1
|
Rajkumari S, Singh J, Agrawal U, Agrawal S. Myeloid-derived suppressor cells in cancer: Current knowledge and future perspectives. Int Immunopharmacol 2024; 142:112949. [PMID: 39236460 DOI: 10.1016/j.intimp.2024.112949] [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: 05/30/2024] [Revised: 08/12/2024] [Accepted: 08/13/2024] [Indexed: 09/07/2024]
Abstract
MDSCs (myeloid-derived suppressor cells) are crucial for immune system evasion in cancer. They accumulate in peripheral blood and tumor microenvironment, suppressing immune cells like T-cells, natural killer cells and dendritic cells. They promote tumor angiogenesis and metastasis by secreting cytokines and growth factors and contribute to a tumor-promoting environment. The accumulation of MDSCs in cancer patients has been linked to poor prognosis and resistance to various cancer therapies. Targeting MDSCs and their immunosuppressive mechanisms may improve treatment outcomes and enhance immune surveillance by developing drugs that inhibit MDSC function, by preventing their accumulation and by disrupting the tumor-promoting environment. This review presents a detailed overview of the MDSC research in cancer with regulation of their development and function. The relevance of MDSC as a prognostic and predictive biomarker in different types of cancers, along with recent advancements on the therapeutic approaches to target MDSCs are discussed in detail.
Collapse
Affiliation(s)
- Sunanda Rajkumari
- ICMR National Institute of Medical Statistics, Ansari Nagar, New Delhi 110029, India
| | - Jaspreet Singh
- ICMR National Institute of Pathology, Safdarjung Hospital Campus, Ansari Nagar, New Delhi 110029, India
| | - Usha Agrawal
- Asian Institute of Public Health University (AIPH) University, 1001 Haridamada, Jatani, Near IIT Bhubaneswar, Bhubaneswar 751002, India
| | - Sandeep Agrawal
- Discovery Research Division, Indian Council of Medical Research, Ansari Nagar, New Delhi 110029, India.
| |
Collapse
|
2
|
Valentina B, Jessica B, Michelle P, Nadia W, Roland H, Matthias D, Jo C, Guillaume M. CD38 as theranostic target in oncology. J Transl Med 2024; 22:998. [PMID: 39501292 PMCID: PMC11539646 DOI: 10.1186/s12967-024-05768-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Accepted: 10/15/2024] [Indexed: 11/08/2024] Open
Abstract
CD38 is a multifunctional transmembrane glycoprotein found in multiple tissues and overexpressed in many cancer cells, notably in hematological malignancies such as leukemia and multiple myeloma (MM). Therefore, targeting CD38 remains an attractive strategy for cancer treatment in hematological malignancies as well as in solid tumors. It plays a critical role in the progression of these diseases through its ADP-ribosyl cyclase and cADPR-hydrolase activities. Its importance has led to the development of various anti-CD38 monoclonal antibodies (mAbs), including daratumumab and isatuximab, approved for MM treatment. These mAbs exert their anti-tumor effects through Fc-dependent immune mechanisms and immunomodulation, enhancing T-cell and NK-cell-mediated responses. However, resistance mechanisms arise during the treatment with daratumumab, creating the necessity for new therapies. This review explains current knowledge about the role of CD38 as a target in oncology and aims to delineate the use of single domain antibodies (sdAbs) as innovative theranostic tools in nuclear medicine. For diagnostic purposes, PET radionuclides like 68 Ga, 64Cu, and SPECT radionuclides like 99mTc and 111In, are commonly used. Significant progress has been made in anti-CD38 radioligand therapy (RLT), with anti-CD38 antibodies providing insights into tumor biology and treatment efficacy. In terms of therapy, RLT is a promising approach that offers precise targeting of malignant cells while minimizing exposure to healthy tissue. This involves the use of radionuclides emitting α particles, like 225Ac, 212Pb or 211At, and β--particles like 90Y, 131I, or 177Lu, to exert cytotoxic effects. Derived from Camelidae heavy chain antibodies, sdAbs offer advantages over conventional mAbs such as small size, high stability, specificity, and ability to recognize hidden epitopes. CD38-specific sdAbs, such as sdAb 2F8, characterized by our laboratory, showing excellent tumor targeting and their engineered constructs, such as biparatopic antibodies and chimeric antibodies, represent a new generation of theranostic agents for diagnosis and treatment CD38-expressing malignancies.
Collapse
Affiliation(s)
- Bocuzzi Valentina
- Laboratory of Hematology, GIGA I3, University of Liège, Liège, Belgium
- Center for Protein Engineering, University of Liège, Liège, Belgium
| | - Bridoux Jessica
- Molecular Imaging and Therapy Laboratory (MITH), Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | | | - Withofs Nadia
- Department of Nuclear Medicine and Oncology, CHU de Liège, Liège, Belgium
| | - Hustinx Roland
- Department of Nuclear Medicine, CHU de Liège, Liège, Belgium
| | - D'Huyvetter Matthias
- Molecular Imaging and Therapy Laboratory (MITH), Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Caers Jo
- Department of Hematology, CHU de Liège, Liège, Belgium.
| | - Marcion Guillaume
- Laboratory of Hematology, GIGA I3, University of Liège, Liège, Belgium
- Center for Protein Engineering, University of Liège, Liège, Belgium
| |
Collapse
|
3
|
Li C, Li J. Dysregulation of systemic immunity in colorectal cancer and its clinical applications as biomarkers and therapeutics. Crit Rev Oncol Hematol 2024; 204:104543. [PMID: 39454739 DOI: 10.1016/j.critrevonc.2024.104543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 10/13/2024] [Accepted: 10/21/2024] [Indexed: 10/28/2024] Open
Abstract
The immune system plays critical roles in the initiation and progression of colorectal cancer (CRC), and the majority of studies have focused on immune perturbations within the tumor microenvironment. In recent years, systemic immunity, which mainly occurs in the periphery, has attracted much attention. In CRC, both the tumor itself and treatments have extensive effects on systemic immunity, characterized by alterations in circulating cytokines and immune cells. In addition, intact systemic immunity is critical for the efficacy of therapies for CRC, especially immunotherapy. Therefore, various strategies aimed at alleviating the detrimental effects of traditional therapies or directly harnessing the components of systemic immunity for CRC treatment have been developed. However, whether these improvements can translate to survival benefits requires further study. This review aims to comprehensively outline the current knowledge of systemic immunity in CRC.
Collapse
Affiliation(s)
- Changqin Li
- Department of Clinical Laboratory, the Third Hospital of Mianyang, Sichuan Mental Health Center, Mianyang, Sichuan, China
| | - Jian Li
- Department of General Surgery, the Third Hospital of Mianyang, Sichuan Mental Health Center, Mianyang, Sichuan, China.
| |
Collapse
|
4
|
Calabrò A, Drommi F, Sidoti Migliore G, Pezzino G, Vento G, Freni J, Costa G, Cavaliere R, Bonaccorsi I, Sionne M, Nigro S, Navarra G, Ferlazzo G, De Pasquale C, Campana S. Neutrophil-like Monocytes Increase in Patients with Colon Cancer and Induce Dysfunctional TIGIT+ NK Cells. Int J Mol Sci 2024; 25:8470. [PMID: 39126041 PMCID: PMC11313383 DOI: 10.3390/ijms25158470] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 07/26/2024] [Accepted: 07/28/2024] [Indexed: 08/12/2024] Open
Abstract
Myeloid-derived suppressor cells (MDSCs) are a heterogeneous family of immune cells including granulocytic (CD14neg/CD15+/HLA-DRneg) and monocytic subtypes (CD14+/CD15neg/HLA-DRneg). In the present study, we found a population of monocytes expressing the granulocyte marker CD15 that significantly increased in both peripheral blood (PB) and tumoral tissues of patients with colorectal cancer (CRC). Further phenotypical analysis confirmed the granulocytic-like features of this monocyte subpopulation that is associated with an increase in granulocyte-monocyte precursors (GMPs) in the PB of these patients (pts). Mechanistically, this granulocyte-like monocyte population suppressed NK cell activity by inducing TIGIT and engaging NKp30. Accordingly, an increased frequency of TIGIT+ NK cells with impaired functions was found in both the PB and tumoral tissue of CRC pts. Collectively, we provided new mechanistic explanations for tumor immune escape occurring in CRC by showing the increase in this new kind of MDSC, in both PB and CRC tissue, which is able to significantly impair the effector functions of NK cells, thereby representing a potential therapeutic target for cancer immunotherapy.
Collapse
Affiliation(s)
- Alessia Calabrò
- Laboratory of Immunology and Biotherapy, Department Human Pathology “G. Barresi”, University of Messina, Via Consolare Valeria 1, 98125 Messina, Italy; (A.C.); (F.D.); (G.P.); (G.C.); (I.B.); (C.D.P.); (S.C.)
| | - Fabiana Drommi
- Laboratory of Immunology and Biotherapy, Department Human Pathology “G. Barresi”, University of Messina, Via Consolare Valeria 1, 98125 Messina, Italy; (A.C.); (F.D.); (G.P.); (G.C.); (I.B.); (C.D.P.); (S.C.)
| | - Giacomo Sidoti Migliore
- Translational Immunobiology Unit, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, BLDG 50, RM 6308, Bethesda, MD 20892, USA;
| | - Gaetana Pezzino
- Laboratory of Immunology and Biotherapy, Department Human Pathology “G. Barresi”, University of Messina, Via Consolare Valeria 1, 98125 Messina, Italy; (A.C.); (F.D.); (G.P.); (G.C.); (I.B.); (C.D.P.); (S.C.)
| | - Grazia Vento
- Department of Experimental Medicine (DIMES), University of Genoa, Via Leon Battista Alberti 2, 16132 Genova, Italy;
| | - José Freni
- Laboratory of Histology, Department of Biomedical, Dental, Morphological and Functional Imaging Sciences, University of Messina, Via Consolare Valeria 1, 98125 Messina, Italy;
| | - Gregorio Costa
- Laboratory of Immunology and Biotherapy, Department Human Pathology “G. Barresi”, University of Messina, Via Consolare Valeria 1, 98125 Messina, Italy; (A.C.); (F.D.); (G.P.); (G.C.); (I.B.); (C.D.P.); (S.C.)
- Clinical Pathology Unit, University Hospital Policlinico G. Martino, 98125 Messina, Italy;
| | - Riccardo Cavaliere
- Clinical Pathology Unit, University Hospital Policlinico G. Martino, 98125 Messina, Italy;
| | - Irene Bonaccorsi
- Laboratory of Immunology and Biotherapy, Department Human Pathology “G. Barresi”, University of Messina, Via Consolare Valeria 1, 98125 Messina, Italy; (A.C.); (F.D.); (G.P.); (G.C.); (I.B.); (C.D.P.); (S.C.)
- Clinical Pathology Unit, University Hospital Policlinico G. Martino, 98125 Messina, Italy;
| | - Mariagrazia Sionne
- Oncologic Surgery, Department of Human Pathology of Adult and Evolutive Age, University of Messina, Via Consolare Valeria 1, 98125 Messina, Italy; (M.S.); (S.N.); (G.N.)
| | - Stefania Nigro
- Oncologic Surgery, Department of Human Pathology of Adult and Evolutive Age, University of Messina, Via Consolare Valeria 1, 98125 Messina, Italy; (M.S.); (S.N.); (G.N.)
| | - Giuseppe Navarra
- Oncologic Surgery, Department of Human Pathology of Adult and Evolutive Age, University of Messina, Via Consolare Valeria 1, 98125 Messina, Italy; (M.S.); (S.N.); (G.N.)
| | - Guido Ferlazzo
- Department of Experimental Medicine (DIMES), University of Genoa, Via Leon Battista Alberti 2, 16132 Genova, Italy;
- Unit of Experimental Pathology and Immunology, IRCCS Ospedale Policlinico San Martino, Largo R. Benzi 10, 16132 Genova, Italy
| | - Claudia De Pasquale
- Laboratory of Immunology and Biotherapy, Department Human Pathology “G. Barresi”, University of Messina, Via Consolare Valeria 1, 98125 Messina, Italy; (A.C.); (F.D.); (G.P.); (G.C.); (I.B.); (C.D.P.); (S.C.)
| | - Stefania Campana
- Laboratory of Immunology and Biotherapy, Department Human Pathology “G. Barresi”, University of Messina, Via Consolare Valeria 1, 98125 Messina, Italy; (A.C.); (F.D.); (G.P.); (G.C.); (I.B.); (C.D.P.); (S.C.)
| |
Collapse
|
5
|
Fortin BM, Pfeiffer SM, Insua-Rodríguez J, Alshetaiwi H, Moshensky A, Song WA, Mahieu AL, Chun SK, Lewis AN, Hsu A, Adam I, Eng OS, Pannunzio NR, Seldin MM, Marazzi I, Marangoni F, Lawson DA, Kessenbrock K, Masri S. Circadian control of tumor immunosuppression affects efficacy of immune checkpoint blockade. Nat Immunol 2024; 25:1257-1269. [PMID: 38806707 PMCID: PMC11374317 DOI: 10.1038/s41590-024-01859-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Accepted: 04/29/2024] [Indexed: 05/30/2024]
Abstract
The circadian clock is a critical regulator of immunity, and this circadian control of immune modulation has an essential function in host defense and tumor immunosurveillance. Here we use a single-cell RNA sequencing approach and a genetic model of colorectal cancer to identify clock-dependent changes to the immune landscape that control the abundance of immunosuppressive cells and consequent suppression of cytotoxic CD8+ T cells. Of these immunosuppressive cell types, PD-L1-expressing myeloid-derived suppressor cells (MDSCs) peak in abundance in a rhythmic manner. Disruption of the epithelial cell clock regulates the secretion of cytokines that promote heightened inflammation, recruitment of neutrophils and the subsequent development of MDSCs. We also show that time-of-day anti-PD-L1 delivery is most effective when synchronized with the abundance of immunosuppressive MDSCs. Collectively, these data indicate that circadian gating of tumor immunosuppression informs the timing and efficacy of immune checkpoint inhibitors.
Collapse
Affiliation(s)
- Bridget M Fortin
- Department of Biological Chemistry, University of California Irvine, Irvine, CA, USA
| | - Shannon M Pfeiffer
- Department of Biological Chemistry, University of California Irvine, Irvine, CA, USA
| | - Jacob Insua-Rodríguez
- Department of Biological Chemistry, University of California Irvine, Irvine, CA, USA
- Department of Physiology and Biophysics, University of California Irvine, Irvine, CA, USA
| | - Hamad Alshetaiwi
- Department of Pathology, University of Hail, Hail, Saudi Arabia
- Chao Family Comprehensive Cancer Center, University of California Irvine, Irvine, CA, USA
| | - Alexander Moshensky
- Department of Physiology and Biophysics, University of California Irvine, Irvine, CA, USA
| | - Wei A Song
- Department of Biological Chemistry, University of California Irvine, Irvine, CA, USA
| | - Alisa L Mahieu
- Department of Biological Chemistry, University of California Irvine, Irvine, CA, USA
| | - Sung Kook Chun
- Department of Biological Chemistry, University of California Irvine, Irvine, CA, USA
| | - Amber N Lewis
- Department of Biological Chemistry, University of California Irvine, Irvine, CA, USA
| | - Alex Hsu
- Department of Biological Chemistry, University of California Irvine, Irvine, CA, USA
| | - Isam Adam
- Department of Physiology and Biophysics, University of California Irvine, Irvine, CA, USA
| | - Oliver S Eng
- Chao Family Comprehensive Cancer Center, University of California Irvine, Irvine, CA, USA
- Department of Surgery, Division of Surgical Oncology, University of California Irvine, Orange, CA, USA
| | - Nicholas R Pannunzio
- Department of Biological Chemistry, University of California Irvine, Irvine, CA, USA
- Chao Family Comprehensive Cancer Center, University of California Irvine, Irvine, CA, USA
- Department of Medicine, Division of Hematology/Oncology, University of California Irvine, Irvine, CA, USA
- Center for Epigenetics and Metabolism, University of California Irvine, Irvine, CA, USA
| | - Marcus M Seldin
- Department of Biological Chemistry, University of California Irvine, Irvine, CA, USA
- Chao Family Comprehensive Cancer Center, University of California Irvine, Irvine, CA, USA
- Center for Epigenetics and Metabolism, University of California Irvine, Irvine, CA, USA
| | - Ivan Marazzi
- Department of Biological Chemistry, University of California Irvine, Irvine, CA, USA
- Chao Family Comprehensive Cancer Center, University of California Irvine, Irvine, CA, USA
- Center for Epigenetics and Metabolism, University of California Irvine, Irvine, CA, USA
| | - Francesco Marangoni
- Department of Physiology and Biophysics, University of California Irvine, Irvine, CA, USA
- Chao Family Comprehensive Cancer Center, University of California Irvine, Irvine, CA, USA
| | - Devon A Lawson
- Department of Physiology and Biophysics, University of California Irvine, Irvine, CA, USA
- Chao Family Comprehensive Cancer Center, University of California Irvine, Irvine, CA, USA
- Center for Epigenetics and Metabolism, University of California Irvine, Irvine, CA, USA
| | - Kai Kessenbrock
- Department of Biological Chemistry, University of California Irvine, Irvine, CA, USA
- Chao Family Comprehensive Cancer Center, University of California Irvine, Irvine, CA, USA
- Center for Epigenetics and Metabolism, University of California Irvine, Irvine, CA, USA
| | - Selma Masri
- Department of Biological Chemistry, University of California Irvine, Irvine, CA, USA.
- Chao Family Comprehensive Cancer Center, University of California Irvine, Irvine, CA, USA.
- Center for Epigenetics and Metabolism, University of California Irvine, Irvine, CA, USA.
| |
Collapse
|
6
|
Pettinella F, Mariotti B, Lattanzi C, Bruderek K, Donini M, Costa S, Marini O, Iannoto G, Gasperini S, Caveggion E, Castellucci M, Calzetti F, Bianchetto-Aguilera F, Gardiman E, Giani M, Dusi S, Cantini M, Vassanelli A, Pavone D, Milella M, Pilotto S, Biondani P, Höing B, Schleupner MC, Hussain T, Hadaschik B, Kaspar C, Visco C, Tecchio C, Koenderman L, Bazzoni F, Tamassia N, Brandau S, Cassatella MA, Scapini P. Surface CD52, CD84, and PTGER2 mark mature PMN-MDSCs from cancer patients and G-CSF-treated donors. Cell Rep Med 2024; 5:101380. [PMID: 38242120 PMCID: PMC10897522 DOI: 10.1016/j.xcrm.2023.101380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 08/11/2023] [Accepted: 12/18/2023] [Indexed: 01/21/2024]
Abstract
Precise molecular characterization of circulating polymorphonuclear myeloid-derived suppressor cells (PMN-MDSCs) is hampered by their mixed composition of mature and immature cells and lack of specific markers. Here, we focus on mature CD66b+CD10+CD16+CD11b+ PMN-MDSCs (mPMN-MDSCs) from either cancer patients or healthy donors receiving G-CSF for stem cell mobilization (GDs). By RNA sequencing (RNA-seq) experiments, we report the identification of a distinct gene signature shared by the different mPMN-MDSC populations under investigation, also validated in mPMN-MDSCs from GDs and tumor-associated neutrophils (TANs) by single-cell RNA-seq (scRNA-seq) experiments. Analysis of such a gene signature uncovers a specific transcriptional program associated with mPMN-MDSC differentiation and allows us to identify that, in patients with either solid or hematologic tumors and in GDs, CD52, CD84, and prostaglandin E receptor 2 (PTGER2) represent potential mPMN-MDSC-associated markers. Altogether, our findings indicate that mature PMN-MDSCs distinctively undergo specific reprogramming during differentiation and lay the groundwork for selective immunomonitoring, and eventually targeting, of mature PMN-MDSCs.
Collapse
Affiliation(s)
- Francesca Pettinella
- Section of General Pathology, Department of Medicine, University of Verona, 37134 Verona, Italy
| | - Barbara Mariotti
- Section of General Pathology, Department of Medicine, University of Verona, 37134 Verona, Italy
| | - Chiara Lattanzi
- Section of General Pathology, Department of Medicine, University of Verona, 37134 Verona, Italy
| | - Kirsten Bruderek
- Research Division, Department of Otorhinolaryngology, University Hospital Essen, 45122 Essen, Germany
| | - Marta Donini
- Section of General Pathology, Department of Medicine, University of Verona, 37134 Verona, Italy
| | - Sara Costa
- Section of General Pathology, Department of Medicine, University of Verona, 37134 Verona, Italy
| | - Olivia Marini
- Section of General Pathology, Department of Medicine, University of Verona, 37134 Verona, Italy
| | - Giulia Iannoto
- Section of General Pathology, Department of Medicine, University of Verona, 37134 Verona, Italy
| | - Sara Gasperini
- Section of General Pathology, Department of Medicine, University of Verona, 37134 Verona, Italy
| | - Elena Caveggion
- Section of General Pathology, Department of Medicine, University of Verona, 37134 Verona, Italy
| | | | - Federica Calzetti
- Section of General Pathology, Department of Medicine, University of Verona, 37134 Verona, Italy
| | | | - Elisa Gardiman
- Section of General Pathology, Department of Medicine, University of Verona, 37134 Verona, Italy
| | - Matteo Giani
- Section of General Pathology, Department of Medicine, University of Verona, 37134 Verona, Italy
| | - Stefano Dusi
- Section of General Pathology, Department of Medicine, University of Verona, 37134 Verona, Italy
| | - Maurizio Cantini
- Transfusion Medicine Department, University and Hospital Trust (AOUI), Verona, Italy
| | - Aurora Vassanelli
- Transfusion Medicine Department, University and Hospital Trust (AOUI), Verona, Italy
| | - Denise Pavone
- Transfusion Medicine Department, University and Hospital Trust (AOUI), Verona, Italy
| | - Michele Milella
- Section of Innovation Biomedicine - Oncology Area, Department of Engineering for Innovation Medicine (DIMI), University of Verona, Verona, Italy
| | - Sara Pilotto
- Section of Innovation Biomedicine - Oncology Area, Department of Engineering for Innovation Medicine (DIMI), University of Verona, Verona, Italy
| | - Pamela Biondani
- Section of Oncology, University and Hospital Trust (AOUI) of Verona, Verona, Italy
| | - Benedikt Höing
- Department of Otorhinolaryngology, University Hospital Essen, Essen, Germany
| | | | - Timon Hussain
- Department of Otorhinolaryngology, University Hospital Essen, Essen, Germany
| | - Boris Hadaschik
- Department of Urology, University Hospital Essen, Essen, Germany
| | - Cordelia Kaspar
- Department of Urology, University Hospital Essen, Essen, Germany
| | - Carlo Visco
- Section of Hematology and Bone Marrow Transplant Unit, Department of Engineering for Innovation Medicine (DIMI), University of Verona, Verona, Italy
| | - Cristina Tecchio
- Section of Hematology and Bone Marrow Transplant Unit, Department of Engineering for Innovation Medicine (DIMI), University of Verona, Verona, Italy
| | - Leo Koenderman
- Department of Respiratory Medicine and Center for Translational Immunology, University Medical Center Utrecht, 3584CX Utrecht, the Netherlands
| | - Flavia Bazzoni
- Section of General Pathology, Department of Medicine, University of Verona, 37134 Verona, Italy
| | - Nicola Tamassia
- Section of General Pathology, Department of Medicine, University of Verona, 37134 Verona, Italy
| | - Sven Brandau
- Research Division, Department of Otorhinolaryngology, University Hospital Essen, 45122 Essen, Germany; German Cancer Consortium, Partner Site Essen-Düsseldorf, Essen, Germany
| | - Marco A Cassatella
- Section of General Pathology, Department of Medicine, University of Verona, 37134 Verona, Italy.
| | - Patrizia Scapini
- Section of General Pathology, Department of Medicine, University of Verona, 37134 Verona, Italy.
| |
Collapse
|
7
|
Sartorius D, Blume ML, Fleischer JR, Ghadimi M, Conradi LC, De Oliveira T. Implications of Rectal Cancer Radiotherapy on the Immune Microenvironment: Allies and Foes to Therapy Resistance and Patients' Outcome. Cancers (Basel) 2023; 15:5124. [PMID: 37958298 PMCID: PMC10650490 DOI: 10.3390/cancers15215124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Revised: 10/16/2023] [Accepted: 10/19/2023] [Indexed: 11/15/2023] Open
Abstract
Aside from surgical resection, locally advanced rectal cancer is regularly treated with neoadjuvant chemoradiotherapy. Since the concept of cancer treatment has shifted from only focusing on tumor cells as drivers of disease progression towards a broader understanding including the dynamic tumor microenvironment (TME), the impact of radiotherapy on the TME and specifically the tumor immune microenvironment (TIME) is increasingly recognized. Both promoting as well as suppressing effects on anti-tumor immunity have been reported in response to rectal cancer (chemo-)radiotherapy and various targets for combination therapies are under investigation. A literature review was conducted searching the PubMed database for evidence regarding the pleiotropic effects of (chemo-)radiotherapy on the rectal cancer TIME, including alterations in cytokine levels, immune cell populations and activity as well as changes in immune checkpoint proteins. Radiotherapy can induce immune-stimulating and -suppressive alterations, potentially mediating radioresistance. The response is influenced by treatment modalities, including the dosage administered and the highly individual intrinsic pre-treatment immune status. Directly addressing the main immune cells of the TME, this review aims to highlight therapeutical implications since efficient rectal cancer treatment relies on personalized strategies combining conventional therapies with immune-modulating approaches, such as immune checkpoint inhibitors.
Collapse
Affiliation(s)
| | | | | | | | - Lena-Christin Conradi
- Department of General, Visceral and Pediatric Surgery, University Medical Center Göttingen, Robert-Koch-Straβe 40, 37075 Göttingen, Germany; (D.S.); (M.L.B.); (J.R.F.); (M.G.)
| | - Tiago De Oliveira
- Department of General, Visceral and Pediatric Surgery, University Medical Center Göttingen, Robert-Koch-Straβe 40, 37075 Göttingen, Germany; (D.S.); (M.L.B.); (J.R.F.); (M.G.)
| |
Collapse
|
8
|
Chang CH, Chen CJ, Yu CF, Tsai HY, Chen FH, Chiang CS. Targeting M-MDSCs enhances the therapeutic effect of BNCT in the 4-NQO-induced murine head and neck squamous cell carcinoma model. Front Oncol 2023; 13:1263873. [PMID: 37886177 PMCID: PMC10598372 DOI: 10.3389/fonc.2023.1263873] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Accepted: 09/15/2023] [Indexed: 10/28/2023] Open
Abstract
Purpose Malignant head and neck squamous cell carcinoma (HNSCC) is characterized by a poor prognosis and resistance to conventional radiotherapy. Infiltrating myeloid-derived suppressive cells (MDSCs) is prominent in HNSCC and is linked to immune suppression and tumor aggressiveness. This study aimed to investigate the impact of boron neutron capture therapy (BNCT) on the MDSCs in the tumor microenvironment and peripheral blood and to explore the potential for MDSCs depletion combined with BNCT to reactivate antitumor immunity. Methods and materials Carcinogen, 4-NQO, -induced oral tumors were irradiated with a total physical dose of 2 Gy BNCT in Tsing Hua Open Reactor (THOR). Flow cytometry and immunohistochemistry accessed the dynamics of peripheral MDSCs and infiltrated MDSCs within the tumor microenvironment. Mice were injected with an inhibitor of CSF-1 receptor (CSF-1R), PLX3397, to determine whether modulating M-MDSCs could affect mice survival after BNCT. Results Peripheral CD11b+Ly6ChighLy6G- monocytic-MDSCs (M-MDSCs), but not CD11b+Ly6CloLy6Ghigh polymorphonuclear-MDSCs (PMN-MDSCs), increased as tumor progression. After BNCT treatment, there were temporarily decreased and persistent increases of M-MDSCs thereafter, either in peripheral blood or in tumors. The administration of PLX-3397 hindered BNCT-caused M-MDSCs infiltration, prolonged mice survival, and activated tumor immunity by decreasing tumor-associated macrophages (TAMs) and increasing CD8+ T cells. Conclusion M-MDSCs were recruited into 4-NQO-induced tumors after BNCT, and their number was also increased in peripheral blood. Assessment of M-MDSCs levels in peripheral blood could be an index to determine the optimal intervention window. Their temporal alteration suggests an association with tumor recurrence after BNCT, making M-MDSCs a potential intervention target. Our preliminary results showed that PLX-3397 had strong M-MDSCs, TAMs, and TIL (tumor-infiltrating lymphocyte) modulating effects that could synergize tumor control when combined with BNCT.
Collapse
Affiliation(s)
- Chun-Hsiang Chang
- Department of Biomedical Engineering and Environment Sciences, National Tsing Hua University, Hsinchu, Taiwan
| | - Chi-Jui Chen
- Department of Biomedical Engineering and Environment Sciences, National Tsing Hua University, Hsinchu, Taiwan
| | - Ching-Fang Yu
- Institute for Radiological Research, Chang Gung University, Taoyuan, Taiwan
- Department of Radiation Oncology, Chang Gung Memorial Hospital Linkou Branch, Taoyuan, Taiwan
| | - Hui-Yu Tsai
- Institute of Nuclear Engineering and Science, National Tsing Hua University, Hsinchu, Taiwan
| | - Fang-Hsin Chen
- Institute of Nuclear Engineering and Science, National Tsing Hua University, Hsinchu, Taiwan
| | - Chi-Shiun Chiang
- Department of Biomedical Engineering and Environment Sciences, National Tsing Hua University, Hsinchu, Taiwan
- Institute of Nuclear Engineering and Science, National Tsing Hua University, Hsinchu, Taiwan
| |
Collapse
|
9
|
Stagg J, Golden E, Wennerberg E, Demaria S. The interplay between the DNA damage response and ectonucleotidases modulates tumor response to therapy. Sci Immunol 2023; 8:eabq3015. [PMID: 37418547 PMCID: PMC10394739 DOI: 10.1126/sciimmunol.abq3015] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Accepted: 06/14/2023] [Indexed: 07/09/2023]
Abstract
The extracellular nucleoside adenosine reduces tissue inflammation and is generated by irreversible dephosphorylation of adenosine monophosphate (AMP) mediated by the ectonucleotidase CD73. The pro-inflammatory nucleotides adenosine triphosphate, nicotinamide adenine dinucleotide, and cyclic guanosine -monophosphate-AMP (cGAMP), which are produced in the tumor microenvironment (TME) during therapy-induced immunogenic cell death and activation of innate immune signaling, can be converted into AMP by ectonucleotidases CD39, CD38, and CD203a/ENPP1. Thus, ectonucleotidases shape the TME by converting immune-activating signals into an immunosuppressive one. Ectonucleotidases also hinder the ability of therapies including radiation therapy, which enhance the release of pro-inflammatory nucleotides in the extracellular milieu, to induce immune-mediated tumor rejection. Here, we review the immunosuppressive effects of adenosine and the role of different ectonucleotidases in modulating antitumor immune responses. We discuss emerging opportunities to target adenosine generation and/or its ability to signal via adenosine receptors expressed by immune and cancer cells in the context of combination immunotherapy and radiotherapy.
Collapse
Affiliation(s)
- John Stagg
- Centre de Recherche du Centre Hospitalier de
l’Université de Montréal, 900 St-Denis street, Montreal,
Quebec, Canada, H2X 0A9
| | - Encouse Golden
- Department of Radiation Oncology, Weill Cornell Medicine,
New York, NY 10065, USA
| | - Erik Wennerberg
- Division of Radiotherapy and Imaging, Institute of Cancer
Research, London SM2 5NG, UK
| | - Sandra Demaria
- Department of Radiation Oncology, Weill Cornell Medicine,
New York, NY 10065, USA
- Department of Pathology and Laboratory Medicine, Weill
Cornell Medicine, New York, NY, 10065, USA
| |
Collapse
|
10
|
Sato R, Oikawa M, Kakita T, Okada T, Abe T, Tsuchiya H, Akazawa N, Ohira T, Harada Y, Okano H, Ito K, Tsuchiya T. A decreased preoperative platelet-to-lymphocyte ratio, systemic immune-inflammation index, and pan-immune-inflammation value are associated with the poorer survival of patients with a stent inserted as a bridge to curative surgery for obstructive colorectal cancer. Surg Today 2023; 53:409-419. [PMID: 35987967 DOI: 10.1007/s00595-022-02575-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Accepted: 07/19/2022] [Indexed: 10/15/2022]
Abstract
PURPOSE Inflammation is one of the hallmarks of cancer, and inflammation-based markers that are calculated easily from laboratory results have shown predictive abilities. We investigated the prognostic values of the preoperative platelet-to-lymphocyte ratio (PLR), systemic immune-inflammation index (SII), and pan-immune-inflammation value (PIV) in patients with non-metastatic obstructive colorectal cancer (OCRC) and a self-expandable metallic stent inserted as a bridge to curative surgery. METHODS The subjects of this retrospective study were 86 patients with pathological stage I to III OCRC. We examined the associations of these biomarkers with short- and long-term outcomes. RESULTS Multivariate analyses revealed that a preoperative PLR < 149, SII < 597, and PIV < 209 were independently associated with poorer relapse-free survival (RFS) (P = 0.007, P < 0.001, and P = 0.002, respectively) and that a PIV < 209 was independently associated with poorer cancer-specific survival (P = 0.030). A platelet count < 240 was significantly associated with worse RFS, whereas the lymphocyte count was not. Pre-stenting PLR < 221 was an independent poor prognostic factor for RFS (P = 0.045). CONCLUSION This study showed that decreased preoperative PLR, SII, PIV, and pre-stenting PLR were associated with poorer RFS, contrary to the findings of most previous studies. Our results suggest that platelets and obstruction contributed primarily to the opposite relationships, which might provide new insight into the possible pathophysiology of platelet-tumor interactions generated in the OCRC environment.
Collapse
Affiliation(s)
- Ryuichiro Sato
- Department of Gastroenterological Surgery, Sendai City Medical Center Sendai Open Hospital, 5-22-1 Tsurugaya, Miyagino-ku, Sendai, 983-0824, Japan.
- Department of Surgery, Japanese Red Cross Sendai Hospital, 2-43-3 Yagiyama hon-cho, Taihaku-ku, Sendai, 982-8501, Japan.
| | - Masaya Oikawa
- Department of Gastroenterological Surgery, Sendai City Medical Center Sendai Open Hospital, 5-22-1 Tsurugaya, Miyagino-ku, Sendai, 983-0824, Japan
| | - Tetsuya Kakita
- Department of Gastroenterological Surgery, Sendai City Medical Center Sendai Open Hospital, 5-22-1 Tsurugaya, Miyagino-ku, Sendai, 983-0824, Japan
| | - Takaho Okada
- Department of Gastroenterological Surgery, Sendai City Medical Center Sendai Open Hospital, 5-22-1 Tsurugaya, Miyagino-ku, Sendai, 983-0824, Japan
| | - Tomoya Abe
- Department of Gastroenterological Surgery, Sendai City Medical Center Sendai Open Hospital, 5-22-1 Tsurugaya, Miyagino-ku, Sendai, 983-0824, Japan
| | - Haruyuki Tsuchiya
- Department of Gastroenterological Surgery, Sendai City Medical Center Sendai Open Hospital, 5-22-1 Tsurugaya, Miyagino-ku, Sendai, 983-0824, Japan
| | - Naoya Akazawa
- Department of Gastroenterological Surgery, Sendai City Medical Center Sendai Open Hospital, 5-22-1 Tsurugaya, Miyagino-ku, Sendai, 983-0824, Japan
| | - Tetsuya Ohira
- Department of Gastroenterology, Sendai City Medical Center Sendai Open Hospital, 5-22-1 Tsurugaya, Miyagino-ku, Sendai, 983-0824, Japan
| | - Yoshihiro Harada
- Department of Gastroenterology, Sendai City Medical Center Sendai Open Hospital, 5-22-1 Tsurugaya, Miyagino-ku, Sendai, 983-0824, Japan
| | - Haruka Okano
- Department of Gastroenterology, Sendai City Medical Center Sendai Open Hospital, 5-22-1 Tsurugaya, Miyagino-ku, Sendai, 983-0824, Japan
| | - Kei Ito
- Department of Gastroenterology, Sendai City Medical Center Sendai Open Hospital, 5-22-1 Tsurugaya, Miyagino-ku, Sendai, 983-0824, Japan
| | - Takashi Tsuchiya
- Department of Gastroenterological Surgery, Sendai City Medical Center Sendai Open Hospital, 5-22-1 Tsurugaya, Miyagino-ku, Sendai, 983-0824, Japan
| |
Collapse
|
11
|
Masuda Y, Nakayama Y, Shimizu R, Naito K, Miyamoto E, Tanaka A, Konishi M. Maitake α-glucan promotes differentiation of monocytic myeloid-derived suppressor cells into M1 macrophages. Life Sci 2023; 317:121453. [PMID: 36709912 DOI: 10.1016/j.lfs.2023.121453] [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: 11/06/2022] [Revised: 01/17/2023] [Accepted: 01/24/2023] [Indexed: 01/27/2023]
Abstract
AIMS Myeloid-derived suppressor cells (MDSCs) are major components of the tumor microenvironment and systemically accumulate in tumor-bearing hosts and patients with cancer, facilitating cancer progression. Maitake macromolecular α-glucan YM-2A, isolated from Grifola frondosa, inhibits tumor growth by enhancing immune responses. The present study investigated the effects of YM-2A on the immunosuppressive potential of MDSCs. MAIN METHODS YM-2A was orally administered to CT26 tumor-bearing mice, and the number of immune cells in the spleen and tumor was measured. Splenic MDSCs isolated from the CT26 tumor-bearing mice were treated with YM-2A and co-cultured with T cells to measure their inhibitory effect on T cell proliferation. For adoptive transfer of monocytic MDSCs (M-MDSCs), YM-2A-treated M-MDSCs mixed with CT26 cells were implanted subcutaneously in the mice to measure the tumor growth rate. KEY FINDINGS YM-2A selectively reduced the accumulation of M-MDSCs but not that of polymorphonuclear MDSCs (PMN-MDSCs) in CT26 tumor-bearing mice. In tumor tissues, YM-2A treatment induced the polarity of immunostimulatory M1-phenotype; furthermore, it increased the infiltration of dendritic, natural killer, and CD4+ and CD8+ T cells. YM-2A treatment of purified M-MDSCs from CT-26 tumor-bearing mice induced dectin-1-dependent differentiation into M1 macrophages. YM-2A-treated M-MDSCs lost their inhibitory activity against proliferation and activation of CD8+ T cells. Furthermore, adoptive transfer of M-MDSCs treated with YM-2A inhibited CT26 tumor growth. SIGNIFICANCE YM-2A promotes the differentiation of M-MDSCs into immunostimulatory M1 macrophages, thereby enhancing the efficacy of cancer immunotherapy.
Collapse
Affiliation(s)
- Yuki Masuda
- Department of Microbial Chemistry, Kobe Pharmaceutical University, Kobe, Japan
| | - Yoshiaki Nakayama
- Department of Microbial Chemistry, Kobe Pharmaceutical University, Kobe, Japan
| | - Ryohei Shimizu
- Department of Microbial Chemistry, Kobe Pharmaceutical University, Kobe, Japan
| | - Kenta Naito
- Research and Development Department, Yukiguni Maitake Co., Ltd., Niigata, Japan
| | - Eri Miyamoto
- Research and Development Department, Yukiguni Maitake Co., Ltd., Niigata, Japan
| | - Akihiro Tanaka
- Research and Development Department, Yukiguni Maitake Co., Ltd., Niigata, Japan
| | - Morichika Konishi
- Department of Microbial Chemistry, Kobe Pharmaceutical University, Kobe, Japan.
| |
Collapse
|
12
|
Gao L, Du X, Li J, Qin FXF. Evolving roles of CD38 metabolism in solid tumour microenvironment. Br J Cancer 2023; 128:492-504. [PMID: 36396822 PMCID: PMC9938187 DOI: 10.1038/s41416-022-02052-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 10/20/2022] [Accepted: 10/27/2022] [Indexed: 11/19/2022] Open
Abstract
Given that plenty of clinical findings and reviews have already explained in detail on the progression of CD38 in multiple myeloma and haematological system tumours, here we no longer give unnecessary discussion on the above progression. Though therapeutic antibodies have been regarded as a greatest breakthrough in multiple myeloma immunotherapies due to the durable anti-tumour responses in the clinic, but the role of CD38 in the immunologic regulation and evasion of non-hematopoietic solid tumours are just initiated and controversial. Therefore, we will focus on the bio-function of CD38 enzymatic substrates or metabolites in the variety of non-hematopoietic malignancies and the potential therapeutic value of targeting the CD38-NAD+ or CD38-cADPR/ADPR signal axis. Though limited, we review some ongoing researches and clinical trials on therapeutic approaches in solid tumour as well.
Collapse
Affiliation(s)
- Long Gao
- Department of Infectious Disease, The First Affiliated Hospital of Anhui Medical University, 230022, Hefei, China
| | - Xiaohong Du
- Institute of Clinical Medicine Research, Suzhou Science and Technology Town Hospital, Suzhou, China
| | - Jiabin Li
- Department of Infectious Disease, The First Affiliated Hospital of Anhui Medical University, 230022, Hefei, China.
| | - F Xiao-Feng Qin
- Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, 100005, Beijing, China.
- Suzhou Institute of Systems Medicine, 215123, Suzhou, China.
| |
Collapse
|
13
|
Christofides A, Katopodi XL, Cao C, Karagkouni D, Aliazis K, Yenyuwadee S, Aksoylar HI, Pal R, Mahmoud MAA, Strauss L, Tijaro-Ovalle NM, Boon L, Asara J, Vlachos IS, Patsoukis N, Boussiotis VA. SHP-2 and PD-1-SHP-2 signaling regulate myeloid cell differentiation and antitumor responses. Nat Immunol 2023; 24:55-68. [PMID: 36581713 PMCID: PMC9810534 DOI: 10.1038/s41590-022-01385-x] [Citation(s) in RCA: 28] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Accepted: 11/03/2022] [Indexed: 12/31/2022]
Abstract
The inhibitory receptor PD-1 suppresses T cell activation by recruiting the phosphatase SHP-2. However, mice with a T-cell-specific deletion of SHP-2 do not have improved antitumor immunity. Here we showed that mice with conditional targeting of SHP-2 in myeloid cells, but not in T cells, had diminished tumor growth. RNA sequencing (RNA-seq) followed by gene set enrichment analysis indicated the presence of polymorphonuclear myeloid-derived suppressor cells and tumor-associated macrophages (TAMs) with enriched gene expression profiles of enhanced differentiation, activation and expression of immunostimulatory molecules. In mice with conditional targeting of PD-1 in myeloid cells, which also displayed diminished tumor growth, TAMs had gene expression profiles enriched for myeloid differentiation, activation and leukocyte-mediated immunity displaying >50% overlap with enriched profiles of SHP-2-deficient TAMs. In bone marrow, GM-CSF induced the phosphorylation of PD-1 and recruitment of PD-1-SHP-2 to the GM-CSF receptor. Deletion of SHP-2 or PD-1 enhanced GM-CSF-mediated phosphorylation of the transcription factors HOXA10 and IRF8, which regulate myeloid differentiation and monocytic-moDC lineage commitment, respectively. Thus, SHP-2 and PD-1-SHP-2 signaling restrained myelocyte differentiation resulting in a myeloid landscape that suppressed antitumor immunity.
Collapse
Affiliation(s)
- Anthos Christofides
- Division of Hematology-Oncology, Beth Israel Deaconess Medical Center, Boston, MA, USA
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
- Cancer Center, Beth Israel Deaconess Medical Center, Boston, MA, USA
- Department of Medicine, Yale University, New Haven, CT, USA
| | - Xanthi-Lida Katopodi
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
- Harvard Medical School Initiative for RNA Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Carol Cao
- Division of Hematology-Oncology, Beth Israel Deaconess Medical Center, Boston, MA, USA
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
- Harvard College, Cambridge, MA, USA
| | - Dimitra Karagkouni
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
- Harvard Medical School Initiative for RNA Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Konstantinos Aliazis
- Division of Hematology-Oncology, Beth Israel Deaconess Medical Center, Boston, MA, USA
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
- Cancer Center, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Sasitorn Yenyuwadee
- Division of Hematology-Oncology, Beth Israel Deaconess Medical Center, Boston, MA, USA
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
- Cancer Center, Beth Israel Deaconess Medical Center, Boston, MA, USA
- Department of Dermatology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Halil-Ibrahim Aksoylar
- Division of Hematology-Oncology, Beth Israel Deaconess Medical Center, Boston, MA, USA
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
- Cancer Center, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Rinku Pal
- Division of Hematology-Oncology, Beth Israel Deaconess Medical Center, Boston, MA, USA
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
- Cancer Center, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Mohamed A A Mahmoud
- Division of Hematology-Oncology, Beth Israel Deaconess Medical Center, Boston, MA, USA
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
- Heidelberg University, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Laura Strauss
- Division of Hematology-Oncology, Beth Israel Deaconess Medical Center, Boston, MA, USA
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
- Cancer Center, Beth Israel Deaconess Medical Center, Boston, MA, USA
- Sanofi /Tidal, Cambridge, MA, USA
| | - Natalia M Tijaro-Ovalle
- Division of Hematology-Oncology, Beth Israel Deaconess Medical Center, Boston, MA, USA
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
- Department of Medicine, Yale University, New Haven, CT, USA
| | | | - John Asara
- Cancer Center, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Ioannis S Vlachos
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
- Harvard Medical School Initiative for RNA Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Cancer Research Institute, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Nikolaos Patsoukis
- Division of Hematology-Oncology, Beth Israel Deaconess Medical Center, Boston, MA, USA
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
- Cancer Center, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Vassiliki A Boussiotis
- Division of Hematology-Oncology, Beth Israel Deaconess Medical Center, Boston, MA, USA.
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA.
- Cancer Center, Beth Israel Deaconess Medical Center, Boston, MA, USA.
| |
Collapse
|
14
|
Hong Y, Chen L, Sun J, Xing L, Yang Y, Jin X, Cai H, Dong L, Zhou L, Zhang Z. Single-cell transcriptome profiling reveals heterogeneous neutrophils with prognostic values in sepsis. iScience 2022; 25:105301. [PMID: 36304125 PMCID: PMC9593767 DOI: 10.1016/j.isci.2022.105301] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 08/16/2022] [Accepted: 09/29/2022] [Indexed: 11/20/2022] Open
Abstract
Neutrophils constitute the largest proportion of nucleated peripheral blood cells, and neutrophils have substantial heterogeneity. We profiled nearly 300,000 human peripheral blood cells in this study using single-cell RNA sequencing. A large proportion (>50%) of these cells were annotated as neutrophils. Neutrophils were further clustered into four subtypes, including Neu1, Neu2, Neu3, and Neu4. Neu1 is characterized by high expression of MMP9, HP, and RGL4. Neu1 was associated with septic shock and significantly correlated with the sequential organ failure assessment (SOFA) score. A gene expression module in Neu1 named Neu1_C (characterized by expression of NFKBIA, CXCL8, G0S2, and FTH1) was highly predictive of septic shock with an area under the curve of 0.81. The results were extensively validated in external bulk datasets by using single-cell deconvolution methods. In summary, our study establishes a general framework for studying neutrophil-related mechanisms, prognostic biomarkers, and potential therapeutic targets for septic shock. Neutrophils were clustered into four subtypes, including Neu1, Neu2, Neu3, and Neu4 Neu1 was associated with septic shock Neu1 was correlated with the sequential organ failure assessment (SOFA) score A gene expression module in Neu1 named Neu1_C was highly predictive of septic shock
Collapse
Affiliation(s)
- Yucai Hong
- Department of Emergency Medicine, Key Laboratory of Precision Medicine in Diagnosis and Monitoring Research of Zhejiang Province, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310016, China
| | - Lin Chen
- Department of Critical Care Medicine, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, China
| | - Jian Sun
- Department of Critical Care Medicine, Lishui Center Hospital, Lishui, Zhejiang 323000, China
| | - Lifeng Xing
- Department of Emergency Medicine, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310016, China
| | - Yi Yang
- Department of Emergency Medicine, The Second Hospital of Jiaxing, Jiaxing, 314000, P.R.China
| | - Xiaohong Jin
- Department of Emergency Medicine, The Affiliated Wenling Hospital of Wenzhou Medical University, Wenling 317500, Zhejiang Province, China
| | - Huabo Cai
- Department of Emergency Medicine, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310016, China
| | - Lianlian Dong
- Department of Emergency Medicine, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310016, China
| | - Liping Zhou
- Department of Emergency Medicine, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310016, China
| | - Zhongheng Zhang
- Department of Emergency Medicine, Key Laboratory of Precision Medicine in Diagnosis and Monitoring Research of Zhejiang Province, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310016, China,Key Laboratory of Digital Technology in Medical Diagnostics Of Zhejiang Province, Hangzhou, Zhejiang, China,Corresponding author
| |
Collapse
|
15
|
Al-Mterin MA, Elkord E. Myeloid-derived suppressor cells in colorectal cancer: prognostic biomarkers and therapeutic targets. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2022; 3:497-510. [PMID: 36081407 PMCID: PMC9448663 DOI: 10.37349/etat.2022.00097] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 07/20/2022] [Indexed: 12/24/2022] Open
Abstract
Myeloid-derived suppressor cells (MDSCs) are a group of immature myeloid cells, which are expanded in most cancer patients. MDSCs suppress host immune responses, leading to cancer growth and progression. Several studies demonstrated that there was a relationship between levels of MDSCs and tumorigenesis in colorectal cancer (CRC) patients. MDSCs are now being investigated for their role as possible therapeutic targets in cancer treatment. This review summarizes available studies that investigated MDSC expansion in CRC patients, as well as their role in CRC tumorigenesis, prognosis, and targeting. Based on the available studies, there is a possible relationship between high levels of MDSCs and CRC progression. Additionally, targeting MDSCs in CRC patients selectively represents a significant challenge for the development of targeted treatments. Targeting of MDSCs could be exploited in different ways including MDSC depletion, inhibition of MDSC function and recruitment, and enhancing MDSC differentiation. Overall, MDSCs could be exploited as prognostic biomarkers and potential therapeutic targets in CRC.
Collapse
Affiliation(s)
| | - Eyad Elkord
- Natural and Medical Sciences Research Center, University of Nizwa, Nizwa 616, Oman;Biomedical Research Center, School of Science, Engineering and Environment, University of Salford, M5 4WT Manchester, UK
| |
Collapse
|
16
|
Targeting CD38 in Neoplasms and Non-Cancer Diseases. Cancers (Basel) 2022; 14:cancers14174169. [PMID: 36077708 PMCID: PMC9454480 DOI: 10.3390/cancers14174169] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 08/21/2022] [Accepted: 08/25/2022] [Indexed: 01/12/2023] Open
Abstract
Simple Summary CD38 remains an interesting target for anticancer therapy. Its relatively high abundance in neoplasms and crucial impact on NAD+/cADPR metabolism and the activity of T cells allows for changing the immune response in autoimmune diseases, neoplasms, and finally the induction of cell death. Antibody-dependent cell cytotoxicity is responsible for cell death induced by targeting the tumor with anti-CD38 antibodies, such as daratumumab. A wide range of laboratory experiments and clinical trials show an especially promising role of anti-CD38 therapy against multiple myeloma, NK cell lymphomas, and CD19- B-cell malignancies. More studies are required to include more diseases in the therapeutic protocols involving the modulation of CD38 activity. Abstract CD38 is a myeloid antigen present both on the cell membrane and in the intracellular compartment of the cell. Its occurrence is often enhanced in cancer cells, thus making it a potential target in anticancer therapy. Daratumumab and isatuximab already received FDA approval, and novel agents such as MOR202, TAK079 and TNB-738 undergo clinical trials. Also, novel therapeutics such as SAR442085 aim to outrank the older antibodies against CD38. Multiple myeloma and immunoglobulin light-chain amyloidosis may be effectively treated with anti-CD38 immunotherapy. Its role in other hematological malignancies is also important concerning both diagnostic process and potential treatment in the future. Aside from the hematological malignancies, CD38 remains a potential target in gastrointestinal, neurological and pulmonary system disorders. Due to the strong interaction of CD38 with TCR and CD16 on T cells, it may also serve as the biomarker in transplant rejection in renal transplant patients. Besides, CD38 finds its role outside oncology in systemic lupus erythematosus and collagen-induced arthritis. CD38 plays an important role in viral infections, including AIDS and COVID-19. Most of the undergoing clinical trials focus on the use of anti-CD38 antibodies in the therapy of multiple myeloma, CD19- B-cell malignancies, and NK cell lymphomas. This review focuses on targeting CD38 in cancer and non-cancerous diseases using antibodies, cell-based therapies and CD38 inhibitors. We also provide a summary of current clinical trials targeting CD38.
Collapse
|
17
|
Bai H, Li QZ, Qi YC, Zhai YY, Jin W. The prediction of tumor and normal tissues based on the DNA methylation values of ten key sites. BIOCHIMICA ET BIOPHYSICA ACTA. GENE REGULATORY MECHANISMS 2022; 1865:194841. [PMID: 35798200 DOI: 10.1016/j.bbagrm.2022.194841] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/19/2022] [Revised: 05/28/2022] [Accepted: 06/28/2022] [Indexed: 06/15/2023]
Abstract
Abnormal DNA methylation can alter the gene expression to promote or inhibit tumorigenesis in colon adenocarcinoma (COAD). However, the finding important genes and key sites of abnormal DNA methylation which result in the occurrence of COAD is still an eventful task. Here, we studied the effects of DNA methylation in the 12 types of genomic features on the changes of gene expression in COAD, the 10 important COAD-related genes and the key abnormal DNA methylation sites were identified. The effects of important genes on the prognosis were verified by survival analysis. Moreover, it was shown that the important genes were participated in cancer pathways and were hub genes in a co-expression network. Based on the DNA methylation levels in the ten sites, the least diversity increment algorithm for predicting tumor tissues and normal tissues in seventeen cancer types are proposed. The better results are obtained in jackknife test. For example, the predictive accuracies are 94.17 %, 91.28 %, 89.04 % and 88.89 %, respectively, for COAD, rectum adenocarcinoma, pancreatic adenocarcinoma and cholangiocarcinoma. Finally, by computing enrichment score of infiltrating immunocytes and the activity of immune pathways, we found that the genes are highly correlated with immune microenvironment.
Collapse
Affiliation(s)
- Hui Bai
- Laboratory of Theoretical Biophysics, School of Physical Science and Technology, Inner Mongolia University, Hohhot 010021, China
| | - Qian-Zhong Li
- Laboratory of Theoretical Biophysics, School of Physical Science and Technology, Inner Mongolia University, Hohhot 010021, China; The State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, Inner Mongolia University, Hohhot 010070, China.
| | - Ye-Chen Qi
- Laboratory of Theoretical Biophysics, School of Physical Science and Technology, Inner Mongolia University, Hohhot 010021, China
| | - Yuan-Yuan Zhai
- Laboratory of Theoretical Biophysics, School of Physical Science and Technology, Inner Mongolia University, Hohhot 010021, China
| | - Wen Jin
- Inner Mongolia key laboratory of gene regulation of the metabolic disease, Department of Clinical Medical Research Center, Inner Mongolia People's Hospital, Hohhot 010010, China
| |
Collapse
|
18
|
Zhang Q, Zhang W, Lin T, Lu W, He X, Ding Y, Chen W, Diao W, Ding M, Shen P, Guo H. Mass cytometry reveals immune atlas of urothelial carcinoma. BMC Cancer 2022; 22:677. [PMID: 35725444 PMCID: PMC9210814 DOI: 10.1186/s12885-022-09788-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 06/16/2022] [Indexed: 11/10/2022] Open
Abstract
Immunotherapy has emerged as a robust clinical strategy for cancer treatment. PD1/PD-L1 inhibitors have been used as second-line therapy for urothelial carcinoma due to the high tumor mutational burden. Despite the efficacy of the treatment is significant, the response rate is still poor. The tumor immune microenvironment plays a key role in the regulation of immunotherapeutic efficacy. However, a comprehensive understanding of the intricate microenvironment in clinical samples remains unclear. To obtain detailed systematic tumor immune profile, we performed an in-depth immunoassay on 12 human urothelial carcinoma tissue samples and 14 paratumor tissue samples using mass cytometry. Among the large number of cells assayed, we identified 71 T-cell phenotypes, 30 tumor-associated macrophage phenotypes. T cell marker expression profiles showed that almost all T cells in the tumor tissue were in a state of exhaustion. CD38 expression on tumor-associated macrophages (TAMs) was significantly higher than PDL1, and CD38+ TAMs were closely associated with immunosuppression. CD38 may be a more suitable target for immunotherapy in urothelial carcinoma compared to PD1/PDL1. This single-cell analysis of clinical samples expands our insights into the immune microenvironment of urothelial carcinoma and reveals potential biomarkers and targets for immunotherapy development.
Collapse
Affiliation(s)
- Qing Zhang
- Department of Urology, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Institute of Urology, Nanjing University, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, China
| | - Wenlong Zhang
- Department of Urology, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Institute of Urology, Nanjing University, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, China
| | - Tingsheng Lin
- Department of Urology, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Institute of Urology, Nanjing University, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, China
| | - Wenfeng Lu
- Department of Urology, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Institute of Urology, Nanjing University, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, China
| | - Xin He
- Department of Urology, Drum Tower Hospital Clinical College of Nanjing Medical University, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, China
| | - Yuanzhen Ding
- Department of Urology, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Institute of Urology, Nanjing University, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, China
| | - Wei Chen
- Department of Urology, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Institute of Urology, Nanjing University, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, China
| | - Wenli Diao
- Department of Urology, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Institute of Urology, Nanjing University, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, China
| | - Meng Ding
- Department of Urology, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Institute of Urology, Nanjing University, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, China
| | - Pingping Shen
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Urology, School of Life Science, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing University, Nanjing, 210023, Jiangsu, China.
| | - Hongqian Guo
- Department of Urology, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Institute of Urology, Nanjing University, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, China.
| |
Collapse
|
19
|
Zhang Y, Li W, Ma K, Zhai J, Jin Y, Zhang L, Chen C. Elevated CD38 expression characterizes impaired CD8+ T cell immune response in metastatic pleural effusions. Immunol Lett 2022; 245:61-68. [DOI: 10.1016/j.imlet.2022.04.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2022] [Revised: 04/02/2022] [Accepted: 04/11/2022] [Indexed: 11/28/2022]
|
20
|
Sato S, Shimizu T, Ishizuka M, Suda K, Shibuya N, Hachiya H, Iso Y, Takagi K, Aoki T, Kubota K. The preoperative pan-immune-inflammation value is a novel prognostic predictor for with stage I-III colorectal cancer patients undergoing surgery. Surg Today 2022; 52:1160-1169. [PMID: 35015151 DOI: 10.1007/s00595-021-02448-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Accepted: 11/15/2021] [Indexed: 12/20/2022]
Abstract
PURPOSE The pan-immune-inflammation value (PIV) is useful for stratifying outcomes in patients with metastatic colorectal cancer. However, it is unclear whether preoperative PIV can predict the surgical outcomes of patients with stage I-III colorectal cancer who receive surgery. METHODS The records of 758 patients with stage I-III colorectal cancer who received surgical treatment were retrospectively reviewed. The preoperative PIV was calculated as follows: (neutrophil count × platelet count × monocyte count)/lymphocyte count. The cut-off value was determined using a receiver operating characteristic curve for overall survival. RESULTS The cut-off value of the preoperative PIV was 376. Five hundred sixty-eight patients (74.9%) had low values (≤ 376), and 190 (25.1%) had high values (> 376). Univariate and multivariate analyses revealed that the PIV (> 376/ ≤ 376) (HR 2.485; 95% CI 1.552-3.981, P < 0.001) was significantly associated with overall survival, as well as age (> 60/ ≤ 60, years) (HR 1.988; 95% CI 1.038-3.807, P = 0.038), globulin-to-albumin ratio (> 0.83/ ≤ 0.83) (HR 2.013; 95% CI 1.231-3.290, P = 0.005) and postoperative complication (C-D grade III-V/0-II) (HR 1.991; 95% CI 1.154-3.438, P = 0.013). The Kaplan-Meier method and log-rank test showed significant differences in overall survival between patients with stage I-III disease with high (> 376) and low (≤ 376) PIVs. CONCLUSION The preoperative PIV is useful for predicting surgical outcomes in patients with stage I-III colorectal cancer.
Collapse
Affiliation(s)
- Shun Sato
- Second Department of Surgery, Dokkyo Medical University, 880 Kitakobayashi, Mibu, Tochigi, 321-0293, Japan
| | - Takayuki Shimizu
- Second Department of Surgery, Dokkyo Medical University, 880 Kitakobayashi, Mibu, Tochigi, 321-0293, Japan.
| | - Mitsuru Ishizuka
- Second Department of Surgery, Dokkyo Medical University, 880 Kitakobayashi, Mibu, Tochigi, 321-0293, Japan
| | - Kotaro Suda
- Second Department of Surgery, Dokkyo Medical University, 880 Kitakobayashi, Mibu, Tochigi, 321-0293, Japan
| | - Norisuke Shibuya
- Second Department of Surgery, Dokkyo Medical University, 880 Kitakobayashi, Mibu, Tochigi, 321-0293, Japan
| | - Hiroyuki Hachiya
- Second Department of Surgery, Dokkyo Medical University, 880 Kitakobayashi, Mibu, Tochigi, 321-0293, Japan
| | - Yukihiro Iso
- Second Department of Surgery, Dokkyo Medical University, 880 Kitakobayashi, Mibu, Tochigi, 321-0293, Japan
| | - Kazutoshi Takagi
- Second Department of Surgery, Dokkyo Medical University, 880 Kitakobayashi, Mibu, Tochigi, 321-0293, Japan
| | - Taku Aoki
- Second Department of Surgery, Dokkyo Medical University, 880 Kitakobayashi, Mibu, Tochigi, 321-0293, Japan
| | - Keiichi Kubota
- Second Department of Surgery, Dokkyo Medical University, 880 Kitakobayashi, Mibu, Tochigi, 321-0293, Japan
| |
Collapse
|
21
|
Colombo G, Gelardi ELM, Balestrero FC, Moro M, Travelli C, Genazzani AA. Insight Into Nicotinamide Adenine Dinucleotide Homeostasis as a Targetable Metabolic Pathway in Colorectal Cancer. Front Pharmacol 2021; 12:758320. [PMID: 34880756 PMCID: PMC8645963 DOI: 10.3389/fphar.2021.758320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Accepted: 11/02/2021] [Indexed: 11/13/2022] Open
Abstract
Tumour cells modify their cellular metabolism with the aim to sustain uncontrolled proliferation. Cancer cells necessitate adequate amounts of NAD and NADPH to support several enzymes that are usually overexpressed and/or overactivated. Nicotinamide adenine dinucleotide (NAD) is an essential cofactor and substrate of several NAD-consuming enzymes, such as PARPs and sirtuins, while NADPH is important in the regulation of the redox status in cells. The present review explores the rationale for targeting the key enzymes that maintain the cellular NAD/NADPH pool in colorectal cancer and the enzymes that consume or use NADP(H).
Collapse
Affiliation(s)
- Giorgia Colombo
- Department of Pharmaceutical Sciences, Università Del Piemonte Orientale, Novara, Italy
| | | | | | - Marianna Moro
- Department of Pharmaceutical Sciences, Università Del Piemonte Orientale, Novara, Italy
| | - Cristina Travelli
- Department of Drug Sciences, Università Degli Studi di Pavia, Pavia, Italy
| | - Armando A. Genazzani
- Department of Pharmaceutical Sciences, Università Del Piemonte Orientale, Novara, Italy
| |
Collapse
|
22
|
TGF-β orchestrates the phenotype and function of monocytic myeloid-derived suppressor cells in colorectal cancer. Cancer Immunol Immunother 2021; 71:1583-1596. [PMID: 34727230 PMCID: PMC9188538 DOI: 10.1007/s00262-021-03081-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Accepted: 10/01/2021] [Indexed: 11/06/2022]
Abstract
Background Monocytic myeloid-derived suppressor cells (M-MDSCs) are significantly expanded in the blood of colorectal cancer (CRC) patients. However, their presence and underlying mechanisms in the tumour microenvironment of CRC have not been examined in detail. Methods Tumour tissues and peripheral blood from CRC patients were analysed for the presence of M-MDSCs. The mechanisms of suppression were analysed by blocking pathways by which MDSCs abrogate T cell proliferation. Co-culture of CRC cells with monocytes were performed with and without cytokine blocking antibodies to determine the mechanism by which CRC cells polarise monocytes. Multi-spectral IHC was used to demonstrate the intra-tumoral location of M-MDSCs. Results Tumour tissues and blood of CRC patients contain M-MDSCs which inhibit T cell proliferation. Whilst inhibition of arginase and nitric oxide synthase 2 fail to rescue T cell proliferation, blockade of IL-10 released by these HLA-DR− cells abrogates the suppresivity of M-MDSCs. Tumour conditioned media (TCM) significantly reduces HLA-DR expression, increases IL-10 release from monocytes and causes them to become suppressive. TGF-β is highly expressed in the TCM and accumulates in the plasma. TGF-β reduces HLA-DR expression and drives monocyte immunosuppressivity. The invasive margin of CRC is enriched in CD14+ HLA-DR− cells in close proximity to T cells. Conclusions Our study demonstrates the cross-talk between CRC cells, M-MDSCs and T cells. Characterisation of CRC M-MDSCs point to therapeutic avenues to target these cells in addition to TGF-β blockade. Supplementary Information The online version contains supplementary material available at 10.1007/s00262-021-03081-5.
Collapse
|
23
|
Ding Z, He Y, Fu Y, Zhu N, Zhao M, Song Y, Huang X, Chen S, Yang Y, Zhang C, Hu Q, Ni Y, Ding L. CD38 Multi-Functionality in Oral Squamous Cell Carcinoma: Prognostic Implications, Immune Balance, and Immune Checkpoint. Front Oncol 2021; 11:687430. [PMID: 34211854 PMCID: PMC8239289 DOI: 10.3389/fonc.2021.687430] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Accepted: 05/27/2021] [Indexed: 01/09/2023] Open
Abstract
Background CD38 belongs to the ribosyl cyclase family and is expressed on various hematological cells and involved in immunosuppression and tumor promotion. Although targeting CD38 antibodies has been approved for treatment of multiple myeloma, the function of CD38 in solid tumor, oral squamous cell carcinoma (OSCC) etc., has not been investigated. Methods This retrospective study included 92 OSCC samples and analyzed the spatial distribution of CD38 by immunohistochemistry (IHC). The values of diagnosis and prognosis of CD38 were evaluated. Additionally, 53 OSCC preoperative peripheral blood samples were used to be analyzed by flow cytometry. Tumor Immune Estimation Resource (TIMER) and cBioPortal databases were used to study CD38 level in various tumors and its correlation with tumor immune microenvironment in head and neck squamous cell carcinoma (HNSCC). Results CD38 ubiquitously presented in tumor cells (TCs), fibroblast-like cells (FLCs), and tumor-infiltrating lymphocytes (TILs). Patients with highly expressed CD38 in TCs (CD38TCs) had higher TNM stage and risk of lymph node metastasis. Upregulation of CD38 in FLCs (CD38FLCs) was significantly associated with poor WPOI. Escalated CD38 in TILs (CD38TILs) led to higher Ki-67 level of tumor cells. Moreover, patients with enhanced CD38TCs were susceptible to postoperative metastasis occurrence, and those with highly expressed CD38TILs independently predicted shorter overall and disease-free survival. Strikingly, patients with highly expressed CD38TILs, but not CD38TCs and CD38FLCs, had significantly lower CD3+CD4+ T cells and higher ratio of CD3-CD16+CD56+NK cells. The imbalance of immune system is attributed to dysregulated immune checkpoint molecules (VISTA, PD-1, LAG-3, CTLA-4, TIGIT, GITR) as well as particular immune cell subsets, which were positively correlated with CD38 expression in HNSCC. Conclusion CD38 is a poor prognostic biomarker for OSCC patients and plays a vital role in governing immune microenvironment and circulating lymphocyte homeostasis. Co-expression between CD38 and immune checkpoint molecules provides new insight into immune checkpoint therapy.
Collapse
Affiliation(s)
- Zhuang Ding
- Central Laboratory of Stomatology, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China
| | - Yijia He
- Central Laboratory of Stomatology, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China
| | - Yong Fu
- Central Laboratory of Stomatology, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China
| | - Nisha Zhu
- Central Laboratory of Stomatology, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China
| | - Mengxiang Zhao
- Central Laboratory of Stomatology, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China
| | - Yuxian Song
- Central Laboratory of Stomatology, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China
| | - Xiaofeng Huang
- Department of Oral Pathology, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China
| | - Sheng Chen
- Department of Oral Pathology, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China
| | - Yan Yang
- Department of Oral Pathology, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China
| | - Caihong Zhang
- Research Institute of Superconductor Electronics, School of Electronic Science and Engineering, Nanjing University, Nanjing, China
| | - Qingang Hu
- Central Laboratory of Stomatology, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China
| | - Yanhong Ni
- Central Laboratory of Stomatology, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China
| | - Liang Ding
- Central Laboratory of Stomatology, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China
| |
Collapse
|
24
|
Ge Y, Cheng D, Jia Q, Xiong H, Zhang J. Mechanisms Underlying the Role of Myeloid-Derived Suppressor Cells in Clinical Diseases: Good or Bad. Immune Netw 2021; 21:e21. [PMID: 34277111 PMCID: PMC8263212 DOI: 10.4110/in.2021.21.e21] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Revised: 05/16/2021] [Accepted: 05/18/2021] [Indexed: 12/24/2022] Open
Abstract
Myeloid-derived suppressor cells (MDSCs) have strong immunosuppressive activity and are morphologically similar to conventional monocytes and granulocytes. The development and classification of these cells have, however, been controversial. The activation network of MDSCs is relatively complex, and their mechanism of action is poorly understood, creating an avenue for further research. In recent years, MDSCs have been found to play an important role in immune regulation and in effectively inhibiting the activity of effector lymphocytes. Under certain conditions, particularly in the case of tissue damage or inflammation, MDSCs play a leading role in the immune response of the central nervous system. In cancer, however, this can lead to tumor immune evasion and the development of related diseases. Under cancerous conditions, tumors often alter bone marrow formation, thus affecting progenitor cell differentiation, and ultimately, MDSC accumulation. MDSCs are important contributors to tumor progression and play a key role in promoting tumor growth and metastasis, and even reduce the efficacy of immunotherapy. Currently, a number of studies have demonstrated that MDSCs play a key regulatory role in many clinical diseases. In light of these studies, this review discusses the origin of MDSCs, the mechanisms underlying their activation, their role in a variety of clinical diseases, and their function in immune response regulation.
Collapse
Affiliation(s)
- Yongtong Ge
- Institute of Immunology and Molecular Medicine, Basic Medical School, Jining Medical University, Jining 272067, China
| | - Dalei Cheng
- Institute of Immunology and Molecular Medicine, Basic Medical School, Jining Medical University, Jining 272067, China
| | - Qingzhi Jia
- Affiliated Hospital of Jining Medical College, Jining Medical University, Jining 272067, China
| | - Huabao Xiong
- Institute of Immunology and Molecular Medicine, Basic Medical School, Jining Medical University, Jining 272067, China
| | - Junfeng Zhang
- Institute of Immunology and Molecular Medicine, Basic Medical School, Jining Medical University, Jining 272067, China
| |
Collapse
|
25
|
Lampis A, Ratti M, Ghidini M, Mirchev MB, Okuducu AF, Valeri N, Hahne JC. Challenges and perspectives for immunotherapy in oesophageal cancer: A look to the future (Review). Int J Mol Med 2021; 47:97. [PMID: 33846775 PMCID: PMC8041478 DOI: 10.3892/ijmm.2021.4930] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Accepted: 02/25/2021] [Indexed: 12/16/2022] Open
Abstract
Oesophageal cancer is one of the most aggressive malignancies with limited treatment options, thus resulting in a high morbidity and mortality. With 5‑year survival rates of only 5‑10%, oesophageal cancer holds a dismal prognosis for patients. In order to improve overall survival, the early diagnosis and tools for patient stratification for personalized treatment are urgent needs. A minority of oesophageal cancers belong to the spectrum of Lynch syndrome‑associated cancers and are characterized by microsatellite instability (MSI). Microsatellite instability is a consequence of defective mismatch repair protein functions and it has been well characterized in other gastrointestinal tumours, such as colorectal and gastric cancer. In the latter, high levels of MSI are associated with a better prognosis and with an increased benefit to immune‑based therapies. Therefore, similar therapeutic approaches could offer an opportunity of treatment for oesophageal cancer patients with MSI. Apart from immune checkpoint inhibitors, other immunotherapies such as adoptive T‑cell transfer, peptide vaccine and oncolytic viruses are under investigation in oesophageal cancer patients. In the present review, the rationale and current knowledge about immunotherapies in oesophageal cancer are summarised.
Collapse
Affiliation(s)
- Andrea Lampis
- Division of Molecular Pathology, The Institute of Cancer Research, Sutton SM25NG, UK
- Centre for Evolution and Cancer, The Institute of Cancer Research, Sutton SM25NG, UK
| | - Margherita Ratti
- Division of Molecular Pathology, The Institute of Cancer Research, Sutton SM25NG, UK
- Centre for Evolution and Cancer, The Institute of Cancer Research, Sutton SM25NG, UK
- Medical Department, Division of Oncology, Hospital Trust of Cremona, I-26100 Cremona, Italy
| | - Michele Ghidini
- Division of Medical Oncology, Hospital Policlinic 'Fondazione IRCCS Ca' Granda Ospedale Maggiore', I-20122 Milan, Italy
| | - Milko B. Mirchev
- Clinic of Gastroenterology, Medical University, 9002 Varna, Bulgaria
| | | | - Nicola Valeri
- Division of Molecular Pathology, The Institute of Cancer Research, Sutton SM25NG, UK
- Centre for Evolution and Cancer, The Institute of Cancer Research, Sutton SM25NG, UK
- Department of Medicine, The Royal Marsden NHS Foundation Trust, Sutton SM25NG, UK
| | - Jens Claus Hahne
- Division of Molecular Pathology, The Institute of Cancer Research, Sutton SM25NG, UK
- Centre for Evolution and Cancer, The Institute of Cancer Research, Sutton SM25NG, UK
| |
Collapse
|
26
|
Sharma V, Aggarwal A, Jacob J, Sahni D. Myeloid-derived suppressor cells: Bridging the gap between inflammation and pancreatic adenocarcinoma. Scand J Immunol 2021; 93:e13021. [PMID: 33455004 DOI: 10.1111/sji.13021] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 01/09/2021] [Accepted: 01/10/2021] [Indexed: 12/29/2022]
Abstract
Pancreatic cancer has been identified as one of the deadliest malignancies because it remains asymptomatic and usually presents in the advanced stage. Tumour immune evasion is a well-known mechanism of tumorigenesis in various forms of human malignancies. Chronic inflammation via complex networking of various inflammatory cytokines in the local tissue microenvironment dysregulates the immune system and support tumour development. Pro-inflammatory mediators present in the tumour microenvironment increase the tumour burden by causing immune suppression through the generation of myeloid-derived suppressor cells (MDSCs) and T regulatory cells. These cells, along-with myofibroblasts, create a highly immunosuppressive and resistant tumour microenvironment and are thus considered as one of the culprits for the failure of anti-cancer chemotherapies in pancreatic adenocarcinoma patients. Targeting these MDSCs using various combinatorial approaches might have the potential for abrogating the resistance and suppressive nature of the pancreatic tumour microenvironment. Therefore, there is more curiosity in studying the crosstalk of MDSCs with other immune cells during pathological conditions and the underlying mechanisms of immunosuppression in the current scenario. In this article, the possible role of MDSCs in inflammation-mediated tumour progression of pancreatic adenocarcinoma has been discussed.
Collapse
Affiliation(s)
- Vinit Sharma
- Department of Anatomy, Post Graduate Institute of Medical Education and Research (PGIMER), Chandigarh, India
| | - Anjali Aggarwal
- Department of Anatomy, Post Graduate Institute of Medical Education and Research (PGIMER), Chandigarh, India
| | - Justin Jacob
- Department of Anatomy, Post Graduate Institute of Medical Education and Research (PGIMER), Chandigarh, India
| | - Daisy Sahni
- Department of Anatomy, Post Graduate Institute of Medical Education and Research (PGIMER), Chandigarh, India
| |
Collapse
|
27
|
Yan X, Xie Y, Yang F, Hua Y, Zeng T, Sun C, Yang M, Huang X, Wu H, Fu Z, Li W, Jiao S, Yin Y. Comprehensive description of the current breast cancer microenvironment advancements via single-cell analysis. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2021; 40:142. [PMID: 33906694 PMCID: PMC8077685 DOI: 10.1186/s13046-021-01949-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Accepted: 04/15/2021] [Indexed: 02/07/2023]
Abstract
Breast cancer is a heterogeneous disease with a complex microenvironment consisting of tumor cells, immune cells, fibroblasts and vascular cells. These cancer-associated cells shape the tumor microenvironment (TME) and influence the progression of breast cancer and the therapeutic responses in patients. The exact composition of the intra-tumoral cells is mixed as the highly heterogeneous and dynamic nature of the TME. Recent advances in single-cell technologies such as single-cell DNA sequencing (scDNA-seq), single-cell RNA sequencing (scRNA-seq) and mass cytometry have provided new insights into the phenotypic and functional diversity of tumor-infiltrating cells in breast cancer. In this review, we have outlined the recent progress in single-cell characterization of breast tumor ecosystems, and summarized the phenotypic diversity of intra-tumoral cells and their potential prognostic relevance.
Collapse
Affiliation(s)
- Xueqi Yan
- Department of Oncology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Yinghong Xie
- Department of Oncology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Fan Yang
- Department of Oncology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Yijia Hua
- Department of Oncology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Tianyu Zeng
- Department of Oncology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Chunxiao Sun
- Department of Oncology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Mengzhu Yang
- Department of Oncology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Xiang Huang
- Department of Oncology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Hao Wu
- Department of Oncology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Ziyi Fu
- Department of Oncology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Wei Li
- Department of Oncology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China.
| | - Shiping Jiao
- Department of Hepatobiliary Surgery, the Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, 210029, Jiangsu Province, China. .,Drum Tower Institute of clinical medicine, Nanjing University, Nanjing, 210029, Jiangsu Province, China.
| | - Yongmei Yin
- Department of Oncology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China. .,Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Personalized Cancer Medicine, Nanjing Medical University, Nanjing, 211166, China.
| |
Collapse
|
28
|
Corti F, Lonardi S, Intini R, Salati M, Fenocchio E, Belli C, Borelli B, Brambilla M, Prete AA, Quarà V, Antista M, Fassan M, Morano F, Spallanzani A, Ambrosini M, Curigliano G, de Braud F, Zagonel V, Fucà G, Pietrantonio F. The Pan-Immune-Inflammation Value in microsatellite instability-high metastatic colorectal cancer patients treated with immune checkpoint inhibitors. Eur J Cancer 2021; 150:155-167. [PMID: 33901794 DOI: 10.1016/j.ejca.2021.03.043] [Citation(s) in RCA: 56] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 03/10/2021] [Accepted: 03/18/2021] [Indexed: 01/26/2023]
Abstract
BACKGROUND Immune checkpoint inhibitors (ICIs) yielded unprecedented efficacy in patients with microsatellite instability (MSI)-high metastatic colorectal cancer (mCRC). Since the Pan-Immune-Inflammation Value (PIV) is a blood-based biomarker with prognostic usefulness in mCRC, it might predict clinical outcomes and primary resistance to ICIs. METHODS We retrospectively analysed the association of PIV and its early modulation at 3/4 weeks after treatment initiation with the outcomes of MSI-high mCRC patients receiving anti-programmed death-(ligand)1 (PD-[L]1) +/- anti-cytotoxic T-lymphocyte antigen 4 (CTLA-4) agents. PIV was calculated as follows: (neutrophil count × platelet count × monocyte count)/lymphocyte count. PIV cut-offs were determined using the maximally selected rank statistics. RESULTS A total of 163 patients were included. In the multivariable models for overall survival (OS) and progression-free survival (PFS), both high (>492) baseline PIV (OS: adjusted [a] HR, 3.00; 95% CI, 1.49-6.04, p = 0.002; PFS: aHR, 1.91; 95% CI, 1.06-3.44, p = 0.031) and early PIV increase ≥+30% (OS: aHR, 3.21; 95% CI, 1.65-6.23, p < 0.001; PFS: aHR, 2.25; 95% CI, 1.30-3.89, p = 0.003) confirmed an independent prognostic impact. After stratifying patients according to baseline PIV and ICI regimen, OS and PFS were significantly worse in subjects with high PIV receiving anti-PD-1/PD-L1 monotherapy. Early PIV increase was an independent predictor of clinical benefit (aOR, 0.23; 95% CI, 0.08-0.66; p = 0.007), whereas a trend was observed for baseline PIV (aOR, 0.33; 95% CI, 0.10-1.07; p = 0.065). CONCLUSION PIV is a strong predictor of outcomes in MSI-high mCRC patients receiving ICIs. Prospective validation of these results is required to establish its role as a stratification factor for personalised combination strategies.
Collapse
Affiliation(s)
- Francesca Corti
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Giacomo Venezian 1, 20133, Milan, Italy.
| | - Sara Lonardi
- Department of Oncology, Istituto Oncologico Veneto, IRCCS, Via Gattamelata, 64, 35128, Padua, Italy.
| | - Rossana Intini
- Department of Oncology, Istituto Oncologico Veneto, IRCCS, Via Gattamelata, 64, 35128, Padua, Italy.
| | - Massimiliano Salati
- Department of Oncology and Hematology, University Hospital of Modena, Via del Pozzo, 71, 41125, Modena, Italy; PhD Program in Clinical and Experimental Medicine, University of Modena and Reggio Emilia, Via Università, 4, 41121, Modena Italy.
| | - Elisabetta Fenocchio
- Multidisciplinary Outpatient Oncology Clinic, Candiolo Cancer Institute, FPO-IRCCS, Strada Provinciale 142, km 3.95, 10060 Candiolo (TO), Italy.
| | - Carmen Belli
- Division of Early Drugs Development for Innovative Therapies, Istituto Europeo di Oncologia, IRCCS, Via Ripamonti 435, 20141, Milan, Italy.
| | - Beatrice Borelli
- Unit of Medical Oncology, Azienda Ospedaliero-Universitaria Pisana, Via Roma 67, 56126, Pisa, Italy.
| | - Marta Brambilla
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Giacomo Venezian 1, 20133, Milan, Italy.
| | - Alessandra A Prete
- Department of Oncology, Istituto Oncologico Veneto, IRCCS, Via Gattamelata, 64, 35128, Padua, Italy.
| | - Virginia Quarà
- Department of Oncology, University of Turin, Via Giuseppe Verdi, 8, 10124, Torino, Italy.
| | - Maria Antista
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Giacomo Venezian 1, 20133, Milan, Italy.
| | - Matteo Fassan
- Surgical Pathology Unit, Department of Medicine (DIMED), University of Padua, Via Giustiniani 2, 35128, Padua, Italy.
| | - Federica Morano
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Giacomo Venezian 1, 20133, Milan, Italy.
| | - Andrea Spallanzani
- Department of Oncology and Hematology, University Hospital of Modena, Via del Pozzo, 71, 41125, Modena, Italy.
| | - Margherita Ambrosini
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Giacomo Venezian 1, 20133, Milan, Italy.
| | - Giuseppe Curigliano
- Division of Early Drugs Development for Innovative Therapies, Istituto Europeo di Oncologia, IRCCS, Via Ripamonti 435, 20141, Milan, Italy; Oncology and Hemato-oncology Department, University of Milan, via Festa del Perdono 7, 20122, Milan, Italy.
| | - Filippo de Braud
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Giacomo Venezian 1, 20133, Milan, Italy; Oncology and Hemato-oncology Department, University of Milan, via Festa del Perdono 7, 20122, Milan, Italy.
| | - Vittorina Zagonel
- Department of Oncology, Istituto Oncologico Veneto, IRCCS, Via Gattamelata, 64, 35128, Padua, Italy.
| | - Giovanni Fucà
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Giacomo Venezian 1, 20133, Milan, Italy.
| | - Filippo Pietrantonio
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Giacomo Venezian 1, 20133, Milan, Italy; Oncology and Hemato-oncology Department, University of Milan, via Festa del Perdono 7, 20122, Milan, Italy.
| |
Collapse
|
29
|
Frosch J, Leontari I, Anderson J. Combined Effects of Myeloid Cells in the Neuroblastoma Tumor Microenvironment. Cancers (Basel) 2021; 13:1743. [PMID: 33917501 PMCID: PMC8038814 DOI: 10.3390/cancers13071743] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 04/02/2021] [Accepted: 04/02/2021] [Indexed: 12/13/2022] Open
Abstract
Despite multimodal treatment, survival chances for high-risk neuroblastoma patients remain poor. Immunotherapeutic approaches focusing on the activation and/or modification of host immunity for eliminating tumor cells, such as chimeric antigen receptor (CAR) T cells, are currently in development, however clinical trials have failed to reproduce the preclinical results. The tumor microenvironment is emerging as a major contributor to immune suppression and tumor evasion in solid cancers and thus has to be overcome for therapies relying on a functional immune response. Among the cellular components of the neuroblastoma tumor microenvironment, suppressive myeloid cells have been described as key players in inhibition of antitumor immune responses and have been shown to positively correlate with more aggressive disease, resistance to treatments, and overall poor prognosis. This review article summarizes how neuroblastoma-driven inflammation induces suppressive myeloid cells in the tumor microenvironment and how they in turn sustain the tumor niche through suppressor functions, such as nutrient depletion and generation of oxidative stress. Numerous preclinical studies have suggested a range of drug and cellular therapy approaches to overcome myeloid-derived suppression in neuroblastoma that warrant evaluation in future clinical studies.
Collapse
Affiliation(s)
| | | | - John Anderson
- UCL Institute of Child Health, Developmental Biology and Cancer Section, University College London, London WC1N 1EH, UK; (J.F.); (I.L.)
| |
Collapse
|
30
|
Cui C, Lan P, Fu L. The role of myeloid-derived suppressor cells in gastrointestinal cancer. Cancer Commun (Lond) 2021; 41:442-471. [PMID: 33773092 PMCID: PMC8211353 DOI: 10.1002/cac2.12156] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 02/09/2021] [Accepted: 03/10/2021] [Indexed: 12/24/2022] Open
Abstract
Gastrointestinal (GI) cancer encompasses a range of malignancies that originate in the digestive system, which together represent the most common form of cancer diagnosed worldwide. However, despite numerous advances in both diagnostics and treatment, the incidence and mortality rate of GI cancer are on the rise. Myeloid-derived suppressor cells (MDSCs) are a heterogeneous population of immature myeloid cells that increase in number under certain pathological conditions, such as infection and inflammation, and this expansion is of particular relevance to cancer. MDSCs are heavily involved in the regulation of the immune system and act to dampen its response to tumors, favoring the escape of tumor cells from immunosurveillance and increasing both metastasis and recurrence. Several recent studies have supported the use of MDSCs as a prognostic and predictive biomarker in patients with cancer, and potentially as a novel treatment target. In the present review, the mechanisms underlying the immunosuppressive functions of MDSCs are described, and recent researches concerning the involvement of MDSCs in the progression, prognosis, and therapies of GI cancer are reviewed. The aim of this work was to present the development of novel treatments targeting MDSCs in GI cancer in the hope of improving outcomes for patients with this condition.
Collapse
Affiliation(s)
- Cheng Cui
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Department of Pharmacology and International Cancer Centre, Shenzhen University School of Medicine, Shenzhen, Guangdong, 518055, P. R. China
| | - Penglin Lan
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Department of Pharmacology and International Cancer Centre, Shenzhen University School of Medicine, Shenzhen, Guangdong, 518055, P. R. China
| | - Li Fu
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Department of Pharmacology and International Cancer Centre, Shenzhen University School of Medicine, Shenzhen, Guangdong, 518055, P. R. China
| |
Collapse
|
31
|
Guo C, Crespo M, Gurel B, Dolling D, Rekowski J, Sharp A, Petremolo A, Sumanasuriya S, Rodrigues DN, Ferreira A, Pereira R, Figueiredo I, Mehra N, Lambros MBK, Neeb A, Gil V, Seed G, Terstappen L, Alimonti A, Drake CG, Yuan W, de Bono JS. CD38 in Advanced Prostate Cancers. Eur Urol 2021; 79:736-746. [PMID: 33678520 PMCID: PMC8175332 DOI: 10.1016/j.eururo.2021.01.017] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Accepted: 01/12/2021] [Indexed: 12/13/2022]
Abstract
Background CD38, a druggable ectoenzyme, is involved in the generation of adenosine, which is implicated in tumour immune evasion. Its expression and role in prostate tumour-infiltrating immune cells (TIICs) have not been elucidated. Objective To characterise CD38 expression on prostate cancer (PC) epithelial cells and TIICs, and to associate this expression with clinical outcomes. Design, setting, and participants RNAseq from 159 patients with metastatic castration-resistant prostate cancer (mCRPC) in the International Stand Up To Cancer/Prostate Cancer Foundation (SU2C/PCF) cohort and 171 mCRPC samples taken from 63 patients in the Fred Hutchinson Cancer Research Centre cohort were analysed. CD38 expression was immunohistochemically scored by a validated assay on 51 castration-resistant PC (CRPC) and matching, same-patient castration-sensitive PC (CSPC) biopsies obtained between 2016 and 2018, and was associated with retrospectively collected clinical data. Outcome measurements and statistical analysis mCRPC transcriptomes were analysed for associations between CD38 expression and gene expression signatures. Multiplex immunofluorescence determined CD38 expression in PC biopsies. Differences in CD38+ TIIC densities between CSPC and CRPC biopsies were analysed using a negative binomial mixed model. Differences in the proportions of CD38+ epithelial cells between non-matched benign prostatic epithelium and PC were compared using Fisher’s exact test. Differences in the proportions of biopsies containing CD38+ tumour epithelial cells between matched CSPC and CRPC biopsies were compared by McNemar’s test. Univariable and multivariable survival analyses were performed using Cox regression models. Results and limitations CD38 mRNA expression in mCRPC was most significantly associated with upregulated immune signalling pathways. CD38 mRNA expression was associated with interleukin (IL)-12, IL-23, and IL-27 signalling signatures as well as immunosuppressive adenosine signalling and T cell exhaustion signatures. CD38 protein was frequently expressed on phenotypically diverse TIICs including B cells and myeloid cells, but largely absent from tumour epithelial cells. CD38+ TIIC density increased with progression to CRPC and was independently associated with worse overall survival. Future studies are required to dissect TIIC CD38 function. Conclusions CD38+ prostate TIICs associate with worse survival and immunosuppressive mechanisms. The role of CD38 in PC progression warrants investigation as insights into its functions may provide rationale for CD38 targeting in lethal PC. Patient summary CD38 is expressed on the surface of white blood cells surrounding PC cells. These cells may impact PC growth and treatment resistance. Patients with PC with more CD38-expressing white blood cells are more likely to die earlier.
Collapse
Affiliation(s)
- Christina Guo
- The Institute of Cancer Research, London, UK; The Royal Marsden NHS Foundation Trust, Sutton, UK
| | | | - Bora Gurel
- The Institute of Cancer Research, London, UK
| | | | | | - Adam Sharp
- The Institute of Cancer Research, London, UK; The Royal Marsden NHS Foundation Trust, Sutton, UK
| | | | - Semini Sumanasuriya
- The Institute of Cancer Research, London, UK; The Royal Marsden NHS Foundation Trust, Sutton, UK
| | - Daniel N Rodrigues
- The Institute of Cancer Research, London, UK; The Royal Marsden NHS Foundation Trust, Sutton, UK
| | | | | | | | - Niven Mehra
- The Institute of Cancer Research, London, UK
| | | | - Antje Neeb
- The Institute of Cancer Research, London, UK
| | | | - George Seed
- The Institute of Cancer Research, London, UK
| | | | - Andrea Alimonti
- Institute of Oncology Research, Bellinzona, Switzerland; Faculty of Biomedical Sciences, Università della Svizzera Italiana, Lugano, Switzerland; Department of Medicine, University of Padova, Padova, Italy; Veneto Institute of Molecular Medicine, Padova, Italy
| | | | - Wei Yuan
- The Institute of Cancer Research, London, UK
| | - Johann S de Bono
- The Institute of Cancer Research, London, UK; The Royal Marsden NHS Foundation Trust, Sutton, UK.
| | | |
Collapse
|
32
|
Yin K, Xia X, Rui K, Wang T, Wang S. Myeloid-Derived Suppressor Cells: A New and Pivotal Player in Colorectal Cancer Progression. Front Oncol 2020; 10:610104. [PMID: 33384962 PMCID: PMC7770157 DOI: 10.3389/fonc.2020.610104] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Accepted: 11/10/2020] [Indexed: 12/12/2022] Open
Abstract
Colorectal cancer (CRC) remains a devastating human malignancy with poor prognosis. Of the various factors, immune evasion mechanisms play pivotal roles in CRC progression and impede the effects of cancer therapy. Myeloid-derived suppressor cells (MDSCs) constitute an immature population of myeloid cells that are typical during tumor progression. These cells have the ability to induce strong immunosuppressive effects within the tumor microenvironment (TME) and promote CRC development. Indeed, MDSCs have been shown to accumulate in both tumor-bearing mice and CRC patients, and may therefore become an obstacle for cancer immunotherapy. Consequently, numerous studies have focused on the characterization of MDSCs and their immunosuppressive capacity, as well as developing novel approaches to suppress MDSCs function with different approaches. Current therapeutic strategies that target MDSCs in CRC include inhibition of their recruitment and alteration of their function, alone or in combination with other therapies including chemotherapy, radiotherapy and immunotherapy. Herein, we summarize the recent roles and mechanisms of MDSCs in CRC progression. In addition, a brief review of MDSC-targeting approaches for potential CRC therapy is presented.
Collapse
Affiliation(s)
- Kai Yin
- Department of General Surgery, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Xueli Xia
- Department of Immunology, Jiangsu Key Laboratory of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Ke Rui
- Department of Laboratory Medicine, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Tingting Wang
- Department of Laboratory Medicine, Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi Children's Hospital, Wuxi, China
| | - Shengjun Wang
- Department of Immunology, Jiangsu Key Laboratory of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, China
| |
Collapse
|
33
|
Feng J, Chen S, Li S, Wu B, Lu J, Tan L, Li J, Song Y, Shi G, Shi YG, Jiang J. The association between monocytic myeloid-derived suppressor cells levels and the anti-tumor efficacy of anti-PD-1 therapy in NSCLC patients. Transl Oncol 2020; 13:100865. [PMID: 32920330 PMCID: PMC7492992 DOI: 10.1016/j.tranon.2020.100865] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 07/24/2020] [Accepted: 08/03/2020] [Indexed: 12/26/2022] Open
Abstract
Monocytic myeloid-derived suppressor cells (M-MDSCs), granulocytic MDSC (G-MDSCs) and regulatory T cells (Tregs) inhibit adaptive anti-tumor immunity and undermine the efficacy of anti-PD-1 therapy. However, the impact of anti-PD-1 treatment on these immunosuppressive cells has not been clearly defined in non-small cell lung cancer (NSCLC). In this retrospective study, 27 advanced NSCLC patients were divided into partial response (PR), stable disease (SD), and progressive disease (PD) groups. The impact of anti-PD-1 therapy on circulating Tregs, G-MDSCs, and M-MDSCs was assessed by flow cytometer. Here, we found that anti-PD-1 treatment boosted circulating Tregs levels, which presented the most remarkable augment during the first two therapeutic cycles, in NSCLC patients. In contrast, anti-PD-1 therapy did not overall change G-MDSCs and M-MDSCs levels. However, the PR group had a higher baseline level of M-MDSCs, which exhibited a significant decrease after the first cycle of anti-PD-1 treatment. Besides, M-MDSCs levels in the PR group were maintained at a low level in the following therapeutic cycles. Consistently, Tregs levels robustly increased in the syngeneic tumor model after anti-mouse PD-1 Ab treatment. Accordingly, M-MDSCs neutralization by anti-mouse ly6c Ab enhanced the anti-tumor efficacy of anti-PD-1 therapy in mice. Finally, the decreased M-MDSCs levels were associated with the enhanced effector CD8+ T cells expansion in the PR group and mice. In conclusion, anti-PD-1 therapy upregulates Tregs levels in NSCLC patients, and the M-MDSC levels are associated with the anti-tumor efficacy of anti-PD-1 treatment. Neutralization of M-MDSCs may be a promising option to augment anti-PD-1 therapy efficacy in NSCLC. Anti-PD-1 therapy upregulates Tregs levels in NSCLC patients and mouse model. M-MDSCs exhibit a significant decrease in the PR group after anti-PD-1 therapy. M-MDSCs neutralization augments anti-PD-1 efficacy in mouse model. The increased effector CD8+ T cells are associated with the better anti-PD-1 efficacy in the PR group and mouse model.
Collapse
Affiliation(s)
- Jiuxing Feng
- Key Laboratory of Medical Epigenetics and Metabolism, Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China
| | - Shujing Chen
- Department of Pulmonary and Critical Care Medicine, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Shuangqi Li
- Key Laboratory of Medical Epigenetics and Metabolism, Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China
| | - Baitong Wu
- School of Medicine, Tongji University, Shanghai 200120, China
| | - Jiacheng Lu
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Li Tan
- Key Laboratory of Medical Epigenetics and Metabolism, Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China
| | - Jiamin Li
- Department of Pulmonary and Critical Care Medicine, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Yuanlin Song
- Department of Pulmonary and Critical Care Medicine, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Guoming Shi
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Yujiang Geno Shi
- Key Laboratory of Medical Epigenetics and Metabolism, Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China; Division of Endocrinology, Diabetes and Hypertension, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA.
| | - Jinjun Jiang
- Department of Pulmonary and Critical Care Medicine, Zhongshan Hospital, Fudan University, Shanghai 200032, China.
| |
Collapse
|
34
|
Cassetta L, Bruderek K, Skrzeczynska-Moncznik J, Osiecka O, Hu X, Rundgren IM, Lin A, Santegoets K, Horzum U, Godinho-Santos A, Zelinskyy G, Garcia-Tellez T, Bjelica S, Taciak B, Kittang AO, Höing B, Lang S, Dixon M, Müller V, Utikal JS, Karakoç D, Yilmaz KB, Górka E, Bodnar L, Anastasiou OE, Bourgeois C, Badura R, Kapinska-Mrowiecka M, Gotic M, Ter Laan M, Kers-Rebel E, Król M, Santibañez JF, Müller-Trutwin M, Dittmer U, de Sousa AE, Esendağlı G, Adema G, Loré K, Ersvær E, Umansky V, Pollard JW, Cichy J, Brandau S. Differential expansion of circulating human MDSC subsets in patients with cancer, infection and inflammation. J Immunother Cancer 2020; 8:jitc-2020-001223. [PMID: 32907925 PMCID: PMC7481096 DOI: 10.1136/jitc-2020-001223] [Citation(s) in RCA: 119] [Impact Index Per Article: 29.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/24/2020] [Indexed: 01/25/2023] Open
Abstract
Background Myeloid-derived suppressor cells (MDSC) are a functional myeloid cell subset that includes myeloid cells with immune suppressive properties. The presence of MDSC has been reported in the peripheral blood of patients with several malignant and non-malignant diseases. So far, direct comparison of MDSC across different diseases and Centers is hindered by technical pitfalls and a lack of standardized methodology. To overcome this issue, we formed a network through the COST Action Mye-EUNITER (www.mye-euniter.eu) with the goal to standardize and facilitate the comparative analysis of human circulating MDSC in cancer, inflammation and infection. In this manuscript, we present the results of the multicenter study Mye-EUNITER MDSC Monitoring Initiative, that involved 13 laboratories and compared circulating MDSC subsets across multiple diseases, using a common protocol for the isolation, identification and characterization of these cells. Methods We developed, tested, executed and optimized a standard operating procedure for the isolation and immunophenotyping of MDSC using blood from healthy donors. We applied this procedure to the blood of almost 400 patients and controls with different solid tumors and non-malignant diseases. The latter included viral infections such as HIV and hepatitis B virus, but also psoriasis and cardiovascular disorders. Results We observed that the frequency of MDSC in healthy donors varied substantially between centers and was influenced by technical aspects such as the anticoagulant and separation method used. Expansion of polymorphonuclear (PMN)-MDSC exceeded the expansion of monocytic MDSC (M-MDSC) in five out of six solid tumors. PMN-MDSC expansion was more pronounced in cancer compared with infection and inflammation. Programmed death-ligand 1 was primarily expressed in M-MDSC and e-MDSC and was not upregulated as a consequence of disease. LOX-1 expression was confined to PMN-MDSC. Conclusions This study provides improved technical protocols and workflows for the multi-center analysis of circulating human MDSC subsets. Application of these workflows revealed a predominant expansion of PMN-MDSC in solid tumors that exceeds expansion in chronic infection and inflammation.
Collapse
Affiliation(s)
- Luca Cassetta
- MRC Centre for Reproductive Health, The University of Edinburgh The Queen's Medical Research Institute, Edinburgh, Edinburgh, UK
| | - Kirsten Bruderek
- Department of Otorhinolaryngology, University Hospital Essen, Essen, Germany
| | - Joanna Skrzeczynska-Moncznik
- Department of Immunology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Małopolska, Poland
| | - Oktawia Osiecka
- Department of Immunology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Małopolska, Poland
| | - Xiaoying Hu
- Clinical Cooperation Unit Dermato-Oncology, DKFZ, Heidelberg, Baden-Württemberg, Germany.,Department of Dermatology, Venereology and Allergology, University Medical Centre Mannheim, Mannheim, Baden-Württemberg, Germany
| | - Ida Marie Rundgren
- Department of Biomedical Laboratory Scientist Education and Chemical Engineering, Faculty of Engineering and Natural Sciences, Western Norway University of Applied Sciences, Bergen, Hordaland, Norway
| | - Ang Lin
- Division of Immunology and Allergy, Department of Medicine Solna, Karolinska Institute, Stockholm, Stockholm, Sweden.,Center for Molecular Medicine, Karolinska Institute, Stockholm, Stockholm, Sweden
| | - Kim Santegoets
- Medical Center, Radiotherapy & OncoImmunology Laboratory, Department of Radiation Oncology, Radboud University, Nijmegen, Gelderland, The Netherlands
| | - Utku Horzum
- Department of Basic Oncology, Cancer Institute, Hacettepe University, Ankara, Ankara, Turkey
| | - Ana Godinho-Santos
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, University of Lisbon, Lisboa, Lisboa, Portugal
| | - Gennadiy Zelinskyy
- Institute for Virology, University Hospital Essen, Essen, Nordrhein-Westfalen, Germany
| | - Thalia Garcia-Tellez
- HIV Inflammation and Persistence, Pasteur Institute, Paris, Île-de-France, France
| | - Sunčica Bjelica
- Department of Molecular Oncology, Institute for Medical Research, University of Belgrade, Beograd, Beograd, Serbia
| | - Bartłomiej Taciak
- Department of Cancer Biology, Institute of Biology, Warsaw University of Life Sciences, Warszawa, Poland.,Cellis AG, Zurich, Switzerland
| | | | - Benedikt Höing
- Department of Otorhinolaryngology, University Hospital Essen, Essen, Germany
| | - Stephan Lang
- Department of Otorhinolaryngology, University Hospital Essen, Essen, Germany
| | - Michael Dixon
- Edinburgh Breast Unit and Breast Cancer Now Research Unit, The University of Edinburgh, Edinburgh, Edinburgh, UK
| | - Verena Müller
- Clinical Cooperation Unit Dermato-Oncology, DKFZ, Heidelberg, Baden-Württemberg, Germany.,Department of Dermatology, Venereology and Allergology, University Medical Centre Mannheim, Mannheim, Baden-Württemberg, Germany
| | - Jochen Sven Utikal
- Department of Dermatology, Venereology and Allergology, University Medical Centre Mannheim, Mannheim, Baden-Württemberg, Germany.,Clinical Cooperation Unit Dermato-Oncology, German Cancer Research Centre, Heidelberg, Baden-Württemberg, Germany
| | - Derya Karakoç
- Department of Medical and Surgical Research, Institute of Health Sciences, Hacettepe University, Ankara, Ankara, Turkey.,Department of General Surgery, Faculty of Medicine, Hacettepe University, Ankara, Ankara, Turkey
| | - Kerim Bora Yilmaz
- Department of Medical and Surgical Research, Institute of Health Sciences, Hacettepe University, Ankara, Ankara, Turkey.,Department of General Surgery, Gulhane Egitim ve Arastirma Hastanesi, Ankara, Ankara, Turkey
| | - Emilia Górka
- Department of Cancer Biology, Institute of Biology, Warsaw University of Life Sciences, Warszawa, Poland.,Cellis AG, Zurich, Switzerland
| | - Lubomir Bodnar
- Department of Oncology and Immunooncology, Hospital Ministry of the Interior and Administration & Warmia and Masuria Oncology Centre, Olsztyn, Poland.,Department of Oncology, University of Warmia and Mazury in Olsztyn, Olsztyn, Poland
| | | | - Christine Bourgeois
- Center for Immunology of Viral Infections and Autoimmune Diseases, IDMIT Department, IBFJ, CEA, Université Paris-Sud, Saint-Aubin, Île-de-France, France
| | - Robert Badura
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, University of Lisbon, Lisboa, Lisboa, Portugal.,Serviço de Doenças Infecciosas, Northern Lisbon University Hospital Centre, Lisboa, Lisboa, Portugal
| | | | - Mirjana Gotic
- Clinic of Hematology, Clinical Center of Serbia, Beograd, Beograd, Serbia
| | - Mark Ter Laan
- Medical Center, Department of Neurosurgery, Radboud University, Nijmegen, Gelderland, The Netherlands
| | - Esther Kers-Rebel
- Medical Center, Radiotherapy & OncoImmunology Laboratory, Department of Radiation Oncology, Radboud University, Nijmegen, Gelderland, The Netherlands
| | - Magdalena Król
- Department of Cancer Biology, Institute of Biology, Warsaw University of Life Sciences, Warszawa, Poland.,Cellis AG, Zurich, Switzerland
| | - Juan Francisco Santibañez
- Department of Molecular Oncology, Institute for Medical Research, University of Belgrade, Beograd, Beograd, Serbia.,Centro Integrativo de Biología y Química Aplicada (CIBQA), Universidad Bernardo O'Higgins, Santiago, Chile
| | | | - Ulf Dittmer
- Institute for Virology, University Hospital Essen, Essen, Nordrhein-Westfalen, Germany
| | - Ana Espada de Sousa
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, University of Lisbon, Lisboa, Lisboa, Portugal
| | - Güneş Esendağlı
- Department of Basic Oncology, Cancer Institute, Hacettepe University, Ankara, Ankara, Turkey.,Department of Medical and Surgical Research, Institute of Health Sciences, Hacettepe University, Ankara, Ankara, Turkey
| | - Gosse Adema
- Department of Radiation Oncology, Radboud University Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands
| | - Karin Loré
- Division of Immunology and Allergy, Department of Medicine Solna, Karolinska Institute, Stockholm, Stockholm, Sweden.,Center for Molecular Medicine, Karolinska Institute, Stockholm, Stockholm, Sweden
| | - Elisabeth Ersvær
- Department of Biomedical Laboratory Scientist Education and Chemical Engineering, Faculty of Engineering and Natural Sciences, Western Norway University of Applied Sciences, Bergen, Hordaland, Norway
| | - Viktor Umansky
- Department of Dermatology, Venereology and Allergology, University Medical Centre Mannheim, Mannheim, Baden-Württemberg, Germany.,Clinical Cooperation Unit Dermato-Oncology, German Cancer Research Centre, Heidelberg, Baden-Württemberg, Germany
| | - Jeffrey W Pollard
- MRC Centre for Reproductive Health, The University of Edinburgh The Queen's Medical Research Institute, Edinburgh, Edinburgh, UK
| | - Joanna Cichy
- Department of Immunology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Małopolska, Poland
| | - Sven Brandau
- Department of Otorhinolaryngology, University Hospital Essen, Essen, Germany .,German Cancer Consortium, Partner Site Essen-Düsseldorf, Germany
| |
Collapse
|
35
|
Jiao Y, Yi M, Xu L, Chu Q, Yan Y, Luo S, Wu K. CD38: targeted therapy in multiple myeloma and therapeutic potential for solid cancers. Expert Opin Investig Drugs 2020; 29:1295-1308. [PMID: 32822558 DOI: 10.1080/13543784.2020.1814253] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
INTRODUCTION CD38 is expressed by some cells of hematological malignancies and tumor-related immunosuppressive cells, including regulatory T cells, regulatory B cells, and myeloid-derived suppressor cells. CD38 is an effective target in some hematological malignancies such as multiple myeloma (MM). Daratumumab (Dara), a CD38-targeting antibody, can eliminate CD38high immune suppressor cells and is regarded as a standard therapy for MM because of its outstanding clinical efficacy. Other CD38 monospecific antibodies, such as isatuximab, MOR202, and TAK079, showed promising effects in clinical trials. AREA COVERED This review examines the expression, function, and targeting of CD38 in MM and its potential to deplete immunosuppressive cells in solid cancers. We summarize the distribution and biological function of CD38 and discuss the application of anti-CD38 drugs in hematological malignancies. We also analyz the role of CD38+ immune cells in the tumor microenvironment to encourage additional investigations that target CD38 in solid cancers. PubMed and ClinicalTrials were searched to identify relevant literature from the database inception to 30 April 2020. EXPERT OPINION There is convincing evidence that CD38-targeted immunotherapeutics reduce CD38+ immune suppressor cells. This result suggests that CD38 can be exploited to treat solid tumors by regulating the immunosuppressive microenvironment.
Collapse
Affiliation(s)
- Ying Jiao
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology , Wuhan, China
| | - Ming Yi
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology , Wuhan, China
| | - Linping Xu
- Department of Medical Oncology, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital , Zhengzhou, China
| | - Qian Chu
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology , Wuhan, China
| | - Yongxiang Yan
- R & D Department, Wuhan YZY Biopharma Co., Ltd , Wuhan, China
| | - Suxia Luo
- Department of Medical Oncology, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital , Zhengzhou, China
| | - Kongming Wu
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology , Wuhan, China.,Department of Medical Oncology, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital , Zhengzhou, China
| |
Collapse
|
36
|
Davidov V, Jensen G, Mai S, Chen SH, Pan PY. Analyzing One Cell at a TIME: Analysis of Myeloid Cell Contributions in the Tumor Immune Microenvironment. Front Immunol 2020; 11:1842. [PMID: 32983100 PMCID: PMC7492293 DOI: 10.3389/fimmu.2020.01842] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Accepted: 07/09/2020] [Indexed: 12/30/2022] Open
Abstract
Tumor-mediated regulation of the host immune system involves an intricate signaling network that results in the tumor's inherent survival benefit. Myeloid cells are central in orchestrating the mechanisms by which tumors escape immune detection and continue their proliferative programming. Myeloid cell activation has historically been classified using a dichotomous system of classical (M1-like) and alternative (M2-like) states, defining general pro- and anti-inflammatory functions, respectively. Explosions in bioinformatics analyses have rapidly expanded the definitions of myeloid cell pro- and anti-inflammatory states with different combinations of tissue- and disease-specific phenotypic and functional markers. These new definitions have allowed researchers to target specific subsets of disease-propagating myeloid cells in order to modify or arrest the natural progression of the associated disease, especially in the context of tumor-immune interactions. Here, we discuss the myeloid cell contribution to solid tumor initiation and maintenance, and strategies to reprogram their phenotypic and functional fate, thereby disabling the network that benefits tumor survival.
Collapse
Affiliation(s)
- Vitaliy Davidov
- Texas A&M College of Medicine, Bryan, TX, United States.,Center for Immunotherapy Research, Cancer Center of Excellence, Houston Methodist Research Institute, Houston, TX, United States
| | - Garrett Jensen
- Texas A&M College of Medicine, Bryan, TX, United States.,Center for Immunotherapy Research, Cancer Center of Excellence, Houston Methodist Research Institute, Houston, TX, United States
| | - Sunny Mai
- Center for Immunotherapy Research, Cancer Center of Excellence, Houston Methodist Research Institute, Houston, TX, United States
| | - Shu-Hsia Chen
- Texas A&M College of Medicine, Bryan, TX, United States.,Center for Immunotherapy Research, Cancer Center of Excellence, Houston Methodist Research Institute, Houston, TX, United States
| | - Ping-Ying Pan
- Texas A&M College of Medicine, Bryan, TX, United States.,Center for Immunotherapy Research, Cancer Center of Excellence, Houston Methodist Research Institute, Houston, TX, United States
| |
Collapse
|
37
|
Hong EH, Cho J, Ahn JH, Kwon BE, Kweon MN, Seo SU, Yoon BI, Chang SY, Ko HJ. Plasmacytoid dendritic cells regulate colitis-associated tumorigenesis by controlling myeloid-derived suppressor cell infiltration. Cancer Lett 2020; 493:102-112. [PMID: 32810576 DOI: 10.1016/j.canlet.2020.08.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 07/23/2020] [Accepted: 08/03/2020] [Indexed: 12/11/2022]
Abstract
Toll-like receptor (TLR)3 and TLR7 are important for stimulating plasmacytoid dendritic cells (pDCs), which secrete type I interferon. Mice deficient for TLR3 and TLR7 (TLR3-/-TLR7-/-) reportedly exhibit deteriorated colitis because of impaired pDCs. However, the role of pDCs in tumorigenesis-associated inflammation progression has not been studied. We treated wild-type or TLR3-/-TLR7-/- mice with dextran sulfate sodium (DSS) and/or azoxymethane (AOM) and examined colon mucosa, measured body weight and colon length of mice, and examined pDC and myeloid-derived suppressor cell (MDSC) accumulation. Further, we depleted pDCs in AOM/DSS-treated wild-type mice by treating them with anti-PDCA-1 antibodies. We found that MDSCs significantly increased, while pDCs decreased in TLR3-/-TLR7-/- mice. Moreover, TLR3-/-TLR7-/- mice developed colitis-associated colon cancer following AOM/DSS treatment. Additionally, we showed that a defect in TLR7 of pDCs is responsible for the aggravation of colitis-associated colon cancer. Further, we showed that TLR7 ligand mitigates colitis-associated colon cancer. Collectively, our results demonstrate that gut pDCs play a crucial role in reducing colorectal cancer development via the regulation of infiltrating MDSCs.
Collapse
Affiliation(s)
- Eun-Hye Hong
- Laboratory of Microbiology and Immunology, College of Pharmacy, Kangwon National University, Chuncheon, 24341, South Korea
| | - Jaewon Cho
- Laboratory of Microbiology and Immunology, College of Pharmacy, Kangwon National University, Chuncheon, 24341, South Korea
| | - Jae-Hee Ahn
- Laboratory of Microbiology and Immunology, College of Pharmacy, Kangwon National University, Chuncheon, 24341, South Korea
| | - Bo-Eun Kwon
- Laboratory of Microbiology and Immunology, College of Pharmacy, Kangwon National University, Chuncheon, 24341, South Korea
| | - Mi-Na Kweon
- Mucosal Immunology Laboratory, Department of Convergence Medicine, University of Ulsan College of Medicine/Asan Medical Center, Seoul, 05505, South Korea
| | - Sang-Uk Seo
- Department of Microbiology, College of Medicine, The Catholic University of Korea, Seoul, 05505, South Korea
| | - Byung-Il Yoon
- Laboratory of Histology and Molecular Pathogenesis, College of Veterinary Medicine, Kangwon National University, Chuncheon, 24341, South Korea
| | - Sun-Young Chang
- Laboratory of Microbiology, College of Pharmacy, Ajou University, Suwon, 16499, South Korea
| | - Hyun-Jeong Ko
- Laboratory of Microbiology and Immunology, College of Pharmacy, Kangwon National University, Chuncheon, 24341, South Korea.
| |
Collapse
|
38
|
Sieminska I, Baran J. Myeloid-Derived Suppressor Cells in Colorectal Cancer. Front Immunol 2020; 11:1526. [PMID: 32849517 PMCID: PMC7426395 DOI: 10.3389/fimmu.2020.01526] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Accepted: 06/10/2020] [Indexed: 12/24/2022] Open
Abstract
Colorectal cancer (CRC) remains one of the most common malignancies diagnosed worldwide. The pathogenesis of CRC is complex and involves, among others, accumulation of genetic predispositions and epigenetic factors, dietary habits, alterations in gut microbiota, and lack of physical activity. A growing body of evidence suggests that immune cells play different roles in CRC, comprising both pro- and anti-tumorigenic functions. Immunosuppression observed during cancer development and progression is a result of the orchestration of many cell types, including myeloid-derived suppressor cells (MDSCs). MDSCs, along with other cells, stimulate tumor growth, angiogenesis, and formation of metastases. This article focuses on MDSCs in relation to their role in the initiation and progression of CRC. Possible forms of immunotherapies targeting MDSCs in CRC are also discussed.
Collapse
Affiliation(s)
| | - Jarek Baran
- Department of Clinical Immunology, Jagiellonian University Medical College, Krakow, Poland
| |
Collapse
|
39
|
CD38: T Cell Immuno-Metabolic Modulator. Cells 2020; 9:cells9071716. [PMID: 32709019 PMCID: PMC7408359 DOI: 10.3390/cells9071716] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 07/14/2020] [Accepted: 07/15/2020] [Indexed: 12/22/2022] Open
Abstract
Activation and subsequent differentiation of T cells following antigenic stimulation are triggered by highly coordinated signaling events that lead to instilling cells with a discrete metabolic and transcriptional feature. Compelling studies indicate that intracellular nicotinamide adenine dinucleotide (NAD+) levels have profound influence on diverse signaling and metabolic pathways of T cells, and hence dictate their functional fate. CD38, a major mammalian NAD+ glycohydrolase (NADase), expresses on T cells following activation and appears to be an essential modulator of intracellular NAD+ levels. The enzymatic activity of CD38 in the process of generating the second messenger cADPR utilizes intracellular NAD+, and thus limits its availability to different NAD+ consuming enzymes (PARP, ART, and sirtuins) inside the cells. The present review discusses how the CD38-NAD+ axis affects T cell activation and differentiation through interfering with their signaling and metabolic processes. We also describe the pivotal role of the CD38-NAD+ axis in influencing the chromatin remodeling and rewiring T cell response. Overall, this review emphasizes the crucial contribution of the CD38-NAD+ axis in altering T cell response in various pathophysiological conditions.
Collapse
|
40
|
Han P, Cao P, Hu S, Kong K, Deng Y, Zhao B, Li F. Esophageal Microenvironment: From Precursor Microenvironment to Premetastatic Niche. Cancer Manag Res 2020; 12:5857-5879. [PMID: 32765088 PMCID: PMC7371556 DOI: 10.2147/cmar.s258215] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Accepted: 06/29/2020] [Indexed: 12/15/2022] Open
Abstract
Esophageal cancer (EC) is the sixth most deadly cancer, and its incidence is still increasing year by year. Although the researches on the molecular mechanisms of EC have been widely carried out and incremental progress has been made, its overall survival rate is still low. There is cumulative evidence showing that the esophageal microenvironment plays a vital role in the development of EC. In precancerous lesions of the esophagus, high-risk environmental factors can promote the development of precancerous lesions by inducing the production of inflammatory factors and the recruitment of immune cells. In the tumor microenvironment, tumor-promoting cells can inhibit anti-tumor immunity and promote tumor progression through a variety of pathways, such as bone marrow-derived suppressor cells (MDSCs), tumor-associated fibroblasts (CAFs), and regulatory T cells (Tregs). The formation of extracellular hypoxia and acidic microenvironment and the change of extracellular matrix stiffness are also important factors affecting tumor progression and metastasis. Simultaneously, primary tumor-derived cytokines and bone marrow-derived immune cells can also promote the formation of pre-metastasis niche of EC lymph nodes, which are beneficial to EC lymph node metastasis. Further research on the specific mechanism of these processes in the occurrence, development, and metastasis of each EC subtype will support us to grasp the overall pre-cancerous prevention, targeted treatment, and metastatic assessment of EC.
Collapse
Affiliation(s)
- Peng Han
- Department of Thoracic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, People's Republic of China
| | - Peng Cao
- Department of Thoracic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, People's Republic of China
| | - Shan Hu
- Department of Thoracic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, People's Republic of China
| | - Kangle Kong
- Department of Thoracic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, People's Republic of China
| | - Yu Deng
- Department of Thoracic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, People's Republic of China
| | - Bo Zhao
- Department of Thoracic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, People's Republic of China
| | - Fan Li
- Department of Thoracic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, People's Republic of China
| |
Collapse
|
41
|
Kuwahara-Ota S, Shimura Y, Steinebach C, Isa R, Yamaguchi J, Nishiyama D, Fujibayashi Y, Takimoto-Shimomura T, Mizuno Y, Matsumura-Kimoto Y, Tsukamoto T, Chinen Y, Kobayashi T, Horiike S, Taniwaki M, Gütschow M, Kuroda J. Lenalidomide and pomalidomide potently interfere with induction of myeloid-derived suppressor cells in multiple myeloma. Br J Haematol 2020; 191:784-795. [PMID: 32558939 DOI: 10.1111/bjh.16881] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2020] [Accepted: 05/22/2020] [Indexed: 12/15/2022]
Abstract
An increase in immunosuppressive myeloid-derived suppressor cells (MDSCs) is associated with disease progression and treatment resistance in multiple myeloma (MM). We investigated the mechanisms underlying MDSC induction, and sought to discover a strategy for prevention of MDSC induction in MM. Using a transwell co-culture system, four of nine examined human myeloma-derived cell lines (HMCLs) were potent in inducing monocytic (M)-MDSCs from normal peripheral blood mononuclear cells (PBMCs). As the results, we identified that secretion of C-C motif chemokine ligand 5 (CCL5) and macrophage migration inhibitory factor (MIF) by myeloma cells is a prerequisite for induction of MDSCs in MM. The immunomodulatory drug (IMiD) compounds, such as lenalidomide (LEN) and pomalidomide (POM), were identified as potent inhibitors of MDSC induction through bidirectional molecular effects of cereblon (CRBN)-dependent and -independent downregulation of CCL5 and MIF in myeloma cells; and downregulation of C-C motif chemokine receptor 5, a receptor for CCL5, and induction of interferon regulatory factor 8, a critical transcription factor for monocytic differentiation, in PBMCs. In the present study of the molecular mechanisms underlying MDSC induction, we identified a novel effect of LEN and POM of inhibiting MDSC induction via overlapping regulatory effects in myeloma cells and normal PBMCs.
Collapse
Affiliation(s)
- Saeko Kuwahara-Ota
- Division of Hematology and Oncology, Department of Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Yuji Shimura
- Division of Hematology and Oncology, Department of Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Christian Steinebach
- Department of Pharmaceutical & Medicinal Chemistry, University of Bonn, Pharmaceutical Institute, Bonn, Germany
| | - Reiko Isa
- Division of Hematology and Oncology, Department of Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Junko Yamaguchi
- Division of Hematology and Oncology, Department of Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Daichi Nishiyama
- Division of Hematology and Oncology, Department of Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Yuto Fujibayashi
- Division of Hematology and Oncology, Department of Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Tomoko Takimoto-Shimomura
- Division of Hematology and Oncology, Department of Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Yoshimi Mizuno
- Division of Hematology and Oncology, Department of Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Yayoi Matsumura-Kimoto
- Division of Hematology and Oncology, Department of Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Taku Tsukamoto
- Division of Hematology and Oncology, Department of Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Yoshiaki Chinen
- Division of Hematology and Oncology, Department of Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan.,Department of Hematology, Fukuchiyama City Hospital, Kyoto, Japan
| | - Tsutomu Kobayashi
- Division of Hematology and Oncology, Department of Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Shigeo Horiike
- Division of Hematology and Oncology, Department of Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Masafumi Taniwaki
- Center for Molecular Diagnostics and Therapeutics, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Michael Gütschow
- Department of Pharmaceutical & Medicinal Chemistry, University of Bonn, Pharmaceutical Institute, Bonn, Germany
| | - Junya Kuroda
- Division of Hematology and Oncology, Department of Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan
| |
Collapse
|
42
|
Fucà G, Guarini V, Antoniotti C, Morano F, Moretto R, Corallo S, Marmorino F, Lonardi S, Rimassa L, Sartore-Bianchi A, Borelli B, Tampellini M, Bustreo S, Claravezza M, Boccaccino A, Murialdo R, Zaniboni A, Tomasello G, Loupakis F, Adamo V, Tonini G, Cortesi E, de Braud F, Cremolini C, Pietrantonio F. The Pan-Immune-Inflammation Value is a new prognostic biomarker in metastatic colorectal cancer: results from a pooled-analysis of the Valentino and TRIBE first-line trials. Br J Cancer 2020; 123:403-409. [PMID: 32424148 PMCID: PMC7403416 DOI: 10.1038/s41416-020-0894-7] [Citation(s) in RCA: 129] [Impact Index Per Article: 32.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2020] [Revised: 04/03/2020] [Accepted: 04/22/2020] [Indexed: 11/25/2022] Open
Abstract
Background Immune-inflammatory biomarkers (IIBs) showed a prognostic relevance in patients with metastatic CRC (mCRC). We aimed at evaluating the prognostic power of a new comprehensive biomarker, the Pan-Immune-Inflammation Value (PIV), in patients with mCRC receiving first-line therapy. Methods In the present pooled-analysis, we included patients enrolled in the Valentino and TRIBE trials. PIV was calculated as: (neutrophil count × platelet count × monocyte count)/lymphocyte count. A cut-off was determined using the maximally selected rank statistics method. Generalised boosted regression (GBR), the Kaplan–Meier method and Cox hazards regression models were used for survival analyses. Results A total of 438 patients were included. Overall, 208 patients (47%) had a low-baseline PIV and 230 (53%) had a high-baseline PIV. Patients with high PIV experienced a worse PFS (HR, 1.66; 95% CI, 1.36–2.03, P < 0.001) and worse OS (HR, 2.01; 95% CI, 1.57–2.57; P < 0.001) compared to patients with low PIV. PIV outperformed the other IIBs in the GBR model and in the multivariable models. Conclusion PIV is a strong predictor of survival outcomes with better performance than other well-known IIBs in patients with mCRC treated with first-line therapy. PIV should be prospectively validated to better stratify mCRC patients undergoing first-line therapy.
Collapse
Affiliation(s)
- Giovanni Fucà
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milan, Italy
| | - Vincenzo Guarini
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milan, Italy
| | - Carlotta Antoniotti
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy.,Unit of Medical Oncology 2, Azienda Ospedaliero-Universitaria Pisana, Pisa, Italy
| | - Federica Morano
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milan, Italy
| | - Roberto Moretto
- Unit of Medical Oncology 2, Azienda Ospedaliero-Universitaria Pisana, Pisa, Italy
| | - Salvatore Corallo
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milan, Italy
| | - Federica Marmorino
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy.,Unit of Medical Oncology 2, Azienda Ospedaliero-Universitaria Pisana, Pisa, Italy
| | - Sara Lonardi
- Unit of Medical Oncology 1, Department of Clinical and Experimental Oncology, Istituto Oncologico Veneto, IRCCS, Padua, Italy
| | - Lorenza Rimassa
- Medical Oncology and Hematology Unit, Humanitas Cancer Center, Humanitas Clinical and Research Center-IRCCS, Rozzano, Italy.,Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Italy
| | - Andrea Sartore-Bianchi
- Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, Milan, Italy.,Oncology and Hemato-oncology Department, University of Milan, Milan, Italy
| | - Beatrice Borelli
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy.,Unit of Medical Oncology 2, Azienda Ospedaliero-Universitaria Pisana, Pisa, Italy
| | - Marco Tampellini
- Department of Oncology, AOU San Luigi di Orbassano, University of Torino, Orbassano, Italy
| | - Sara Bustreo
- Colorectal Cancer Unit, Medical Oncology Division 1, AOU Città della Salute e della Scienza, Torino, Italy
| | - Matteo Claravezza
- Medical Oncology Unit, Ente Ospedaliero Ospedali Galliera, Genoa, Italy
| | - Alessandra Boccaccino
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy.,Unit of Medical Oncology 2, Azienda Ospedaliero-Universitaria Pisana, Pisa, Italy
| | - Roberto Murialdo
- Department of Internal Medicine, University of Genoa and IRCCS AOU San Martino-IST, Genoa, Italy
| | | | - Gianluca Tomasello
- Medical Oncology Unit, Azienda Socio-Sanitaria Territoriale (ASST) Ospedale di Cremona, Cremona, Italy
| | - Fotios Loupakis
- Unit of Medical Oncology 1, Department of Clinical and Experimental Oncology, Istituto Oncologico Veneto, IRCCS, Padua, Italy
| | - Vincenzo Adamo
- Medical Oncology Unit, A.O. Papardo, Messina, Italy.,Department of Human Pathology, University of Messina, Messina, Italy
| | - Giuseppe Tonini
- Oncology Department, Policlinico Campus Bio-Medico di Roma, Rome, Italy
| | | | - Filippo de Braud
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milan, Italy.,Oncology and Hemato-oncology Department, University of Milan, Milan, Italy
| | - Chiara Cremolini
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy.,Unit of Medical Oncology 2, Azienda Ospedaliero-Universitaria Pisana, Pisa, Italy
| | - Filippo Pietrantonio
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milan, Italy. .,Oncology and Hemato-oncology Department, University of Milan, Milan, Italy.
| |
Collapse
|
43
|
Jayakumar A, Bothwell ALM. Functional Diversity of Myeloid-Derived Suppressor Cells: The Multitasking Hydra of Cancer. THE JOURNAL OF IMMUNOLOGY 2020; 203:1095-1103. [PMID: 31427398 DOI: 10.4049/jimmunol.1900500] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Accepted: 06/03/2019] [Indexed: 12/21/2022]
Abstract
Myeloid-derived suppressor cells (MDSCs) are immature suppressive cells found in tumors and immunological niches. In this article, we highlight the ability of MDSCs to promote IL-17-producing T cells (Th17) and regulatory T cells in addition to suppressing cytotoxic T cells in different tumor models. These interactions between MDSCs and T cells support tumor growth because IL-17 is tumorigenic in many cancer types and regulatory T cells suppress antitumor T cells. Besides T cells, MDSCs promote regulatory B cells and suppress overall B cell function; however, tumor-evoked regulatory B cells also regulate MDSC function, suggesting cross-regulation between MDSCs and B cells. These multiple functions shed light on how MDSCs dysregulate several arms of host immune response. Moreover, MDSCs promote tumor cell survival and angiogenesis to support tumors. Therefore, the multifunctional feature of MDSCs make them attractive immunotherapeutic targets.
Collapse
Affiliation(s)
- Asha Jayakumar
- Department of Immunobiology, Yale University, New Haven, CT 06520
| | | |
Collapse
|
44
|
Calabretta E, Carlo-Stella C. The Many Facets of CD38 in Lymphoma: From Tumor-Microenvironment Cell Interactions to Acquired Resistance to Immunotherapy. Cells 2020; 9:E802. [PMID: 32225002 PMCID: PMC7226059 DOI: 10.3390/cells9040802] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 03/19/2020] [Accepted: 03/25/2020] [Indexed: 12/13/2022] Open
Abstract
The CD38 antigen is expressed in several hematological malignancies, and the anti-CD38 monoclonal antibodies Daratumumab and Isatuximab have an established role in the therapy of multiple myeloma. However, data on the therapeutic utility of CD38 targeting in other lymphoid malignancies are limited. In chronic lymphocytic leukemia, the prognostic significance of CD38 expression is well accepted, and preclinical studies on the use of Daratumumab in monotherapy or combination therapy have demonstrated considerable efficacy. In other lymphoproliferative disorders, preclinical and clinical data have not been as compelling; however, CD38 overexpression likely contributes to resistance to checkpoint inhibitors, prompting numerous clinical trials in Hodgkin and non-Hodgkin lymphoma to investigate whether blocking CD38 enhances the efficacy of checkpoint inhibitors. Furthermore, due to its widespread expression in hematological tumors, CD38 represents an attractive target for cellular therapies such as CAR-T cells. The present review discusses current knowledge of CD38 expression and its implications in various lymphoid malignancies. Furthermore, it addresses current and future therapeutic perspectives, with a particular emphasis on the significance of CD38 interaction with immune cells of the tumor microenvironment. Lastly, results of ongoing studies using anti-CD38 antibodies will be reviewed.
Collapse
Affiliation(s)
- Eleonora Calabretta
- Department of Oncology and Hematology, Humanitas Cancer Center, Humanitas Clinical and Research Center, Rozzano, 20089 Milano, Italy;
| | - Carmelo Carlo-Stella
- Department of Oncology and Hematology, Humanitas Cancer Center, Humanitas Clinical and Research Center, Rozzano, 20089 Milano, Italy;
- Department of Biomedical Sciences, Humanitas University, Rozzano, 20089 Milano, Italy
| |
Collapse
|
45
|
Law AMK, Valdes-Mora F, Gallego-Ortega D. Myeloid-Derived Suppressor Cells as a Therapeutic Target for Cancer. Cells 2020; 9:cells9030561. [PMID: 32121014 PMCID: PMC7140518 DOI: 10.3390/cells9030561] [Citation(s) in RCA: 275] [Impact Index Per Article: 68.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Revised: 02/22/2020] [Accepted: 02/24/2020] [Indexed: 12/15/2022] Open
Abstract
The emergence of immunotherapy has been an astounding breakthrough in cancer treatments. In particular, immune checkpoint inhibitors, targeting PD-1 and CTLA-4, have shown remarkable therapeutic outcomes. However, response rates from immunotherapy have been reported to be varied, with some having pronounced success and others with minimal to no clinical benefit. An important aspect associated with this discrepancy in patient response is the immune-suppressive effects elicited by the tumour microenvironment (TME). Immune suppression plays a pivotal role in regulating cancer progression, metastasis, and reducing immunotherapy success. Most notably, myeloid-derived suppressor cells (MDSC), a heterogeneous population of immature myeloid cells, have potent mechanisms to inhibit T-cell and NK-cell activity to promote tumour growth, development of the pre-metastatic niche, and contribute to resistance to immunotherapy. Accumulating research indicates that MDSC can be a therapeutic target to alleviate their pro-tumourigenic functions and immunosuppressive activities to bolster the efficacy of checkpoint inhibitors. In this review, we provide an overview of the general immunotherapeutic approaches and discuss the characterisation, expansion, and activities of MDSCs with the current treatments used to target them either as a single therapeutic target or synergistically in combination with immunotherapy.
Collapse
Affiliation(s)
- Andrew M. K. Law
- Tumour Development Group, The Kinghorn Cancer Centre, Garvan Institute of Medical Research, Sydney, NSW 2010, Australia
- Correspondence: (A.M.K.L.); (F.V.-M.); (D.G.-O.); Tel.: +61-(0)2-9355-5894 (A.M.K.L); +61-(0)2-9385-0143 (F.V.-M); +61-(0)2-9355-5776 (D.G.-O)
| | - Fatima Valdes-Mora
- Histone Variants Group, Garvan Institute of Medical Research, Sydney, NSW 2010, Australia
- St. Vincent’s Clinical School, Faculty of Medicine, University of New South Wales Sydney, Sydney, NSW 2052, Australia
- Correspondence: (A.M.K.L.); (F.V.-M.); (D.G.-O.); Tel.: +61-(0)2-9355-5894 (A.M.K.L); +61-(0)2-9385-0143 (F.V.-M); +61-(0)2-9355-5776 (D.G.-O)
| | - David Gallego-Ortega
- Tumour Development Group, The Kinghorn Cancer Centre, Garvan Institute of Medical Research, Sydney, NSW 2010, Australia
- St. Vincent’s Clinical School, Faculty of Medicine, University of New South Wales Sydney, Sydney, NSW 2052, Australia
- Correspondence: (A.M.K.L.); (F.V.-M.); (D.G.-O.); Tel.: +61-(0)2-9355-5894 (A.M.K.L); +61-(0)2-9385-0143 (F.V.-M); +61-(0)2-9355-5776 (D.G.-O)
| |
Collapse
|
46
|
Pang X, Fan HY, Tang YL, Wang SS, Cao MX, Wang HF, Dai LL, Wang K, Yu XH, Wu JB, Tang YJ, Liang XH. Myeloid derived suppressor cells contribute to the malignant progression of oral squamous cell carcinoma. PLoS One 2020; 15:e0229089. [PMID: 32092078 PMCID: PMC7039453 DOI: 10.1371/journal.pone.0229089] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Accepted: 01/29/2020] [Indexed: 02/05/2023] Open
Abstract
Purpose The tumor-related myeloid derived suppressor cells (MDSCs), important immunosuppressive cells in tumor microenvironment, play an important role in the cancer progression. This study is aimed to investigate the crosstalk between MDSCs and oral squamous cell carcinoma (OSCC) cells and their role in the malignant progression of OSCC. Methods Immunochemistry (IHC) was used to investigate the expression of CD33 in 200 OSCC, 36 premalignant. CD33+ MDSCs were sorted and enriched via magnetic-activated cell sorting (MACS) from OSCC patients or health donor, and their phenotypes were identified by flow cytometry. With a co-culture system of MDSCs and OSCC, the effects of MDSCs on OSCC proliferation, apoptosis, migration invasion, epithelial-mesenchymal transition (EMT), and vasculogenic mimicry formation (VM) formation were assessed, respectively. Besides, peripheral blood mononuclear cells (PBMCs) from health donor were cultured with OSCC supernatant, the level of MDSCs and expressions of Arginase (Arg-1) and inducible nitric oxide synthase (iNOS) were measured. Results The number of MDSCs was increased in tumor tissues of OSCC patients, and was positively related to the T stage, pathological grade, lymph node metastasis and poor prognosis. Tumor-related MDSCs of the co-culture system promoted OSCC progression by contributing to cell proliferation, migration and invasion as well as inducing EMT and VM. In turn, OSCC cells had potential to induce MDSCs differentiation from PBMCs and increase the expression of Arg-1 and iNOS. Conclusion These indicated that the crosstalk between MDSCs and tumor cells facilitated the malignant progression of OSCC cells and the immune suppressive properties of MDSCs, which may provide new insights into tumor treatment on targeting tumor-associated immunosuppressive cells.
Collapse
Affiliation(s)
- Xin Pang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Hua-yang Fan
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Ya-ling Tang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Sha-sha Wang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Ming-xin Cao
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Hao-fan Wang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Lu-ling Dai
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Ke Wang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Xiang-hua Yu
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Jing-biao Wu
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Ya-Jie Tang
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao, China
- * E-mail: (YJT); (XHL)
| | - Xin-hua Liang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
- * E-mail: (YJT); (XHL)
| |
Collapse
|
47
|
Trovato R, Canè S, Petrova V, Sartoris S, Ugel S, De Sanctis F. The Engagement Between MDSCs and Metastases: Partners in Crime. Front Oncol 2020; 10:165. [PMID: 32133298 PMCID: PMC7040035 DOI: 10.3389/fonc.2020.00165] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2019] [Accepted: 01/30/2020] [Indexed: 12/18/2022] Open
Abstract
Tumor metastases represent the major cause of cancer-related mortality, confirming the urgent need to identify key molecular pathways and cell-associated networks during the early phases of the metastatic process to develop new strategies to either prevent or control distal cancer spread. Several data revealed the ability of cancer cells to establish a favorable microenvironment, before their arrival in distant organs, by manipulating the cell composition and function of the new host tissue where cancer cells can survive and outgrow. This predetermined environment is termed “pre-metastatic niche” (pMN). pMN development requires that tumor-derived soluble factors, like cytokines, growth-factors and extracellular vesicles, genetically and epigenetically re-program not only resident cells (i.e., fibroblasts) but also non-resident cells such as bone marrow-derived cells. Indeed, by promoting an “emergency” myelopoiesis, cancer cells switch the steady state production of blood cells toward the generation of pro-tumor circulating myeloid cells defined as myeloid-derived suppressor cells (MDSCs) able to sustain tumor growth and dissemination. MDSCs are a heterogeneous subset of myeloid cells with immunosuppressive properties that sustain metastatic process. In this review, we discuss current understandings of how MDSCs shape and promote metastatic dissemination acting in each fundamental steps of cancer progression from primary tumor to metastatic disease.
Collapse
Affiliation(s)
- Rosalinda Trovato
- Section of Immunology, Department of Medicine, University of Verona, Verona, Italy
| | - Stefania Canè
- Section of Immunology, Department of Medicine, University of Verona, Verona, Italy
| | - Varvara Petrova
- Section of Immunology, Department of Medicine, University of Verona, Verona, Italy
| | - Silvia Sartoris
- Section of Immunology, Department of Medicine, University of Verona, Verona, Italy
| | - Stefano Ugel
- Section of Immunology, Department of Medicine, University of Verona, Verona, Italy
| | - Francesco De Sanctis
- Section of Immunology, Department of Medicine, University of Verona, Verona, Italy
| |
Collapse
|
48
|
Bergenfelz C, Leandersson K. The Generation and Identity of Human Myeloid-Derived Suppressor Cells. Front Oncol 2020; 10:109. [PMID: 32117758 PMCID: PMC7025543 DOI: 10.3389/fonc.2020.00109] [Citation(s) in RCA: 72] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Accepted: 01/21/2020] [Indexed: 12/29/2022] Open
Abstract
Myeloid-derived suppressor cells (MDSCs) are cells of myeloid lineage with a potent immunosuppressive capacity. They are present in cancer patients as well as in patients with severe inflammatory conditions and infections. MDSCs exist as two main subtypes, the granulocytic (G-MDSCs) and the monocytic (Mo-MDSCs) type, as defined by their surface phenotype and functions. While the functions of MDSCs have been investigated in depth, the origin of human MDSCs is less characterized and even controversial. In this review, we recapitulate theories on how MDSCs are generated in mice, and whether this knowledge is translatable into human MDSC biology, as well as on problems of defining MDSCs by their immature cell surface phenotype in relation to the plasticity of myeloid cells. Finally, the challenge of pharmacological targeting of MDSCs in the future is envisioned.
Collapse
Affiliation(s)
- Caroline Bergenfelz
- Department of Translational Medicine, Division of Experimental Infection Medicine, Lund University, Malmö, Sweden
| | - Karin Leandersson
- Department of Translational Medicine, Cancer Immunology, Lund University, Skåne University Hospital, Malmö, Sweden
| |
Collapse
|
49
|
Chen SMY, Krinsky AL, Woolaver RA, Wang X, Chen Z, Wang JH. Tumor immune microenvironment in head and neck cancers. Mol Carcinog 2020; 59:766-774. [PMID: 32017286 DOI: 10.1002/mc.23162] [Citation(s) in RCA: 96] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Revised: 01/18/2020] [Accepted: 01/22/2020] [Indexed: 12/24/2022]
Abstract
Head and neck cancers are a heterogeneous group of tumors that are highly aggressive and collectively represent the sixth most common cancer worldwide. Ninety percent of head and neck cancers are squamous cell carcinomas (HNSCCs). The tumor microenvironment (TME) of HNSCCs consists of many different subsets of cells that infiltrate the tumors and interact with the tumor cells or with each other through various networks. Both innate and adaptive immune cells play a crucial role in mediating immune surveillance and controlling tumor growth. Here, we discuss the different subsets of immune cells and how they contribute to an immunosuppressive TME of HNSCCs. We also briefly summarize recent advances in immunotherapeutic approaches for HNSCC treatment. A better understanding of the multiple factors that play pivotal roles in HNSCC tumorigenesis and tumor progression may help define novel targets to develop more effective immunotherapies for patients with HNSCC.
Collapse
Affiliation(s)
- Samantha M Y Chen
- Department of Immunology and Microbiology, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Alexandra L Krinsky
- Department of Immunology and Microbiology, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Rachel A Woolaver
- Department of Immunology and Microbiology, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Xiaoguang Wang
- Department of Immunology and Microbiology, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Zhangguo Chen
- Department of Immunology and Microbiology, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Jing H Wang
- Department of Immunology and Microbiology, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| |
Collapse
|
50
|
The Good, the Bad and the Unknown of CD38 in the Metabolic Microenvironment and Immune Cell Functionality of Solid Tumors. Cells 2019; 9:cells9010052. [PMID: 31878283 PMCID: PMC7016859 DOI: 10.3390/cells9010052] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Revised: 12/06/2019] [Accepted: 12/18/2019] [Indexed: 02/08/2023] Open
Abstract
The regulation of the immune microenvironment within solid tumors has received increasing attention with the development and clinical success of immune checkpoint blockade therapies, such as those that target the PD-1/PD-L1 axis. The metabolic microenvironment within solid tumors has proven to be an important regulator of both the natural suppression of immune cell functionality and the de novo or acquired resistance to immunotherapy. Enzymatic proteins that generate immunosuppressive metabolites like adenosine are thus attractive targets to couple with immunotherapies to improve clinical efficacy. CD38 is one such enzyme. While the role of CD38 in hematological malignancies has been extensively studied, the impact of CD38 expression within solid tumors is largely unknown, though most current data indicate an immunosuppressive role for CD38. However, CD38 is far from a simple enzyme, and there are several remaining questions that require further study. To effectively treat solid tumors, we must learn as much about this multifaceted protein as possible—i.e., which infiltrating immune cell types express CD38 for functional activities, the most effective CD38 inhibitor(s) to employ, and the influence of other similarly functioning enzymes that may also contribute towards an immunosuppressive microenvironment. Gathering knowledge such as this will allow for intelligent targeting of CD38, the reinvigoration of immune functionality and, ultimately, tumor elimination.
Collapse
|