell-Bio Therapeutics, Hwasun, South Korea.,Department of Microbiology and Clinical Vaccine R&D Center, Chonnam National University Medical School, Hwasun, South Korea
| | - Je-Jung Lee
- Research Center for Cancer Immunotherapy, Chonnam National University Hwasun Hospital, Hwasun, South Korea.,Department of Hematology-Oncology, Chonnam National University Hwasun Hospital and Chonnam National University Medical School, Hwasun, South Korea.,Research Institute, VaxCell-Bio Therapeutics, Hwasun, South Korea
| |
Collapse
|
29
|
López-Relaño J, Martín-Adrados B, Real-Arévalo I, Lozano-Bartolomé J, Abós B, Sánchez-Ramón S, Alonso B, Gómez Del Moral M, Martínez-Naves E. Monocyte-Derived Dendritic Cells Differentiated in the Presence of Lenalidomide Display a Semi-Mature Phenotype, Enhanced Phagocytic Capacity, and Th1 Polarization Capability. Front Immunol 2018; 9:1328. [PMID: 29951065 PMCID: PMC6008535 DOI: 10.3389/fimmu.2018.01328] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Accepted: 05/28/2018] [Indexed: 11/16/2022] Open
Abstract
Lenalidomide is an analog of thalidomide, with potent anticancer activity demonstrated in several hematological malignancies. It has immunomodulatory properties, being able to enhance the activation of different types of immune cells, which results in antitumor activities. Dendritic cells (DCs) are pivotal in the immune response, and different immunotherapeutic approaches targeting these cells are being developed. Since little is known about the effect of lenalidomide on DCs, the goal of the present work was to investigate the phenotype and function of human monocyte-derived DCs differentiated in the presence of lenalidomide (L-DCs). Our results showed that L-DCs display a unique phenotype, with increased cell surface expression of some maturation markers such as CD1d, CD83, CD86, and HLA-DR. This phenotype correlates with a lower expression of the E3 ubiquitin-ligase MARCH-I in L-DCs, upregulating the cell surface expression of CD86 and HLA-DR. In addition, immature L-DCs express higher amounts of DC-SIGN on the cell surface than control immature DCs. After LPS stimulation, production of IL-6 and TNF-α was severely decreased, whereas IL-12 and IL-10 secretion was dramatically upregulated in L-DCs, compared to that in the controls. Functionally, L-DCs are more effectively recognized by NKT cells in cytotoxicity experiments. Furthermore, L-DCs display higher opsonin-independent antigen uptake capability than control DCs. Mixed lymphocyte reaction experiments showed that L-DCs could stimulate naïve CD4 T-cells, polarizing them toward a predominant Th1 phenotype. In summary, DCs derived from monocytes in the presence of lenalidomide present a semi-mature phenotype, increased phagocytic capacity, reduced production of proinflammatory cytokines, and the ability to polarize T-cells toward predominant Th1-type responses; these are qualities that might be useful in the development of new immunotherapeutic treatments.
Collapse
Affiliation(s)
- Juan López-Relaño
- Departamento de Inmunología, Facultad de Medicina, Universidad Complutense, Madrid, Spain.,12 de Octubre Health Research Institute (imas12), Madrid, Spain
| | - Beatriz Martín-Adrados
- Departamento de Inmunología, Facultad de Medicina, Universidad Complutense, Madrid, Spain.,12 de Octubre Health Research Institute (imas12), Madrid, Spain
| | - Irene Real-Arévalo
- Departamento de Inmunología, Facultad de Medicina, Universidad Complutense, Madrid, Spain.,12 de Octubre Health Research Institute (imas12), Madrid, Spain
| | - Javier Lozano-Bartolomé
- Departamento de Inmunología, Facultad de Medicina, Universidad Complutense, Madrid, Spain.,12 de Octubre Health Research Institute (imas12), Madrid, Spain
| | - Beatriz Abós
- Departamento de Inmunología, Facultad de Medicina, Universidad Complutense, Madrid, Spain.,12 de Octubre Health Research Institute (imas12), Madrid, Spain
| | | | | | - Manuel Gómez Del Moral
- 12 de Octubre Health Research Institute (imas12), Madrid, Spain.,Departamento de Biología Celular, Facultad de Medicina, Universidad Complutense, Madrid, Spain
| | - Eduardo Martínez-Naves
- Departamento de Inmunología, Facultad de Medicina, Universidad Complutense, Madrid, Spain.,12 de Octubre Health Research Institute (imas12), Madrid, Spain
| |
Collapse
|
30
|
Vo MC, Nguyen-Pham TN, Lee HJ, Jung SH, Choi NR, Hoang MD, Kim HJ, Lee JJ. Chaetocin enhances dendritic cell function via the induction of heat shock protein and cancer testis antigens in myeloma cells. Oncotarget 2018; 8:46047-46056. [PMID: 28512265 PMCID: PMC5542247 DOI: 10.18632/oncotarget.17517] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2016] [Accepted: 03/24/2017] [Indexed: 12/12/2022] Open
Abstract
Dendritic cells (DC)-based vaccines are considered useful in cancer immuno-therapy, and the interactions of DC and dying tumor cells are important and promising for cancer immunotherapy. We investigated whether chaetocin could be used to induce death of myeloma cells, for loading onto DCs can affect DCs function. In this study, we show that the dying myeloma cells treated with chaetocin resulted in the induction of heat shock protein (HSP) 90, which was inhibited by antioxidant N-acetyl cysteine, and showed an increase in the expression of MAGE-A3 and MAGE-C1/CT7. DCs loaded with chaetocin-treated dying myeloma cells produced low levels of IL-10 and enhanced the cross presentation of DCs. Additionally, these DCs most potently inhibited regulatory T cells, induced Th1 polarization and activated myeloma-specific cytotoxic T lymphocytes compared with DCs loaded with UVB-irradiated dying myeloma cells. These results suggest that the pretreatment of myeloma cells with chaetocin can enhance DC function through the up-regulation of HSP90 and cancer testis antigens in dying myeloma cells and can potently induce the Th1 polarization of DCs and myeloma-specific cytotoxic T lymphocytes.
Collapse
Affiliation(s)
- Manh-Cuong Vo
- Research Center for Cancer Immunotherapy, Chonnam National University Hwasun Hospital, Hwasun, Jeollanamdo, Republic of Korea
| | - Thanh-Nhan Nguyen-Pham
- Research Center for Cancer Immunotherapy, Chonnam National University Hwasun Hospital, Hwasun, Jeollanamdo, Republic of Korea.,Department of Hematology-Oncology, Chonnam National University Hwasun Hospital, Hwasun, Jeollanamdo, Republic of Korea
| | - Hyun-Ju Lee
- Research Center for Cancer Immunotherapy, Chonnam National University Hwasun Hospital, Hwasun, Jeollanamdo, Republic of Korea
| | - Sung-Hoon Jung
- Research Center for Cancer Immunotherapy, Chonnam National University Hwasun Hospital, Hwasun, Jeollanamdo, Republic of Korea.,Department of Hematology-Oncology, Chonnam National University Hwasun Hospital, Hwasun, Jeollanamdo, Republic of Korea
| | - Nu-Ri Choi
- Research Center for Cancer Immunotherapy, Chonnam National University Hwasun Hospital, Hwasun, Jeollanamdo, Republic of Korea
| | - My-Dung Hoang
- Research Center for Cancer Immunotherapy, Chonnam National University Hwasun Hospital, Hwasun, Jeollanamdo, Republic of Korea
| | - Hyeoung-Joon Kim
- Department of Hematology-Oncology, Chonnam National University Hwasun Hospital, Hwasun, Jeollanamdo, Republic of Korea
| | - Je-Jung Lee
- Research Center for Cancer Immunotherapy, Chonnam National University Hwasun Hospital, Hwasun, Jeollanamdo, Republic of Korea.,Department of Hematology-Oncology, Chonnam National University Hwasun Hospital, Hwasun, Jeollanamdo, Republic of Korea
| |
Collapse
|
31
|
Maeda Y, Takahashi H, Nakai N, Yanagita T, Ando N, Okubo T, Saito K, Shiga K, Hirokawa T, Hara M, Ishiguro H, Matsuo Y, Takiguchi S. Apigenin induces apoptosis by suppressing Bcl-xl and Mcl-1 simultaneously via signal transducer and activator of transcription 3 signaling in colon cancer. Int J Oncol 2018; 52:1661-1673. [DOI: 10.3892/ijo.2018.4308] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2017] [Accepted: 01/30/2018] [Indexed: 11/06/2022] Open
Affiliation(s)
- Yuzo Maeda
- Department of Gastroenterological Surgery, Nagoya City University Graduate School of Medical Sciences, Nagoya, Aichi 467-8602, Japan
| | - Hiroki Takahashi
- Department of Gastroenterological Surgery, Nagoya City University Graduate School of Medical Sciences, Nagoya, Aichi 467-8602, Japan
| | - Nozomu Nakai
- Department of Gastroenterological Surgery, Nagoya City University Graduate School of Medical Sciences, Nagoya, Aichi 467-8602, Japan
| | - Takeshi Yanagita
- Department of Gastroenterological Surgery, Nagoya City University Graduate School of Medical Sciences, Nagoya, Aichi 467-8602, Japan
| | - Nanako Ando
- Department of Gastroenterological Surgery, Nagoya City University Graduate School of Medical Sciences, Nagoya, Aichi 467-8602, Japan
| | - Tomotaka Okubo
- Department of Gastroenterological Surgery, Nagoya City University Graduate School of Medical Sciences, Nagoya, Aichi 467-8602, Japan
| | - Kenta Saito
- Department of Gastroenterological Surgery, Nagoya City University Graduate School of Medical Sciences, Nagoya, Aichi 467-8602, Japan
| | - Kazuyoshi Shiga
- Department of Gastroenterological Surgery, Nagoya City University Graduate School of Medical Sciences, Nagoya, Aichi 467-8602, Japan
| | - Takahisa Hirokawa
- Department of Gastroenterological Surgery, Nagoya City University Graduate School of Medical Sciences, Nagoya, Aichi 467-8602, Japan
| | - Masayasu Hara
- Department of Gastroenterological Surgery, Nagoya City University Graduate School of Medical Sciences, Nagoya, Aichi 467-8602, Japan
| | - Hideyuki Ishiguro
- Department of Gastroenterological Surgery, Nagoya City University Graduate School of Medical Sciences, Nagoya, Aichi 467-8602, Japan
| | - Yoichi Matsuo
- Department of Gastroenterological Surgery, Nagoya City University Graduate School of Medical Sciences, Nagoya, Aichi 467-8602, Japan
| | - Shuji Takiguchi
- Department of Gastroenterological Surgery, Nagoya City University Graduate School of Medical Sciences, Nagoya, Aichi 467-8602, Japan
| |
Collapse
|
32
|
Richter J, Kretz AL, Lemke J, Fauler M, Werner JU, Paschke S, Leithäuser F, Henne-Bruns D, Hillenbrand A, Knippschild U. CK1α overexpression correlates with poor survival in colorectal cancer. BMC Cancer 2018; 18:140. [PMID: 29409464 PMCID: PMC5801892 DOI: 10.1186/s12885-018-4019-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2016] [Accepted: 01/22/2018] [Indexed: 12/20/2022] Open
Abstract
Background Colorectal cancer (CRC) is the fourth leading cause of cancer related deaths worldwide and prognosis in advanced tumor stage still remains poor. Since CK1 isoforms have been reported to be deregulated in several tumor entities CK1 has emerged as a novel drug target in cancer therapy. In this study we set out to investigate whether CK1α might have the potential to serve as prognostic marker. Methods CK1α RNA and protein expression levels in healthy and tumor tissue of CRC patients were analyzed using quantitative real-time PCR and Western Blot analysis, respectively. Prognostic relevance was investigated by correlating obtained CK1α expression levels with patients’ survival rate generating Kaplan-Meier survival plots. Results It could be shown that CK1α is overexpressed in colorectal tumor tissue compared to normal tissue and CK1α overexpression in tumor tissue correlates with poor survival in CRC patients. Results become more significant when only considering patients with high-grade tumors, as well as patients assigned to UICC II and UICC III stage. Furthermore, Cox regression analysis revealed that CK1α is an independent prognostic factor. In addition, tumors expressing decreased levels of the kinase reveal positive effects on overall survival when localized in the right colon compared to those in the left side. Conclusion In summary, this study provides evidence for the first time that CK1α RNA levels might serve as prognostic marker for CRC. Electronic supplementary material The online version of this article (10.1186/s12885-018-4019-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Julia Richter
- Department of General and Visceral Surgery, Ulm University Hospital, Albert-Einstein-Allee 23, 89081, Ulm, Germany
| | - Anna-Laura Kretz
- Department of General and Visceral Surgery, Ulm University Hospital, Albert-Einstein-Allee 23, 89081, Ulm, Germany
| | - Johannes Lemke
- Department of General and Visceral Surgery, Ulm University Hospital, Albert-Einstein-Allee 23, 89081, Ulm, Germany
| | - Michael Fauler
- Ulm University, Institute of General Physiology, Albert-Einstein-Allee 11, 89081, Ulm, Germany
| | - Jens-Uwe Werner
- Department of General and Visceral Surgery, Ulm University Hospital, Albert-Einstein-Allee 23, 89081, Ulm, Germany
| | - Stephan Paschke
- Department of General and Visceral Surgery, Ulm University Hospital, Albert-Einstein-Allee 23, 89081, Ulm, Germany
| | - Frank Leithäuser
- Department of Pathology, Ulm University Hospital, Albert-Einstein-Allee 23, 89081, Ulm, Germany
| | - Doris Henne-Bruns
- Department of General and Visceral Surgery, Ulm University Hospital, Albert-Einstein-Allee 23, 89081, Ulm, Germany
| | - Andreas Hillenbrand
- Department of General and Visceral Surgery, Ulm University Hospital, Albert-Einstein-Allee 23, 89081, Ulm, Germany
| | - Uwe Knippschild
- Department of General and Visceral Surgery, Ulm University Hospital, Albert-Einstein-Allee 23, 89081, Ulm, Germany.
| |
Collapse
|