1
|
Huang S, Chung JYF, Li C, Wu Y, Qiao G, To KF, Tang PMK. Cellular dynamics of tumor microenvironment driving immunotherapy resistance in non-small-cell lung carcinoma. Cancer Lett 2024; 604:217272. [PMID: 39326553 DOI: 10.1016/j.canlet.2024.217272] [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: 07/06/2024] [Revised: 09/04/2024] [Accepted: 09/16/2024] [Indexed: 09/28/2024]
Abstract
Immune checkpoint inhibitors (ICIs) have profoundly reshaped the treatment paradigm for non-small cell lung cancer (NSCLC). Despite these advancements, primary and secondary resistance to ICIs remain prevalent challenges in managing advanced NSCLC. Recent studies have highlighted the significant role of the tumor microenvironment (TME) in modulating treatment responses. This review aims to comprehensively examine the interactive roles of immune/stromal cells-such as T cells, B cells, neutrophils, macrophages, and CAFs within the TME, elucidating how these diverse cellular interactions contribute to immunotherapy resistance. It focuses on the dynamic interactions among diverse cell types such as the varying states of T cells under the influence of TME constituents like immune cells and cancer-associated fibroblasts (CAFs). By exploring the mechanisms involved in the complex cellular interactions, we highlight novel therapeutic targets and strategies aimed at overcoming resistance, thereby enhancing the efficacy of ICIs in NSCLC. Our synthesis of recent research provides critical insights into the multifaceted mechanisms of resistance and paves the way for the development of more effective, personalized treatment approaches.
Collapse
Affiliation(s)
- Shujie Huang
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Translational Oncology, Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Jeff Yat-Fai Chung
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Translational Oncology, Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Chunjie Li
- Department of Head and Neck Oncology, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Yi Wu
- MOE Key Laboratory of Environment and Genes Related to Diseases, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, 710061, China
| | - Guibin Qiao
- Department of Thoracic Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Ka-Fai To
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Translational Oncology, Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Patrick Ming-Kuen Tang
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Translational Oncology, Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, Hong Kong; Peter Hung Pain Research Institute, The Chinese University of Hong Kong, Hong Kong.
| |
Collapse
|
2
|
Patin EC, Nenclares P, Chan Wah Hak C, Dillon MT, Patrikeev A, McLaughlin M, Grove L, Foo S, Soliman H, Barata JP, Marsden J, Baldock H, Gkantalis J, Roulstone V, Kyula J, Burley A, Hubbard L, Pedersen M, Smith SA, Clancy-Thompson E, Melcher AA, Ono M, Rullan A, Harrington KJ. Sculpting the tumour microenvironment by combining radiotherapy and ATR inhibition for curative-intent adjuvant immunotherapy. Nat Commun 2024; 15:6923. [PMID: 39134540 PMCID: PMC11319479 DOI: 10.1038/s41467-024-51236-6] [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: 01/05/2024] [Accepted: 07/19/2024] [Indexed: 08/15/2024] Open
Abstract
The combination of radiotherapy/chemoradiotherapy and immune checkpoint blockade can result in poor outcomes in patients with locally advanced head and neck squamous cell carcinoma (HNSCC). Here, we show that combining ATR inhibition (ATRi) with radiotherapy (RT) increases the frequency of activated NKG2A+PD-1+ T cells in animal models of HNSCC. Compared with the ATRi/RT treatment regimen alone, the addition of simultaneous NKG2A and PD-L1 blockade to ATRi/RT, in the adjuvant, post-radiotherapy setting induces a robust antitumour response driven by higher infiltration and activation of cytotoxic T cells in the tumour microenvironment. The efficacy of this combination relies on CD40/CD40L costimulation and infiltration of activated, proliferating memory CD8+ and CD4+ T cells with persistent or new T cell receptor (TCR) signalling, respectively. We also observe increased richness in the TCR repertoire and emergence of numerous and large TCR clonotypes that cluster based on antigen specificity in response to NKG2A/PD-L1/ATRi/RT. Collectively, our data point towards potential combination approaches for the treatment of HNSCC.
Collapse
Affiliation(s)
- Emmanuel C Patin
- Targeted Therapy Team, The Institute of Cancer Research, London, UK.
| | - Pablo Nenclares
- Targeted Therapy Team, The Institute of Cancer Research, London, UK
- The Royal Marsden Hospital, London, UK
| | - Charleen Chan Wah Hak
- Targeted Therapy Team, The Institute of Cancer Research, London, UK
- The Royal Marsden Hospital, London, UK
| | - Magnus T Dillon
- Targeted Therapy Team, The Institute of Cancer Research, London, UK
- The Royal Marsden Hospital, London, UK
| | - Anton Patrikeev
- Targeted Therapy Team, The Institute of Cancer Research, London, UK
| | | | - Lorna Grove
- Targeted Therapy Team, The Institute of Cancer Research, London, UK
- The Royal Marsden Hospital, London, UK
| | - Shane Foo
- Targeted Therapy Team, The Institute of Cancer Research, London, UK
| | | | | | | | - Holly Baldock
- Targeted Therapy Team, The Institute of Cancer Research, London, UK
| | - Jim Gkantalis
- Targeted Therapy Team, The Institute of Cancer Research, London, UK
| | | | - Joan Kyula
- Targeted Therapy Team, The Institute of Cancer Research, London, UK
| | - Amy Burley
- Targeted Therapy Team, The Institute of Cancer Research, London, UK
| | - Lisa Hubbard
- Targeted Therapy Team, The Institute of Cancer Research, London, UK
| | - Malin Pedersen
- Targeted Therapy Team, The Institute of Cancer Research, London, UK
| | | | | | - Alan A Melcher
- Translational Immunotherapy Team, The Institute of Cancer Research, London, UK
| | - Masahiro Ono
- Department of Life Sciences, Imperial College London, London, UK
| | - Antonio Rullan
- Targeted Therapy Team, The Institute of Cancer Research, London, UK
- The Royal Marsden Hospital, London, UK
| | - Kevin J Harrington
- Targeted Therapy Team, The Institute of Cancer Research, London, UK
- The Royal Marsden Hospital, London, UK
| |
Collapse
|
3
|
Plewa N, Poncette L, Blankenstein T. Generation of TGFβR2(-1) neoantigen-specific HLA-DR4-restricted T cell receptors for cancer therapy. J Immunother Cancer 2023; 11:jitc-2022-006001. [PMID: 36822673 PMCID: PMC9950979 DOI: 10.1136/jitc-2022-006001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/06/2023] [Indexed: 02/25/2023] Open
Abstract
BACKGROUND Adoptive transfer of patient's T cells, engineered to express a T cell receptor (TCR) with defined novel antigen specificity, is a convenient form of cancer therapy. In most cases, major histocompatibility complex (MHC) I-restricted TCRs are expressed in CD8+ T cells and the development of CD4+ T cells engineered to express an MHC II-restricted TCR lacks behind. Critical is the choice of the target antigen, whether the epitope is efficiently processed and binds with high affinity to MHC molecules. A mutation in the transforming growth factor β receptor 2 (TGFβR2(-1)) gene creates a frameshift peptide caused by the deletion of one adenine (-1) within a microsatellite sequence. This somatic mutation is recurrent in microsatellite instable colorectal and gastric cancers and, therefore, is a truly tumor-specific antigen detected in many patients. METHODS ABabDR4 mice, which express a diverse human TCR repertoire restricted to human MHC II molecule HLA-DRA/DRB1*0401 (HLA-DR4), were immunized with the TGFβR2(-1) peptide and TGFβR2(-1)-specific TCRs were isolated from responding CD4+ T cells. The TGFβR2(-1)-specific TCRs were expressed in human CD4+ T cells and their potency and safety profile were assessed by co-cultures and other functional assays. RESULTS We demonstrated that TGFβR2(-1) neoantigen is immunogenic and elicited CD4+ T cell responses in ABabDR4 mice. When expressed in human CD4+ T cells, the HLA-DR4 restricted TGFβR2(-1)-specific TCRs induced IFNy expression at low TGFβR2(-1) peptide amounts. The TGFβR2(-1)-specific TCRs recognized HLA-DR4+ lymphoblastoid cells, which endogenously processed and presented the neoantigen, and colorectal cancer cell lines SW48 and HCT116 naturally expressing the TGFβR2(-1) mutation. No MHC II alloreactivity or cross-reactivity to peptides with a similar TCR-recognition motif were observed, indicating the safety of the TCRs. CONCLUSIONS The data suggest that HLA-DR4-restricted TCRs specific for the TGFβR2(-1) recurrent neoantigen can be valuable candidates for adoptive T cell therapy of a sizeable number of patients with cancer.
Collapse
Affiliation(s)
- Natalia Plewa
- Max Delbruck Centre for Molecular Medicine, Berlin, Germany
| | - Lucia Poncette
- Max Delbruck Centre for Molecular Medicine, Berlin, Germany
| | | |
Collapse
|
4
|
Fuchs EJ, McCurdy SR, Solomon SR, Wang T, Herr MM, Modi D, Grunwald MR, Nishihori T, Kuxhausen M, Fingerson S, McKallor C, Bashey A, Kasamon YL, Bolon YT, Saad A, McGuirk J, Paczesny S, Gadalla SM, Marsh SGE, Shaw BE, Spellman SR, Lee SJ, Petersdorf EW. HLA informs risk predictions after haploidentical stem cell transplantation with posttransplantation cyclophosphamide. Blood 2022; 139:1452-1468. [PMID: 34724567 PMCID: PMC8914182 DOI: 10.1182/blood.2021013443] [Citation(s) in RCA: 66] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Accepted: 10/18/2021] [Indexed: 11/20/2022] Open
Abstract
Hematopoietic cell transplantation from HLA-haploidentical related donors is increasingly used to treat hematologic cancers; however, characteristics of the optimal haploidentical donor have not been established. We studied the role of donor HLA mismatching in graft-versus-host disease (GVHD), disease recurrence, and survival after haploidentical donor transplantation with posttransplantation cyclophosphamide (PTCy) for 1434 acute leukemia or myelodysplastic syndrome patients reported to the Center for International Blood and Marrow Transplant Research. The impact of mismatching in the graft-versus-host vector for HLA-A, -B, -C, -DRB1, and -DQB1 alleles, the HLA-B leader, and HLA-DPB1 T-cell epitope (TCE) were studied using multivariable regression methods. Outcome was associated with HLA (mis)matches at individual loci rather than the total number of HLA mismatches. HLA-DRB1 mismatches were associated with lower risk of disease recurrence. HLA-DRB1 mismatching with HLA-DQB1 matching correlated with improved disease-free survival. HLA-B leader matching and HLA-DPB1 TCE-nonpermissive mismatching were each associated with improved overall survival. HLA-C matching lowered chronic GVHD risk, and the level of HLA-C expression correlated with transplant-related mortality. Matching status at the HLA-B leader and HLA-DRB1, -DQB1, and -DPB1 predicted disease-free survival, as did patient and donor cytomegalovirus serostatus, patient age, and comorbidity index. A web-based tool was developed to facilitate selection of the best haploidentical-related donor by calculating disease-free survival based on these characteristics. In conclusion, HLA factors influence the success of haploidentical transplantation with PTCy. HLA-DRB1 and -DPB1 mismatching and HLA-C, -B leader, and -DQB1 matching are favorable. Consideration of HLA factors may help to optimize the selection of haploidentical related donors.
Collapse
Affiliation(s)
- Ephraim J Fuchs
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins Hospital, Baltimore, MD
| | | | - Scott R Solomon
- Northside Hospital Cancer Institute, Blood and Marrow Transplant Program, Atlanta, GA
| | - Tao Wang
- Department of Medicine, Center for International Blood and Marrow Transplant Research (CIBMTR), Medical College of Wisconsin, Milwaukee, WI
- Division of Biostatistics, Institute for Health and Equity, Medical College of Wisconsin, Milwaukee, WI
| | | | | | - Michael R Grunwald
- Department of Hematologic Oncology and Blood Disorders, Levine Cancer Institute, Atrium Health, Charlotte, NC
| | - Taiga Nishihori
- Department of Blood and Marrow Transplant and Cellular Immunotherapy (BMT CI), Moffitt Cancer Center, Tampa, FL
| | - Michelle Kuxhausen
- CIBMTR, National Marrow Donor Program/Be The Match Foundation, Minneapolis, MN
| | - Stephanie Fingerson
- CIBMTR, National Marrow Donor Program/Be The Match Foundation, Minneapolis, MN
| | - Caroline McKallor
- Division of Clinical Research, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Asad Bashey
- Northside Hospital Cancer Institute, Blood and Marrow Transplant Program, Atlanta, GA
| | - Yvette L Kasamon
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins Hospital, Baltimore, MD
| | - Yung-Tsi Bolon
- CIBMTR, National Marrow Donor Program/Be The Match Foundation, Minneapolis, MN
| | - Ayman Saad
- Division of Hematology, Ohio State University, Columbus, OH
| | - Joseph McGuirk
- Division of Hematologic Malignancies and Cellular Therapeutics, The University of Kansas Cancer Center, Kansas City, KS
| | - Sophie Paczesny
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC
| | - Shahinaz M Gadalla
- Division of Cancer Epidemiology and Genetics, National Institutes of Health, National Cancer Institute, Clinical Genetics Branch, Rockville, MD
| | - Steven G E Marsh
- Anthony Nolan Research Institute-University College London Cancer Institute, Royal Free Campus, London, United Kingdom; and
| | - Bronwen E Shaw
- Department of Medicine, Center for International Blood and Marrow Transplant Research (CIBMTR), Medical College of Wisconsin, Milwaukee, WI
| | - Stephen R Spellman
- CIBMTR, National Marrow Donor Program/Be The Match Foundation, Minneapolis, MN
| | - Stephanie J Lee
- Division of Clinical Research, Fred Hutchinson Cancer Research Center, Seattle, WA
- Department of Medicine, CIBMTR, Medical College of Wisconsin, Milwaukee, WI
| | - Effie W Petersdorf
- Division of Clinical Research, Fred Hutchinson Cancer Research Center, Seattle, WA
| |
Collapse
|
5
|
Poncette L, Bluhm J, Blankenstein T. The role of CD4 T cells in rejection of solid tumors. Curr Opin Immunol 2021; 74:18-24. [PMID: 34619457 PMCID: PMC8933281 DOI: 10.1016/j.coi.2021.09.005] [Citation(s) in RCA: 70] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2021] [Accepted: 09/17/2021] [Indexed: 12/22/2022]
Abstract
Versatility of CD4 T cells enables different attack modes towards cancer cells. Cooperation of CD4 and CD8 T cells renders anti-tumor responses most efficient. Integrating CD4 T cells in cancer therapy will improve clinical outcome.
The focus in cancer immunotherapy has mainly been on CD8 T cells, as they can directly recognize cancer cells. CD4 T cells have largely been neglected, because most cancers lack MHC II expression and cannot directly be recognized by CD4 T cells. Yet, tumor antigens can be captured and cross-presented by MHC II-expressing tumor stromal cells. Recent data suggest that CD4 T cells act as a swiss army knife against tumors. They can kill cancer cells, if they express MHC II, induce tumoricidal macrophages, induces cellular senescence of cancer cells, destroy the tumor vasculature through cytokine release and help CD8 T cells in the effector phase. We foresee a great future for CD4 T cells in the clinic, grafted with tumor antigen specificity by T cell receptor gene transfer, either alone or in combination with engineered CD8 T cells.
Collapse
Affiliation(s)
- Lucia Poncette
- T-knife GmbH, Robert-Rössle-Straße 10, 13125 Berlin, Germany; Max-Delbrück-Center for Molecular Medicine, Robert-Rössle-Straße 10, 13125 Berlin, Germany
| | - Julia Bluhm
- T-knife GmbH, Robert-Rössle-Straße 10, 13125 Berlin, Germany; Max-Delbrück-Center for Molecular Medicine, Robert-Rössle-Straße 10, 13125 Berlin, Germany
| | - Thomas Blankenstein
- Max-Delbrück-Center for Molecular Medicine, Robert-Rössle-Straße 10, 13125 Berlin, Germany.
| |
Collapse
|
6
|
Chi A, He X, Hou L, Nguyen NP, Zhu G, Cameron RB, Lee JM. Classification of Non-Small Cell Lung Cancer's Tumor Immune Micro-Environment and Strategies to Augment Its Response to Immune Checkpoint Blockade. Cancers (Basel) 2021; 13:cancers13122924. [PMID: 34208113 PMCID: PMC8230820 DOI: 10.3390/cancers13122924] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 06/03/2021] [Accepted: 06/09/2021] [Indexed: 12/26/2022] Open
Abstract
Simple Summary Immune checkpoint blockade (ICB) has become a major treatment for lung cancer. Better understanding of the tumor immune micro-environment (TIME) in non-small cell lung cancer (NSCLC) is urgently needed to better treat it with this type of therapy. In this review, we describe and explore how NSCLC’s TIME relates to response to ICB, as well as how to treat those with unresponsive types of TIME, which will significantly impact future research in lung cancer immunotherapy. Abstract Immune checkpoint blockade (ICB) with checkpoint inhibitors has led to significant and durable response in a subset of patients with advanced stage EGFR and ALK wild-type non-small cell lung cancer (NSCLC). This has been consistently shown to be correlated with the unique characteristics of each patient’s tumor immune micro-environment (TIME), including the composition and distribution of the tumor immune cell infiltrate; the expression of various checkpoints by tumor and immune cells, such as PD-L1; and the presence of various cytokines and chemokines. In this review, the classification of various types of TIME that are present in NSCLC and their correlation with response to ICB in NSCLC are discussed. This is conducted with a focus on the characteristics and identifiable biomarkers of different TIME subtypes that may also be used to predict NSCLC’s clinical response to ICB. Finally, treatment strategies to augment response to ICB in NSCLC with unresponsive types of TIME are explored.
Collapse
Affiliation(s)
- Alexander Chi
- Department of Radiation Oncology, Beijing Chest Hospital, Capital Medical University, Beijing 101100, China
- Department of Radiation Oncology, Jiangsu Cancer Hospital, Nanjing Medical University, Nanjing 210009, China
- Correspondence: (A.C.); (X.H.)
| | - Xia He
- Department of Radiation Oncology, Jiangsu Cancer Hospital, Nanjing Medical University, Nanjing 210009, China
- Correspondence: (A.C.); (X.H.)
| | - Lin Hou
- Center for Statistical Science, Tsinghua University, Beijing 100084, China;
| | - Nam P. Nguyen
- Department of Radiation Oncology, Howard University, Washington, DC 20060, USA;
| | - Guangying Zhu
- Department of Radiation Oncology, China-Japan Friendship Hospital, Beijing 100029, China;
| | - Robert B. Cameron
- Division of Thoracic Surgery, Department of Surgery, University of California at Los Angeles, Los Angeles, CA 90095, USA; (R.B.C.); (J.M.L.)
| | - Jay M. Lee
- Division of Thoracic Surgery, Department of Surgery, University of California at Los Angeles, Los Angeles, CA 90095, USA; (R.B.C.); (J.M.L.)
| |
Collapse
|
7
|
Lippitz BE, Harris RA. A translational concept of immuno-radiobiology. Radiother Oncol 2019; 140:116-124. [PMID: 31271996 DOI: 10.1016/j.radonc.2019.06.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Revised: 05/30/2019] [Accepted: 06/02/2019] [Indexed: 02/07/2023]
Abstract
BACKGROUND Traditional concepts of radiobiology model the direct radiation-induced cellular cytotoxicity but are not focused on late and sustained effects of radiation. Recent experimental data show the close involvement of immunological processes. METHODS Based on systematic PubMed searches, experimental data on immunological radiation effects are summarized and analyzed in a non-quantitative descriptive manner to provide a translational perspective on the immuno-modulatory impact of radiation in cancer. RESULTS Novel experimental findings document that sustained radiation effects are ultimately mediated through systemic factors such as cytotoxic CD8+ T cells and involve a local immuno-stimulation. Increased tumor infiltration of CD8+ T cell is a prerequisite for long-term radiation effects. CD8+ T cell depletion induces radio-resistance in experimental tumors. The proposed sequence of events involves radiation-damaged cells that release HMGB1, which activates macrophages via TLR4 to a local immuno-stimulation via TNF, which contributes to maturation of DCs. The mature DCs migrate to lymph nodes where they trigger effective CD8+ T cell responses. Radiation effects are boosted, when the physiological self-terminating negative feedback of immune reactions is antagonised via blocking of TGF-β or via checkpoint inhibition with involvement of CD8+ T cells as common denominator. CONCLUSION The concept of immuno-radiobiology emphasizes the necessity for a functional integrity of APCs and T cells for the long-term effects of radiotherapy. Local irradiation at higher doses induces tumor infiltration of CD8+ T cells, which can be boosted by immunotherapy. More systematic research is warranted to better understand the immunological effects of escalating radiation doses.
Collapse
Affiliation(s)
- Bodo E Lippitz
- Dept. of Clinical Neuroscience, Karolinska Institute, Centre for Molecular Medicine L8:04, Karolinska University Hospital, Stockholm, Sweden; Interdisciplinary Centre for Radiosurgery (ICERA), Hamburg, Germany.
| | - Robert A Harris
- Dept. of Clinical Neuroscience, Karolinska Institute, Centre for Molecular Medicine L8:04, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
8
|
Rataj F, Kraus FBT, Chaloupka M, Grassmann S, Heise C, Cadilha BL, Duewell P, Endres S, Kobold S. PD1-CD28 Fusion Protein Enables CD4+ T Cell Help for Adoptive T Cell Therapy in Models of Pancreatic Cancer and Non-hodgkin Lymphoma. Front Immunol 2018; 9:1955. [PMID: 30214445 PMCID: PMC6125378 DOI: 10.3389/fimmu.2018.01955] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Accepted: 08/08/2018] [Indexed: 01/22/2023] Open
Abstract
Background: Interaction of the programmed death receptor 1 (PD-1) and its ligand, PD-L1, suppresses T cell activity and permits tumors to evade T cell-mediated immune surveillance. We have recently demonstrated that antigen-specific CD8+ T cells transduced with a PD1-CD28 fusion protein are protected from PD-1-mediated inhibition. We have now investigated the potential of PD1-CD28 fusion protein-transduced CD4+ T cells alone or in combination with CD8+ T cells for immunotherapy of pancreatic cancer and non-Hodgkin lymphoma. Methods: OVA-specific CD4+ and CD8+ were retrovirally transduced with the PD1-CD28 fusion protein. Cytokine release, proliferation, cytotoxic activity, and phenotype of transduced T cells were assessed in the context of Panc02-OVA (murine pancreatic cancer model) and E.G7-PD-L1 (murine T cell lymphoma model) cells. Results: Stimulation of PD1-CD28 fusion protein-transduced CD4+ T cells with anti-CD3 and recombinant PD-L1 induced specific T cell activation, as measured by IFN-y release and T cell proliferation. Coculture with Panc02-OVA or E.G7-PD-L1 tumor cells also led to specific activation of CD4+ T cells. Cytokine release and T cell proliferation was most effective when tumor cells simultaneously encountered genetically engineered CD4+ and CD8+ T cells. Synergy between both cell populations was also observed for specific tumor cell lysis. T cell cytotoxicity was mediated via granzyme B release and mediated enhanced tumor control in vivo. Transduced CD4+ and CD8+ T cells in co-culture with tumor cells developed a predominant central memory phenotype over time. Different ratios of CD4+ and CD8+ transduced T cells led to a significant increase of IFN-y and IL-2 secretion positively correlating with CD4+ T cell numbers used. Mechanistically, IL-2 and MHC-I were central to the synergistic activity of CD4+ and CD8+ T cells, since neutralization of IL-2 prevented the crosstalk between these cell populations. Conclusion: PD1-CD28 fusion protein-transduced CD4+ T cells significantly improved anti-tumoral effect of fusion protein-transduced CD8+ T cells. Thus, our results indicate that PD1-CD28 fusion protein-transduced CD4+ T cells have the potential to overcome the PD-1-PD-L1 immunosuppressive axis in pancreatic cancer and non-Hodgkin lymphoma.
Collapse
Affiliation(s)
- Felicitas Rataj
- Center of Integrated Protein Science Munich (CIPS-M) and Division of Clinical Pharmacology, Department of Medicine IV, Klinikum der Universität München, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Fabian B T Kraus
- Center of Integrated Protein Science Munich (CIPS-M) and Division of Clinical Pharmacology, Department of Medicine IV, Klinikum der Universität München, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Michael Chaloupka
- Center of Integrated Protein Science Munich (CIPS-M) and Division of Clinical Pharmacology, Department of Medicine IV, Klinikum der Universität München, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Simon Grassmann
- Center of Integrated Protein Science Munich (CIPS-M) and Division of Clinical Pharmacology, Department of Medicine IV, Klinikum der Universität München, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Constanze Heise
- Center of Integrated Protein Science Munich (CIPS-M) and Division of Clinical Pharmacology, Department of Medicine IV, Klinikum der Universität München, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Bruno L Cadilha
- Center of Integrated Protein Science Munich (CIPS-M) and Division of Clinical Pharmacology, Department of Medicine IV, Klinikum der Universität München, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Peter Duewell
- Center of Integrated Protein Science Munich (CIPS-M) and Division of Clinical Pharmacology, Department of Medicine IV, Klinikum der Universität München, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Stefan Endres
- Center of Integrated Protein Science Munich (CIPS-M) and Division of Clinical Pharmacology, Department of Medicine IV, Klinikum der Universität München, Member of the German Center for Lung Research (DZL), Munich, Germany.,German Cancer Research Center (DKTK), Partner Site Munich, Heidelberg, Germany
| | - Sebastian Kobold
- Center of Integrated Protein Science Munich (CIPS-M) and Division of Clinical Pharmacology, Department of Medicine IV, Klinikum der Universität München, Member of the German Center for Lung Research (DZL), Munich, Germany.,German Cancer Research Center (DKTK), Partner Site Munich, Heidelberg, Germany
| |
Collapse
|
9
|
Epidemiology and biology of relapse after stem cell transplantation. Bone Marrow Transplant 2018; 53:1379-1389. [PMID: 29670211 DOI: 10.1038/s41409-018-0171-z] [Citation(s) in RCA: 91] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2017] [Revised: 02/07/2018] [Accepted: 03/12/2018] [Indexed: 12/25/2022]
|
10
|
Shi X, Zhang X, Li J, Mo L, Zhao H, Zhu Y, Hu Z, Gao J, Tan W. PD-1 blockade enhances the antitumor efficacy of GM-CSF surface-modified bladder cancer stem cells vaccine. Int J Cancer 2017; 142:2106-2117. [PMID: 29243219 DOI: 10.1002/ijc.31219] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2017] [Revised: 10/26/2017] [Accepted: 12/07/2017] [Indexed: 01/10/2023]
Affiliation(s)
- Xiaojun Shi
- Department of Urology; Nanfang Hospital, Southern Medical University; Guangzhou China
| | - Xinji Zhang
- Department of Urology; Shunde People's Hospital, Southern Medical University; Guangdong China
| | - Jinlong Li
- Institute of Biotherapy, School of Biotechnology, Southern Medical University; Guangzhou China
| | - Lijun Mo
- Institute of Biotherapy, School of Biotechnology, Southern Medical University; Guangzhou China
| | - Hongfan Zhao
- Department of Urology; Nanfang Hospital, Southern Medical University; Guangzhou China
| | - Yongtong Zhu
- Department of Urology; Nanfang Hospital, Southern Medical University; Guangzhou China
| | - Zhiming Hu
- Institute of Biotherapy, School of Biotechnology, Southern Medical University; Guangzhou China
| | - Jimin Gao
- Zhejiang Provincial Key Laboratory of Medical Genetics, School of Life Sciences; Wenzhou Medical University; Wenzhou China
| | - Wanlong Tan
- Department of Urology; Nanfang Hospital, Southern Medical University; Guangzhou China
| |
Collapse
|
11
|
Wenger A, Werlenius K, Hallner A, Thorén FB, Farahmand D, Tisell M, Smits A, Rydenhag B, Jakola AS, Carén H. Determinants for Effective ALECSAT Immunotherapy Treatment on Autologous Patient-Derived Glioblastoma Stem Cells. Neoplasia 2017; 20:25-31. [PMID: 29190492 PMCID: PMC5715204 DOI: 10.1016/j.neo.2017.10.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2017] [Revised: 10/24/2017] [Accepted: 10/24/2017] [Indexed: 12/13/2022] Open
Abstract
Glioblastoma (GBM) is the most aggressive primary brain tumor with a median survival of less than 15 months, emphasizing the need for better treatments. Immunotherapy as a treatment for improving or aiding the patient's own immune defense to target the tumor has been suggested for GBM. A randomized clinical trial of adoptive cell transfer using ALECSAT (Autologous Lymphoid Effector Cells Specific Against Tumor Cells) is currently ongoing in Sweden. Here we performed a paired pre-clinical study to investigate the composition and in vitro effect of ALECSAT and identify determinants for the effect using autologous GBM-derived cancer stem cells (CSC), immunocytochemistry and flow cytometry. We show a clear dose-response relationship of ALECSAT on CSC, suggesting that the number of infused cells is of importance. In addition, the in vitro effect of ALECSAT on CSC correlated significantly to the blood count of T helper (Th) cells in the patient indicating a potential benefit of collecting cells for ALECSAT preparation at an even earlier stage when patients generally have a better blood count. The factors identified in this study will be important to consider in the design of future immunotherapy trials to achieve prolonged survival.
Collapse
Affiliation(s)
- Anna Wenger
- Sahlgrenska Cancer Center, Department of Pathology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Sweden
| | - Katja Werlenius
- Department of Oncology, Sahlgrenska University Hospital, Gothenburg, Sweden; Sahlgrenska Cancer Center, Department of Oncology, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Sweden
| | - Alexander Hallner
- TIMM laboratory, Sahlgrenska Cancer Center, University of Gothenburg, Gothenburg, Sweden
| | - Fredrik Bergh Thorén
- TIMM laboratory, Sahlgrenska Cancer Center, University of Gothenburg, Gothenburg, Sweden
| | - Dan Farahmand
- Department of Neurosurgery, Sahlgrenska University Hospital, Gothenburg, Sweden; Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Magnus Tisell
- Department of Neurosurgery, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Anja Smits
- Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden; Department of Neuroscience, Neurology, Uppsala University, University Hospital, Uppsala, Sweden
| | - Bertil Rydenhag
- Department of Neurosurgery, Sahlgrenska University Hospital, Gothenburg, Sweden; Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Asgeir S Jakola
- Department of Neurosurgery, Sahlgrenska University Hospital, Gothenburg, Sweden; Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden; Department of Neurosurgery, St. Olavs University Hospital, Trondheim, Norway
| | - Helena Carén
- Sahlgrenska Cancer Center, Department of Pathology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Sweden.
| |
Collapse
|
12
|
Maloney E, Khokhlova T, Pillarisetty VG, Schade GR, Repasky EA, Wang YN, Giuliani L, Primavera M, Hwang JH. Focused ultrasound for immuno-adjuvant treatment of pancreatic cancer: An emerging clinical paradigm in the era of personalized oncotherapy. Int Rev Immunol 2017; 36:338-351. [PMID: 28961038 PMCID: PMC6224292 DOI: 10.1080/08830185.2017.1363199] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Current clinical treatment regimens, including many emergent immune strategies (e.g., checkpoint inhibitors) have done little to affect the devastating course of pancreatic ductal adenocarcinoma (PDA). Clinical trials for PDA often employ multi-modal treatment, and have started to incorporate stromal-targeted therapies, which have shown promising results in early reports. Focused ultrasound (FUS) is one such therapy that is uniquely equipped to address local and systemic limitations of conventional cancer therapies as well as emergent immune therapies for PDA. FUS methods can non-invasively generate mechanical and/or thermal effects that capitalize on the unique oncogenomic/proteomic signature of a tumor. Potential benefits of FUS therapy for PDA include: (1) emulsification of targeted tumor into undenatured antigens in situ, increasing dendritic cell maturation, and increasing intra-tumoral CD8+/ T regulatory cell ratio and CD8+ T cell activity; (2) reduction in intra-tumoral hypoxic stress; (3) modulation of tumor cell membrane protein localization to enhance immunogenicity; (4) modulation of the local cytokine milieu toward a Th1-type inflammatory profile; (5) up-regulation of local chemoattractants; (6) remodeling the tumor stroma; (7) localized delivery of exogenously packaged immune-stimulating antigens, genes and therapeutic drugs. While not all of these results have been studied in experimental PDA models to date, the principles garnered from other solid tumor and disease models have direct relevance to the design of optimal FUS protocols for PDA. In this review, we address the pertinent limitations in current and emergent immune therapies that can be improved with FUS therapy for PDA.
Collapse
Affiliation(s)
- Ezekiel Maloney
- a Department of Radiology , University of Washington , Seattle WA , USA
| | - Tanya Khokhlova
- b Department of Medicine Division of Gastroenterology , University of Washington , Seattle WA , USA
| | | | - George R Schade
- d Department of Urology , University of Washington , Seattle WA , USA
| | - Elizabeth A Repasky
- e Department of Immunology , Roswell Park Cancer Institute , Buffalo NY , USA
| | - Yak-Nam Wang
- f Applied Physics Laboratory , University of Washington , Seattle WA , USA
| | - Lorenzo Giuliani
- g School of Medicine , The Sapienza University of Rome , Rome , Italy
| | - Matteo Primavera
- h School of Medicine , The Sapienza University of Rome , Rome , Italy
| | - Joo Ha Hwang
- i Department of Medicine Division of Gastroenterology , University of Washington , Seattle WA , USA
| |
Collapse
|
13
|
Xu R, Zhao X, Zhao Y, Chen B, Sun L, Xu C, Shen B, Wang M, Xu W, Zhu W. Enhanced gastric cancer growth potential of mesenchymal stem cells derived from gastric cancer tissues educated by CD4 + T cells. Cell Prolif 2017; 51:e12399. [PMID: 29057538 DOI: 10.1111/cpr.12399] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2017] [Accepted: 09/24/2017] [Indexed: 12/18/2022] Open
Abstract
OBJECTIVES Gastric cancer mesenchymal stem cells (GC-MSCs) can promote the development of tumour growth. The tumour-promoting role of tumour-associated MSCs and T cells has been demonstrated. T cells as the major immune cells may influence and induce a pro-tumour phenotype in MSCs. This study focused on whether CD4+ T cells can affect GC-MSCs to promote gastric cancer growth. MATERIALS AND METHODS CD4+ T cells upregulation of programmed death ligand 1 (PD-L1) expression in GC-MSCs through the phosphorylated signal transducer and activator of transcription (p-STAT3) signalling pathway was confirmed by immunofluorescence, western blotting and RT-PCR. Migration of GC cells was detected by Transwell migration assay, and apoptosis of GC cells was measured by flow cytometry using annexin V/propidium iodide double staining. CD4+ T cell-primed GC-MSCs promoted GC growth in a subcutaneously transplanted tumour model in BALB/c nu/nu mice. RESULTS Gastric cancer mesenchymal stem cells stimulated by activated CD4+ T cells promoted migration of GC cells and enhanced GC growth potential in BALB/c nu/nu xenografts. PD-L1 upregulation of GC-MSCs stimulated by CD4+ T cells was mediated through the p-STAT3 signalling pathway. CD4+ T cells-primed GC-MSCs have greater GC volume and growth rate-promoting role than GC-MSCs, with cancer cell-intrinsic PD-1/mammalian target of rapamycin (mTOR) signalling activation. CONCLUSIONS This study showed that GC-MSCs are plastic. The immunophenotype of GC-MSCs stimulated by CD4+ T cells has major changes that may influence tumour cell growth. This research was based on the interaction between tumour cells, MSCs and immune cells, providing a new understanding of the development and immunotherapy of GC.
Collapse
Affiliation(s)
- Rongman Xu
- School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, China.,Department of Clinical Laboratory, Haian People's Hospital, Haian, Jiangsu, China
| | - Xiangdong Zhao
- Zhenjiang Provincial Blood Center, Zhenjiang, Jiangsu, China
| | - Yuanyuan Zhao
- School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Bin Chen
- School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Li Sun
- School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Changgen Xu
- Zhenjiang Provincial Blood Center, Zhenjiang, Jiangsu, China
| | - Bo Shen
- Department of Oncology, Jiangsu Cancer Hospital, Affiliated to Nanjing Medical University, Nanjing, Jiangsu, China
| | - Mei Wang
- School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Wenrong Xu
- School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Wei Zhu
- School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, China
| |
Collapse
|
14
|
Shi X, Zhang X, Li J, Zhao H, Mo L, Shi X, Hu Z, Gao J, Tan W. PD-1/PD-L1 blockade enhances the efficacy of SA-GM-CSF surface-modified tumor vaccine in prostate cancer. Cancer Lett 2017; 406:27-35. [PMID: 28797844 DOI: 10.1016/j.canlet.2017.07.029] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Revised: 07/19/2017] [Accepted: 07/30/2017] [Indexed: 02/07/2023]
Abstract
Program death receptor-1 (PD-1)/program death ligand 1 (PD-L1) signaling plays an important role in tumor adaptive immune resistance. The streptavidin-granulocyte-macrophage colony stimulating factor (SA-GM-CSF) surface-modified tumor cells vaccine developed through our novel protein-anchor technology could significantly promote the activation of dendritic cells. Although GM-CSF vaccine could significantly increase the number of tumor-specific CD8+T-cells, the majority of these CD8+T-cells expressed PD-1. Moreover, GM-CSF vaccine up-regulated the PD-L1 expression of tumor cells, resulting in immune resistance. Adding PD-1/PD-L1 blockade to GM-CSF vaccine therapy could significantly increase the population of CD4+ T, CD8+ T and CD8+ IFN-γ+ T but not CD4+ Foxp3+ T-cells and induced the highest production of IFN-γ. PD-1/PD-L1 blockade could effectively rescue the tumor-specific T lymphocytes generated by the GM-CSF vaccine, resulting in consistent tumor rejection. Taken together, PD-1/PD-L1 blockade combined with SA-GM-CSF-modified vaccine could effectively induce a strong specific antitumor immune response against prostate cancer.
Collapse
Affiliation(s)
- Xiaojun Shi
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xinji Zhang
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou, China; Department of Urology, Shunde People's Hospital, Southern Medical University, Guangdong, China
| | - Jinlong Li
- Institute of Biotherapy, School of Biotechnology, Southern Medical University, Guangzhou, China
| | - Hongfan Zhao
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Lijun Mo
- Institute of Biotherapy, School of Biotechnology, Southern Medical University, Guangzhou, China
| | - Xianghua Shi
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Zhiming Hu
- Institute of Biotherapy, School of Biotechnology, Southern Medical University, Guangzhou, China
| | - Jimin Gao
- Zhejiang Provincial Key Laboratory of Medical Genetics, School of Life Sciences, Wenzhou Medical College, Wenzhou, China
| | - Wanlong Tan
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou, China.
| |
Collapse
|
15
|
De La Maza L, Wu M, Wu L, Yun H, Zhao Y, Cattral M, McCart A, Cho BJ, de Perrot M. In Situ Vaccination after Accelerated Hypofractionated Radiation and Surgery in a Mesothelioma Mouse Model. Clin Cancer Res 2017; 23:5502-5513. [PMID: 28606922 DOI: 10.1158/1078-0432.ccr-17-0438] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Revised: 04/27/2017] [Accepted: 06/05/2017] [Indexed: 11/16/2022]
Abstract
Purpose: How best to sequence and integrate immunotherapy into standard of care is currently unknown. Clinical protocols with accelerated nonablative hypofractionated radiation followed by surgery could provide an opportunity to implement immune checkpoint blockade.Experimental Design: We therefore assessed the impact of nonablative hypofractionated radiation on the immune system in combination with surgery in a mouse mesothelioma model. Blunt surgery (R1 resection) was used to analyze the short-term effect, and radical surgery (R0 resection) was used to analyze the long-term effect of this radiation protocol before surgery.Results: Nonablative hypofractionated radiation led to a specific immune activation against the tumor associated with significant upregulation of CD8+ T cells, limiting the negative effect of an incomplete resection. The same radiation protocol performed 7 days before radical surgery led to a long-term antitumor immune protection that was primarily driven by CD4+ T cells. Radical surgery alone or with a short course of nonablative radiation completed 24 hours before radical surgery did not provide this vaccination effect. Combining this radiation protocol with CTLA-4 blockade provided better results than radiation alone. The effect of PD-1 or PD-L1 blockade with this radiation protocol was less effective than the combination with CTLA-4 blockade.Conclusions: A specific activation of the immune system against the tumor contributes to the benefit of accelerated, hypofractionated radiation before surgery. Nonablative hypofractionated radiation combined with surgery provides an opportunity to introduce immune checkpoint blockades in the clinical setting. Clin Cancer Res; 23(18); 5502-13. ©2017 AACR.
Collapse
Affiliation(s)
- Luis De La Maza
- Latner Thoracic Surgery Research Laboratories, Toronto General Research Institute, University of Toronto, Toronto, Ontario, Canada
| | - Matthew Wu
- Latner Thoracic Surgery Research Laboratories, Toronto General Research Institute, University of Toronto, Toronto, Ontario, Canada
| | - Licun Wu
- Latner Thoracic Surgery Research Laboratories, Toronto General Research Institute, University of Toronto, Toronto, Ontario, Canada
| | - Hana Yun
- Latner Thoracic Surgery Research Laboratories, Toronto General Research Institute, University of Toronto, Toronto, Ontario, Canada
| | - Yidan Zhao
- Latner Thoracic Surgery Research Laboratories, Toronto General Research Institute, University of Toronto, Toronto, Ontario, Canada
| | - Mark Cattral
- Department of General Surgery, Toronto General Hospital, University Health Network, Toronto, Ontario, Canada
| | - Andrea McCart
- Department of General Surgery, Toronto General Hospital, University Health Network, Toronto, Ontario, Canada
| | - Bc John Cho
- Department of Radiation Oncology, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Marc de Perrot
- Latner Thoracic Surgery Research Laboratories, Toronto General Research Institute, University of Toronto, Toronto, Ontario, Canada. .,Division of Thoracic Surgery, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| |
Collapse
|