1
|
Nesbitt C, Van Der Walt A, Butzkueven H, Devitt B, Jokubaitis VG. Multiple sclerosis and cancer: Navigating a dual diagnosis. Mult Scler 2024; 30:1714-1736. [PMID: 39347791 DOI: 10.1177/13524585241274523] [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] [Indexed: 10/01/2024]
Abstract
Healthcare breakthroughs are extending the lives of multiple sclerosis (MS) patients and cancer survivors, creating a growing cohort of individuals navigating a dual diagnosis. Determining the relationship between MS and cancer risk remains challenging, with inconclusive findings confounded by age, risk exposures, comorbidities, genetics and the ongoing introduction of new MS disease-modifying therapies (DMTs) across study periods.This research places significant emphasis on cancer survival, with less attention given to the impact on MS outcomes. Our review explores the existing literature on MS, cancer risk and the intersection of DMTs and cancer treatments. We aim to navigate the complexities of managing MS in cancer survivors to optimise outcomes for both conditions. Continuous research and the formulation of treatment guidelines are essential for guiding future care. Collaboration between neuro-immunology and oncology is crucial, with a need to establish databases for retrospective and ultimately prospective analysis of outcomes in these rapidly evolving fields.
Collapse
Affiliation(s)
- Cassie Nesbitt
- Department of Neuroscience, School of Translational Medicine, Monash University, Melbourne, VIC, Australia
- Department of Neurology, MSNI Multiple Sclerosis and Neuro-Immunology, Alfred Health, Melbourne, VIC, Australia
- Department of Neuroscience, Monash University, The Alfred Centre, Melbourne, VIC, Australia
| | - Anneke Van Der Walt
- Department of Neuroscience, School of Translational Medicine, Monash University, Melbourne, VIC, Australia
- Department of Neurology, MSNI Multiple Sclerosis and Neuro-Immunology, Alfred Health, Melbourne, VIC, Australia
| | - Helmut Butzkueven
- Department of Neuroscience, School of Translational Medicine, Monash University, Melbourne, VIC, Australia
- Department of Neurology, MSNI Multiple Sclerosis and Neuro-Immunology, Alfred Health, Melbourne, VIC, Australia
| | - Bianca Devitt
- Department of Oncology, Eastern Health Clinical School, Monash University, Melbourne, VIC, Australia
- Oncology Clinical Trials Unit, School of Translational Medicine, Monash University, Melbourne, VIC, Australia
| | - Vilija G Jokubaitis
- Department of Neuroscience, School of Translational Medicine, Monash University, Melbourne, VIC, Australia
- Department of Neurology, MSNI Multiple Sclerosis and Neuro-Immunology, Alfred Health, Melbourne, VIC, Australia
- Department of Neuroscience, Monash University, The Alfred Centre, Melbourne, VIC, Australia
| |
Collapse
|
2
|
Dixon-Douglas J, Virassamy B, Clarke K, Hun M, Luen SJ, Savas P, van Geelen CT, David S, Francis PA, Salgado R, Michiels S, Loi S. Sustained lymphocyte decreases after treatment for early breast cancer. NPJ Breast Cancer 2024; 10:94. [PMID: 39433772 PMCID: PMC11493948 DOI: 10.1038/s41523-024-00698-4] [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: 03/26/2024] [Accepted: 09/19/2024] [Indexed: 10/23/2024] Open
Abstract
The role of adaptive immunity in long-term outcomes in early breast cancer is increasingly recognised. Standard (neo)adjuvant chemotherapy can have adverse effects on immune cells. We conducted a retrospective longitudinal study of full blood counts (FBC) of 200 patients receiving (neo)adjuvant chemotherapy for early breast cancer at a single institution. FBC results at four time points from pre-treatment to 12 months post-chemotherapy were analysed. Flow cytometry was performed for patients with matched pre- and post-chemotherapy peripheral blood mononuclear cell samples. A significant decrease in absolute lymphocyte count at 12 months post-chemotherapy was observed (p < 0.01), most pronounced in pre-menopausal patients (n = 73; p < 0.01), patients receiving dose-dense chemotherapy regimens (n = 60; p < 0.01) and patients receiving adjuvant radiotherapy (n = 147, p < 0.01). In pre-menopausal patients, significant changes in CD4+ T cells subsets post-chemotherapy were observed. Further investigation, including long-term clinical outcomes, is needed to meaningfully improve long-term anti-tumour immunity.
Collapse
Affiliation(s)
- Julia Dixon-Douglas
- Division of Cancer Research, Peter MacCallum Cancer Centre, Melbourne, Australia
- Sir Peter MacCallum Department of Medical Oncology, The University of Melbourne, Parkville, Australia
- Institut Gustave Roussy, INSERM U981, PRISM Center, F-94805, Villejuif, France
| | - Balaji Virassamy
- Division of Cancer Research, Peter MacCallum Cancer Centre, Melbourne, Australia
| | - Kylie Clarke
- Division of Cancer Research, Peter MacCallum Cancer Centre, Melbourne, Australia
| | - Michael Hun
- Division of Cancer Research, Peter MacCallum Cancer Centre, Melbourne, Australia
| | - Stephen J Luen
- Division of Cancer Research, Peter MacCallum Cancer Centre, Melbourne, Australia
- Sir Peter MacCallum Department of Medical Oncology, The University of Melbourne, Parkville, Australia
| | - Peter Savas
- Division of Cancer Research, Peter MacCallum Cancer Centre, Melbourne, Australia
- Sir Peter MacCallum Department of Medical Oncology, The University of Melbourne, Parkville, Australia
| | | | - Steven David
- Sir Peter MacCallum Department of Medical Oncology, The University of Melbourne, Parkville, Australia
- Division of Radiation Oncology, Peter MacCallum Cancer Centre, Melbourne, Australia
| | - Prudence A Francis
- Sir Peter MacCallum Department of Medical Oncology, The University of Melbourne, Parkville, Australia
| | - Roberto Salgado
- Division of Cancer Research, Peter MacCallum Cancer Centre, Melbourne, Australia
- Department of Pathology, ZAS-Hospitals, Antwerp, Belgium
| | - Stefan Michiels
- Institut Gustave Roussy, INSERM U981, PRISM Center, F-94805, Villejuif, France
| | - Sherene Loi
- Division of Cancer Research, Peter MacCallum Cancer Centre, Melbourne, Australia.
- Sir Peter MacCallum Department of Medical Oncology, The University of Melbourne, Parkville, Australia.
| |
Collapse
|
3
|
Azizi G, Van den Broek B, Ishikawa LLW, Naziri H, Yazdani R, Zhang GX, Ciric B, Rostami A. IL-7Rα on CD4 + T cells is required for their survival and the pathogenesis of experimental autoimmune encephalomyelitis. J Neuroinflammation 2024; 21:253. [PMID: 39380064 PMCID: PMC11460225 DOI: 10.1186/s12974-024-03224-2] [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: 05/17/2024] [Accepted: 09/04/2024] [Indexed: 10/10/2024] Open
Abstract
BACKGROUND The IL-7 receptor alpha (IL-7Rα) binds both IL-7 and thymic stromal lymphopoietin (TSLP). IL-7Rα is essential for the development and survival of naive CD4+ T cells and their differentiation to effector/memory CD4+ T cells. Mice lacking IL-7Rα have severe lymphopenia and are resistant to experimental autoimmune encephalomyelitis (EAE), a model for multiple sclerosis. However, it has been reported that IL-7Rα on peripheral CD4+ T cells is disposable for their maintenance and EAE pathogenesis, which does not align with the body of knowledge on the role of IL-7Rα in the biology of CD4+ T cells. Given that a definitive study on this important topic is lacking, we revisited it using a novel approach, an inducible knockout of the IL-7Rα gene in CD4+ T cells. METHODS We generated Il7rafl/fl/CD4CreERT2 double transgenic mouse line (henceforth CD4ΔIl7ra), susceptible to tamoxifen-induced knockout of the IL-7Rα gene in CD4+ T cells. CD4ΔIl7ra mice were immunized with MOG35 - 55 for EAE induction and monitored for disease development. The expression of IL-7Rα, CD4+ T cell numbers, and MOG35 - 55-specific CD4+ T cell response was evaluated in the central nervous system (CNS) and lymphoid tissues by flow cytometry. Additionally, splenocytes of CD4ΔIl7ra mice were stimulated with MOG35 - 55 to assess their proliferative response and cytokine production by T helper cells. RESULTS Loss of IL-7Rα from the surface of CD4+ T cells in CD4ΔIl7ra mice was virtually complete several days after tamoxifen treatment. The loss of IL-7Rα in CD4+ T cells led to a gradual and substantial decrease in their numbers in both non-immunized and immunized CD4ΔIl7ra mice, followed by slow repopulation up to the initial numbers. CD4ΔIl7ra mice did not develop EAE. We found a decrease in the total numbers of TNF-, IFN-γ-, IL-17 A-, and GM-CSF-producing CD4+ T cells and regulatory T cells in the spleens and CNS of immunized CD4ΔIl7ra mice. Tracking MOG35 - 55-specific CD4+ T cells revealed a significant reduction in their numbers in CD4ΔIl7ra mice and decreased proliferation and cytokine production in response to MOG35 - 55. CONCLUSION Our study demonstrates that IL-7Rα on peripheral CD4+ T cells is essential for their maintenance, immune response, and EAE pathogenesis.
Collapse
MESH Headings
- Animals
- Encephalomyelitis, Autoimmune, Experimental/immunology
- Encephalomyelitis, Autoimmune, Experimental/pathology
- Encephalomyelitis, Autoimmune, Experimental/metabolism
- Encephalomyelitis, Autoimmune, Experimental/chemically induced
- Encephalomyelitis, Autoimmune, Experimental/genetics
- CD4-Positive T-Lymphocytes/immunology
- CD4-Positive T-Lymphocytes/metabolism
- Mice
- Receptors, Interleukin-7/metabolism
- Receptors, Interleukin-7/genetics
- Mice, Transgenic
- Mice, Inbred C57BL
- Myelin-Oligodendrocyte Glycoprotein/toxicity
- Myelin-Oligodendrocyte Glycoprotein/immunology
- Cell Survival/physiology
- Cell Survival/drug effects
- Peptide Fragments/toxicity
- Peptide Fragments/immunology
- Peptide Fragments/metabolism
- Mice, Knockout
- Cytokines/metabolism
Collapse
Affiliation(s)
- Gholamreza Azizi
- Department of Neurology, Thomas Jefferson University, 900 Walnut Street, Suite 300, Philadelphia, PA, 19107, USA
| | - Bram Van den Broek
- Department of Neurology, Thomas Jefferson University, 900 Walnut Street, Suite 300, Philadelphia, PA, 19107, USA
| | | | - Hamed Naziri
- Department of Neurology, Thomas Jefferson University, 900 Walnut Street, Suite 300, Philadelphia, PA, 19107, USA
| | - Reza Yazdani
- Department of Neurology, Thomas Jefferson University, 900 Walnut Street, Suite 300, Philadelphia, PA, 19107, USA
| | - Guang-Xian Zhang
- Department of Neurology, Thomas Jefferson University, 900 Walnut Street, Suite 300, Philadelphia, PA, 19107, USA
| | - Bogoljub Ciric
- Department of Neurology, Thomas Jefferson University, 900 Walnut Street, Suite 300, Philadelphia, PA, 19107, USA
| | - Abdolmohamad Rostami
- Department of Neurology, Thomas Jefferson University, 900 Walnut Street, Suite 300, Philadelphia, PA, 19107, USA.
| |
Collapse
|
4
|
Jia W, Wu J, Zhang H, Wu Y, Liu D, Wang Z, Wang X, Li C, Hao C. Advancing treatment efficacy: combined therapy of eribulin, anlotinib, and camrelizumab in advanced or metastatic retroperitoneal liposarcoma. Ther Adv Med Oncol 2024; 16:17588359241276968. [PMID: 39281972 PMCID: PMC11402105 DOI: 10.1177/17588359241276968] [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] [Received: 02/11/2024] [Accepted: 08/06/2024] [Indexed: 09/18/2024] Open
Abstract
Background Retroperitoneal liposarcoma (RLPS) typically shows limited response to standard chemotherapy, presenting a challenge in treating advanced or metastatic RLPS. Objective This study aimed to evaluate the potential advantages of a combined therapeutic strategy utilizing eribulin, anlotinib, and camrelizumab. Design Between December 2020 and March 2023, this retrospective study enrolled patients with advanced or metastatic RLPS who received treatment at Peking University Cancer Hospital Sarcoma Center. The treatment regimen involved eribulin plus anlotinib and camrelizumab administered every 3 weeks (Q3W). Methods Efficacy was assessed following the Response Evaluation Criteria in Solid Tumors version 1.1, while safety was evaluated using the Common Terminology Criteria for Adverse Events version 5.0. Results The study included 47 patients with RLPS with a median age of 55.5 years. Patients received a median of 4.5 (range, 2-21) cycles of treatment. Notably, partial response was observed in 8 patients (18.2%), while 25 (56.8%) exhibited stable disease. The objective response rate (ORR) and disease control rate were 18.2% and 75%, respectively. Significant differences in ORR were observed among histological subtypes (well-differentiated vs de-differentiated vs myxoid: 0 vs 17.9% vs 50%; p = 0.039). Six patients underwent surgery before disease progression, and one patient with myxoid liposarcoma (MLPS) had a pathological complete response. With a median follow-up of 21.8 (range, 2.7-30.7) months, the median progression-free survival (mPFS) was 6.9 (95% confidence interval (CI), 4.7-9.1) months, and the 6-month PFS rate was 60.5%. Based on various histological subtypes, the mPFS was 8.4 (95% CI, 4.1-12.7) months with well-differentiated liposarcoma, 5.8 (95% CI, 3.3-8.3) months with de-differentiated liposarcoma and not reached with MLPS, respectively. Treatment-related adverse events (TRAEs) of any grade occurred in 36 (76.6%) patients, with grade 3 or higher TRAEs in 21 (44.7%) patients. The most common TRAEs were neutropenia (53.2%), proteinuria (21.3%), and anorexia (21.3%). Conclusion The combined treatment strategy involving eribulin, anlotinib, and camrelizumab showed promising efficacy and manageable safety in patients with advanced or metastatic RLPS, particularly in those with MLPS.
Collapse
Affiliation(s)
- Weiwei Jia
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Sarcoma Center, Peking University Cancer Hospital & Institute, Beijing, China
| | - Jianhui Wu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Sarcoma Center, Peking University Cancer Hospital & Institute, Beijing, China
| | - Hongtao Zhang
- Jilin Guowen Hospital, Changchun, Jilin Province, China
| | - Yan Wu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Pathology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Daoning Liu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Sarcoma Center, Peking University Cancer Hospital & Institute, Beijing, China
| | - Zhen Wang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Sarcoma Center, Peking University Cancer Hospital & Institute, Beijing, China
| | - Xiaopeng Wang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Sarcoma Center, Peking University Cancer Hospital & Institute, Beijing, China
| | - Chengpeng Li
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Sarcoma Center, Peking University Cancer Hospital & Institute, 52 Fucheng Road, Haidian District, Beijing 100142, China
| | - Chunyi Hao
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Sarcoma Center, Peking University Cancer Hospital & Institute, 52 Fucheng Road, Haidian District, Beijing 100142, China
| |
Collapse
|
5
|
Bharadwaj NS, Zumwalde NA, Kapur A, Patankar M, Gumperz JE. Human CD4 + memory phenotype T cells use mitochondrial metabolism to generate sensitive IFN-γ responses. iScience 2024; 27:109775. [PMID: 38726371 PMCID: PMC11079467 DOI: 10.1016/j.isci.2024.109775] [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] [Received: 01/10/2024] [Revised: 03/12/2024] [Accepted: 04/15/2024] [Indexed: 05/12/2024] Open
Abstract
The transition of naive T lymphocytes into antigenically activated effector cells is associated with a metabolic shift from oxidative phosphorylation to aerobic glycolysis. This shift facilitates production of the key anti-tumor cytokine interferon (IFN)-γ; however, an associated loss of mitochondrial efficiency in effector T cells ultimately limits anti-tumor immunity. Memory phenotype (MP) T cells are a newly recognized subset that arises through homeostatic activation signals following hematopoietic transplantation. We show here that human CD4+ MP cell differentiation is associated with increased glycolytic and oxidative metabolic activity, but MP cells retain less compromised mitochondria compared to effector CD4+ T cells, and their IFN-γ response is less dependent on glucose and more reliant on glutamine. MP cells also produced IFN-γ more efficiently in response to weak T cell receptor (TCR) agonism than effectors and mediated stronger responses to transformed B cells. MP cells may thus be particularly well suited to carry out sustained immunosurveillance against neoplastic cells.
Collapse
Affiliation(s)
- Nikhila S. Bharadwaj
- Department of Medical Microbiology and Immunology, University of Wisconsin School of Medicine and Public Health; Madison, WI 53706, USA
| | - Nicholas A. Zumwalde
- Department of Genetics, University of Wisconsin School of Medicine and Public Health; Madison, WI 53706, USA
| | - Arvinder Kapur
- QIAGEN Sciences Inc., 19300 Germantown Road, Germantown, MD 20874, USA
| | - Manish Patankar
- Department of Obstetrics and Gynecology, University of Wisconsin School of Medicine and Public Health; Madison, WI 53706, USA
| | - Jenny E. Gumperz
- Department of Medical Microbiology and Immunology, University of Wisconsin School of Medicine and Public Health; Madison, WI 53706, USA
| |
Collapse
|
6
|
Schuettfort G, Röther C, Berger A, Fokas E, Fraunholz I, Groh A, Haberl A, Khaykin P, Martin D, Rödel C, Vehreschild M, Stephan C. Differences in the Course of CD4 and CD8 Cells After Chemoradiotherapy in People Living with HIV with Anal Cancer. AIDS Res Hum Retroviruses 2024; 40:198-203. [PMID: 37756364 DOI: 10.1089/aid.2023.0003] [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] [Indexed: 09/29/2023] Open
Abstract
Incidence of anal carcinoma (AC) in people living with HIV (PLWH) is increased compared to the general population. Adverse effects of chemoradiotherapy (CRT) on the immune system are associated with a significant detrimental prognosis on overall survival in patients receiving CRT for solid tumors. The aim of this study was to evaluate immunological factors, in particular the differences in recovery of CD4+ and CD8+ cell counts before and after CRT for AC in PLWH. Retrospective single-center chart review extraction to analyze immunological data collected from PLWH with AC; descriptive statistics were used. Thirty-six PLWH with histologically proven AC were included in the analysis. Absolute CD4 cell count 60 months after CRT was 67.2% of the value at the beginning of CRT, whereas the CD8 cell count reached 82.3%. These differences were statistically significant (p = .048), whereas CD4/CD8-ratio remained stable. The findings of the presented study regarding CD4+ and CD8+ cell recovery after CRT are congruent with results from prior studies in non-HIV infected patients. Although not reaching the level of prior CRT T cell numbers, the ability to generate CD8+ cells seems to be better recovered, while CD4+ regeneration is more impaired. These observations are best explained by faster recovery of CD8+ cells via thymic-independent pathways, which are not available for regeneration of CD4+ cells. Further studies with larger numbers of patients are required to analyze the specific CD4+ and CD8+ cell subsets.
Collapse
Affiliation(s)
- Gundolf Schuettfort
- Department of Infectious Diseases, University Hospital Frankfurt, Frankfurt, Germany
| | | | - Annemarie Berger
- Department of Virology and University Hospital Frankfurt, Frankfurt, Germany
| | - Emmanouil Fokas
- Department of Radiotherapy, University Hospital Frankfurt, Frankfurt, Germany
| | - Ingeborg Fraunholz
- Department of Infectious Diseases, University Hospital Frankfurt, Frankfurt, Germany
| | - Ana Groh
- Department of Infectious Diseases, University Hospital Frankfurt, Frankfurt, Germany
| | - Annette Haberl
- Department of Infectious Diseases, University Hospital Frankfurt, Frankfurt, Germany
| | | | - Daniel Martin
- Department of Radiotherapy, University Hospital Frankfurt, Frankfurt, Germany
| | - Claus Rödel
- Department of Radiotherapy, University Hospital Frankfurt, Frankfurt, Germany
| | - Maria Vehreschild
- Department of Infectious Diseases, University Hospital Frankfurt, Frankfurt, Germany
| | - Christoph Stephan
- Department of Infectious Diseases, University Hospital Frankfurt, Frankfurt, Germany
| |
Collapse
|
7
|
Dinges SS, Amini K, Notarangelo LD, Delmonte OM. Primary and secondary defects of the thymus. Immunol Rev 2024; 322:178-211. [PMID: 38228406 PMCID: PMC10950553 DOI: 10.1111/imr.13306] [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] [Indexed: 01/18/2024]
Abstract
The thymus is the primary site of T-cell development, enabling generation, and selection of a diverse repertoire of T cells that recognize non-self, whilst remaining tolerant to self- antigens. Severe congenital disorders of thymic development (athymia) can be fatal if left untreated due to infections, and thymic tissue implantation is the only cure. While newborn screening for severe combined immune deficiency has allowed improved detection at birth of congenital athymia, thymic disorders acquired later in life are still underrecognized and assessing the quality of thymic function in such conditions remains a challenge. The thymus is sensitive to injury elicited from a variety of endogenous and exogenous factors, and its self-renewal capacity decreases with age. Secondary and age-related forms of thymic dysfunction may lead to an increased risk of infections, malignancy, and autoimmunity. Promising results have been obtained in preclinical models and clinical trials upon administration of soluble factors promoting thymic regeneration, but to date no therapy is approved for clinical use. In this review we provide a background on thymus development, function, and age-related involution. We discuss disease mechanisms, diagnostic, and therapeutic approaches for primary and secondary thymic defects.
Collapse
Affiliation(s)
- Sarah S. Dinges
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
- Department of Pediatric Respiratory Medicine, Immunology and Critical Care Medicine, Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Kayla Amini
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Luigi D. Notarangelo
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Ottavia M. Delmonte
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
8
|
Heiser RA, Cao AT, Zeng W, Ulrich M, Younan P, Anderson ME, Trueblood ES, Jonas M, Thurman R, Law CL, Gardai SJ. Brentuximab Vedotin-Driven Microtubule Disruption Results in Endoplasmic Reticulum Stress Leading to Immunogenic Cell Death and Antitumor Immunity. Mol Cancer Ther 2024; 23:68-83. [PMID: 37775098 PMCID: PMC10762337 DOI: 10.1158/1535-7163.mct-23-0118] [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: 03/03/2023] [Revised: 08/07/2023] [Accepted: 09/26/2023] [Indexed: 10/01/2023]
Abstract
Brentuximab vedotin, a CD30-directed antibody-drug conjugate (ADC), is approved for clinical use in multiple CD30-expressing lymphomas. The cytotoxic payload component of brentuximab vedotin is monomethyl auristatin E (MMAE), a highly potent microtubule-disrupting agent. Preclinical results provided here demonstrate that treatment of cancer cells with brentuximab vedotin or free MMAE leads to a catastrophic disruption of the microtubule network eliciting a robust endoplasmic reticulum (ER) stress response that culminates in the induction of the classic hallmarks of immunogenic cell death (ICD). In accordance with the induction of ICD, brentuximab vedotin-killed lymphoma cells drove innate immune cell activation in vitro and in vivo. In the "gold-standard" test of ICD, vaccination of mice with brentuximab vedotin or free MMAE-killed tumor cells protected animals from tumor rechallenge; in addition, T cells transferred from previously vaccinated animals slowed tumor growth in immunodeficient mice. Immunity acquired from killed tumor cell vaccination was further amplified by the addition of PD-1 blockade. In a humanized model of CD30+ B-cell tumors, treatment with brentuximab vedotin drove the expansion and recruitment of autologous Epstein-Barr virus-reactive CD8+ T cells potentiating the activity of anti-PD-1 therapy. Together, these data support the ability of brentuximab vedotin and MMAE to drive ICD in tumor cells resulting in the activation of antigen-presenting cells and augmented T-cell immunity. These data provide a strong rationale for the clinical combination of brentuximab vedotin and other MMAE-based ADCs with checkpoint inhibitors.
Collapse
|
9
|
Qian H, Ji R, Shen C, Wei Y, Sheng C, Ni Q, Pan J, Chi Y, You H, Miao Y, Shi M, Huang X, Shen A. ATRX is a predictive marker for endocrinotherapy and chemotherapy resistance in HER2-/HR+ breast cancer through the regulation of the AR, GLI3 and GATA2 transcriptional network. Aging (Albany NY) 2023; 15:14996-15024. [PMID: 38126976 PMCID: PMC10781474 DOI: 10.18632/aging.205327] [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: 11/04/2022] [Accepted: 11/01/2023] [Indexed: 12/23/2023]
Abstract
Drug resistance in breast cancer (BC) is a clinical challenge. Exploring the mechanism and identifying a precise predictive biomarker for the drug resistance in BC is critical. Three first-line drug (paclitaxel, doxorubicin and tamoxifen) resistance datasets in BC from GEO were merged to obtain 1,461 differentially expressed genes for weighted correlation network analysis, resulting in identifying ATRX as the hub gene. ATRX is a chromatin remodelling protein, therefore, ATRX-associated transcription factors were explored, thereby identifying the network of AR, GLI3 and GATA2. GO and KEGG analyses revealed immunity, transcriptional regulation and endocrinotherapy/chemotherapy resistance were enriched. Moreover, CIBERSORT revealed immunity regulation was inhibited in the resistance group. ssGSEA showed a significantly lower immune status in the ATRX-Low group compared to the ATRX-High group. Furthermore, the peaks of H3K9me3 ChIP-seq on the four genes were higher in normal tissues than in BC tissues. Notably, the frequency of ATRX mutation was higher than BRCA in BC. Moreover, depressed ATRX revealed worse overall survival and disease-free survival in the human epidermal growth factor receptor 2 (HER2)-/hormone receptor (HR)+ BC. Additionally, depressed ATRX predicted poor results for patients who underwent endocrinotherapy or chemotherapy in the HER2-/HR+ BC subgroup. A nomogram based on ATRX, TILs and ER exhibited a significantly accurate survival prediction ability. Importantly, overexpression of ATRX significantly inhibited the IC50 of the three first-line drugs on MCF-7 cell. Thus, ATRX is an efficient predictive biomarker for endocrinotherapy and chemotherapy resistance in HER2-/HR+ BC and acts by suppressing the AR, GLI3 and GATA2 transcriptional network.
Collapse
Affiliation(s)
- Hongyan Qian
- Cancer Research Center Nantong, Nantong Tumor Hospital, Affiliated Tumor Hospital of Nantong University, Nantong 226361, China
| | - Rui Ji
- Department of Gynecology Oncology, Nantong Tumor Hospital, Affiliated Tumor Hospital of Nantong University, Nantong 226361, China
| | - Cheng Shen
- Department of Computer Science and Engineering, Tandon School of Engineering, New York University, Brooklyn, NY 11201, USA
| | - Yinze Wei
- Department of Pathology, Affiliated Tumor Hospital of Nantong University, Nantong 226361, China
| | - Chenyi Sheng
- Department of Breast Surgery, Affiliated Hospital of Nantong University, Nantong 226001, China
| | - Qichao Ni
- Department of Breast Surgery, Affiliated Hospital of Nantong University, Nantong 226001, China
| | - Jing Pan
- School of Medicine, Nantong University, Nantong 226001, China
| | - Yifan Chi
- School of Medicine, Nantong University, Nantong 226001, China
| | - Huan You
- School of Medicine, Nantong University, Nantong 226001, China
| | - Ying Miao
- School of Medicine, Nantong University, Nantong 226001, China
| | - Minxin Shi
- Department of Surgery, Affiliated Tumor Hospital of Nantong University, Nantong 226361, China
| | - Xianghua Huang
- Department of Surgery, Affiliated Tumor Hospital of Nantong University, Nantong 226361, China
| | - Aiguo Shen
- Cancer Research Center Nantong, Nantong Tumor Hospital, Affiliated Tumor Hospital of Nantong University, Nantong 226361, China
| |
Collapse
|
10
|
Cheng H, Ji S, Wang J, Hua T, Chen Z, Liu J, Shao L, Wang X, Chen W, Sang W, Qi K, Li Z, Sun C, Shi M, Qiao J, Wu Q, Zeng L, Fei X, Huang H, Gu W, Xu K, Zheng J, Cao J. Long-term analysis of cellular immunity in patients with RRMM treated with CAR-T cell therapy. Clin Exp Med 2023; 23:5241-5254. [PMID: 37907623 DOI: 10.1007/s10238-023-01232-9] [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: 08/11/2023] [Accepted: 10/24/2023] [Indexed: 11/02/2023]
Abstract
Chimeric antigen receptor T (CAR-T) cell therapy exhibits remarkable efficacy against refractory or relapsed multiple myeloma (RRMM); however, the immune deficiency following CAR-Ts infusion has not been well studied. In this study, 126 patients who achieved remission post-CAR-Ts infusion were evaluated for cellular immunity. Following lymphodepletion (LD) chemotherapy, the absolute lymphocyte count (ALC) and absolute counts of lymphocyte subsets were significantly lower than baseline at D0. Grade ≥ 3 lymphopenia occurred in 99% of patients within the first 30 days, with most being resolved by 180 days. The median CD4+ T-cell count was consistently below baseline and the lower limit of normal (LLN) levels at follow-up. Conversely, the median CD8+ T-cell count returned to the baseline and LLN levels by D30. The median B-cell count remained lower than baseline level at D60 and returned to baseline and LLN levels at D180. In the first 30 days, 27 (21.4%) patients had 29 infections, with the majority being mild to moderate in severity (21/29; 72.4%). After day 30, 44 (34.9%) patients had 56 infections, including 20 severe infections. One patient died from bacteremia at 3.8 months post-CAR-Ts infusion. In conclusion, most patients with RRMM experienced cellular immune deficiency caused by LD chemotherapy and CAR-Ts infusion. The ALC and most lymphocyte subsets gradually recovered after day 30 of CAR-Ts infusion, except for CD4+ T cells. Some patients experience prolonged CD4+ T-cell immunosuppression without severe infection.
Collapse
Affiliation(s)
- Hai Cheng
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221002, China
| | - Shengwei Ji
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221002, China
| | - Jiaojiao Wang
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221002, China
| | - Tian Hua
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221002, China
| | - Zihan Chen
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221002, China
| | - Jiaying Liu
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221002, China
| | - Lingyan Shao
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221002, China
| | - Xue Wang
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221002, China
| | - Wei Chen
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221002, China
| | - Wei Sang
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221002, China
| | - Kunming Qi
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221002, China
| | - Zhenyu Li
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221002, China
| | - Cai Sun
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221002, China
| | - Ming Shi
- Cancer Institute, Xuzhou Medical University, Xuzhou, 221002, China
| | - Jianlin Qiao
- Jiangsu Bone Marrow Stem Cell Institute, Xuzhou, 221002, China
| | - Qingyun Wu
- Jiangsu Bone Marrow Stem Cell Institute, Xuzhou, 221002, China
| | - Lingyu Zeng
- Jiangsu Bone Marrow Stem Cell Institute, Xuzhou, 221002, China
| | - Xiaoming Fei
- Department of Hematology, The Affiliated Hospital of Jiangsu University, Zhenjiang, 212000, China
| | - Hongming Huang
- Department of Hematology, The Affiliated Hospital of Nantong University, Nantong, 226000, China
| | - Weiying Gu
- Department of Hematology, The First People's Hospital of Changzhou, The Third Affiliated Hospital of Soochow University, Changzhou, 213000, China
| | - Kailin Xu
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221002, China.
| | - Junnian Zheng
- Cancer Institute, Xuzhou Medical University, Xuzhou, 221002, China.
| | - Jiang Cao
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221002, China.
| |
Collapse
|
11
|
He X, Lan H, Jin K, Liu F. Can immunotherapy reinforce chemotherapy efficacy? a new perspective on colorectal cancer treatment. Front Immunol 2023; 14:1237764. [PMID: 37790928 PMCID: PMC10543914 DOI: 10.3389/fimmu.2023.1237764] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Accepted: 08/25/2023] [Indexed: 10/05/2023] Open
Abstract
As one of the main threats to human life (the fourth most dangerous and prevalent cancer), colorectal cancer affects many people yearly, decreases patients' quality of life, and causes irreparable financial and social damages. In addition, this type of cancer can metastasize and involve the liver in advanced stages. However, current treatments can't completely eradicate this disease. Chemotherapy and subsequent surgery can be mentioned among the current main treatments for this disease. Chemotherapy has many side effects, and regarding the treatment of this type of tumor, chemotherapy can lead to liver damage, such as steatohepatitis, steatosis, and sinus damage. These damages can eventually lead to liver failure and loss of its functions. Therefore, it seems that other treatments can be used in addition to chemotherapy to increase its efficiency and reduce its side effects. Biological therapies and immunotherapy are one of the leading suggestions for combined treatment. Antibodies (immune checkpoint blockers) and cell therapy (DC and CAR-T cells) are among the immune system-based treatments used to treat tumors. Immunotherapy targets various aspects of the tumor that may lead to 1) the recruitment of immune cells, 2) increasing the immunogenicity of tumor cells, and 3) leading to the elimination of inhibitory mechanisms established by the tumor. Therefore, immunotherapy can be used as a complementary treatment along with chemotherapy. This review will discuss different chemotherapy and immunotherapy methods for colorectal cancer. Then we will talk about the studies that have dealt with combined treatment.
Collapse
Affiliation(s)
- Xing He
- Department of Gastroenterology, Jinhua Wenrong Hospital, Jinhua, Zhejiang, China
| | - Huanrong Lan
- Department of Surgical Oncology, Hangzhou Cancer Hospital, Hangzhou, Zhejiang, China
| | - Ketao Jin
- Department of Colorectal Surgery, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, Zhejiang, China
| | - Fanlong Liu
- Department of Colorectal Surgery, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| |
Collapse
|
12
|
Elitzur S, Vora A, Burkhardt B, Inaba H, Attarbaschi A, Baruchel A, Escherich G, Gibson B, Liu HC, Loh M, Moorman AV, Möricke A, Pieters R, Uyttebroeck A, Baird S, Bartram J, Barzilai-Birenboim S, Batra S, Ben-Harosh M, Bertrand Y, Buitenkamp T, Caldwell K, Drut R, Geerlinks AV, Gilad G, Grainger J, Haouy S, Heaney N, Huang M, Ingham D, Krenova Z, Kuhlen M, Lehrnbecher T, Manabe A, Niggli F, Paris C, Revel-Vilk S, Rohrlich P, Sinno MG, Szczepanski T, Tamesberger M, Warrier R, Wolfl M, Nirel R, Izraeli S, Borkhardt A, Schmiegelow K. EBV-driven lymphoid neoplasms associated with pediatric ALL maintenance therapy. Blood 2023; 141:743-755. [PMID: 36332176 DOI: 10.1182/blood.2022016975] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 09/19/2022] [Accepted: 10/08/2022] [Indexed: 11/06/2022] Open
Abstract
The development of a second malignancy after the diagnosis of childhood acute lymphoblastic leukemia (ALL) is a rare event. Certain second malignancies have been linked with specific elements of leukemia therapy, yet the etiology of most second neoplasms remains obscure and their optimal management strategies are unclear. This is a first comprehensive report of non-Hodgkin lymphomas (NHLs) following pediatric ALL therapy, excluding stem-cell transplantation. We analyzed data of patients who developed NHL following ALL diagnosis and were enrolled in 12 collaborative pediatric ALL trials between 1980-2018. Eighty-five patients developed NHL, with mature B-cell lymphoproliferations as the dominant subtype (56 of 85 cases). Forty-six of these 56 cases (82%) occurred during or within 6 months of maintenance therapy. The majority exhibited histopathological characteristics associated with immunodeficiency (65%), predominantly evidence of Epstein-Barr virus-driven lymphoproliferation. We investigated 66 cases of post-ALL immunodeficiency-associated lymphoid neoplasms, 52 from our study and 14 additional cases from a literature search. With a median follow-up of 4.9 years, the 5-year overall survival for the 66 patients with immunodeficiency-associated lymphoid neoplasms was 67.4% (95% confidence interval [CI], 56-81). Five-year cumulative risks of lymphoid neoplasm- and leukemia-related mortality were 20% (95% CI, 10.2-30) and 12.4% (95% CI, 2.7-22), respectively. Concurrent hemophagocytic lymphohistiocytosis was associated with increased mortality (hazard ratio, 7.32; 95% CI, 1.62-32.98; P = .01). A large proportion of post-ALL lymphoid neoplasms are associated with an immunodeficient state, likely precipitated by ALL maintenance therapy. Awareness of this underrecognized entity and pertinent diagnostic tests are crucial for early diagnosis and optimal therapy.
Collapse
Affiliation(s)
- Sarah Elitzur
- Department of Pediatric Hematology and Oncology, Schneider Children's Medical Center and Sackler Faculty of Medicine, Tel-Aviv University, Tel Aviv, Israel
| | - Ajay Vora
- Department of Paediatric Haematology, Great Ormond Street Hospital, London, United Kingdom
| | - Birgit Burkhardt
- Pediatric Hematology and Oncology, University Hospital Münster, Münster, Germany
| | - Hiroto Inaba
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, TN
| | - Andishe Attarbaschi
- Department of Pediatric Hematology and Oncology, St. Anna Children's Hospital, Medical University of Vienna, Vienna, Austria
| | - Andre Baruchel
- Department of Pediatric Hematology, Hôpital Robert Debré, Assistance Publique-Hôpitaux de Paris (AP-HP), Paris, France
| | - Gabriele Escherich
- Department of Pediatric Hematology and Oncoogy, University Medical Centre, Hamburg-Eppendorf, Hamburg, Germany
| | - Brenda Gibson
- Department of Paediatric Haematology, Royal Hospital for Children, Glasgow, United Kingdom
| | - Hsi-Che Liu
- Division of Pediatric Hematology/Oncology, Mackay Children's Hospital and Mackay Medical College, Taipei, Taiwan
| | - Mignon Loh
- Division of Pediatric Hematology, Oncology, Bone Marrow Transplant and Cellular Therapy, Seattle Children's Hospital and the Ben Towne Center for Childhood Cancer Research, University of Washington, Seattle, WA
| | - Anthony V Moorman
- Leukaemia Research Cytogenetics Group, Wolfson Childhood Cancer Centre, Clinical and Translational Research Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Anja Möricke
- Department of Pediatrics, Christian-Albrechts-University Kiel and University Medical Center Schleswig-Holstein, Kiel, Germany
| | - Rob Pieters
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - Anne Uyttebroeck
- Department of Paediatric Haematology and Oncology, University Hospital Leuven, Leuven, Leuven, Belgium
| | - Susan Baird
- Department of Haematology, Royal Hospital for Children and Young People, Edinburgh, United Kingdom
| | - Jack Bartram
- Department of Paediatric Haematology, Great Ormond Street Hospital, London, United Kingdom
| | - Shlomit Barzilai-Birenboim
- Department of Pediatric Hematology and Oncology, Schneider Children's Medical Center and Sackler Faculty of Medicine, Tel-Aviv University, Tel Aviv, Israel
| | - Sandeep Batra
- Pediatric Hematology/Oncology, Riley Hospital for Children, Indiana University School of Medicine, Indianapolis, IN
| | - Miriam Ben-Harosh
- Department of Pediatric Hemato-Oncology, Soroka Medical Center, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Yves Bertrand
- Institut d'Hematologie et d'Oncologie Pediatrique, Hospices Civils de Lyon, Lyon, France
| | - Trudy Buitenkamp
- Amsterdam Academic Medical Center, Emma Children's Hospital, Amsterdam, The Netherlands
| | - Kenneth Caldwell
- Cancer and Blood Disorders Institute, Johns Hopkins All Children's Hospital, St Petersburg, FL
| | - Ricardo Drut
- Department of Pathology, School of Medicine, La Plata National University, La Plata, Argentina
| | | | - Gil Gilad
- Department of Pediatric Hematology and Oncology, Schneider Children's Medical Center and Sackler Faculty of Medicine, Tel-Aviv University, Tel Aviv, Israel
| | - John Grainger
- Faculty of Medical & Human Sciences, University of Manchester and Royal Manchester Children's Hospital, Central Manchester University Hospitals NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, United Kingdom
| | - Stephanie Haouy
- Department of Pediatric Oncology, Centre Hospitalier Universitaire de Montpellier, Montpellier, France
| | - Nicholas Heaney
- Beatson West of Scotland Cancer Centre, Glasgow, United Kingdom
| | - Mary Huang
- Department of Pediatric Hematology Oncology, Massachusetts General Hospital for Children, Harvard Medical School, Boston, MA
| | - Danielle Ingham
- Paediatric Oncology, Leeds Children's Hospital, Leeds, United Kingdom
| | - Zdenka Krenova
- Department of Pediatric Oncology and Department of Pediatrics, University Hospital Brno and Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Michaela Kuhlen
- Pediatrics and Adolescent Medicine, University of Augsburg, Augsburg, Germany
| | - Thomas Lehrnbecher
- Pediatric Hematology and Oncology, Hospital for Children and Adolescents, Johann Wolfgang Goethe University, Frankfurt, Germany
| | - Atsushi Manabe
- Department of Pediatrics, Hokkaido University, Graduate School of Medicine, Sapporo, Japan
| | - Felix Niggli
- Department of Pediatric Oncology, University Children's Hospital, Zurich, Switzerland
| | - Claudia Paris
- Department of Pediatric Oncology and Hematology, Hospital Luis Calvo Mackenna, Santiago, Chile
| | - Shoshana Revel-Vilk
- Shaare Zedek Medical Centre and The Faculty of Medicine, Hebrew University, Jerusalem, Israel
| | | | - Mohamad G Sinno
- Phoenix Children's Hospital, Center for Cancer and Blood Disorders, Phoenix, AZ
| | - Tomasz Szczepanski
- Department of Pediatric Hematology and Oncology, Zabrze and Medical University of Silesia, Katowice, Poland
| | - Melanie Tamesberger
- Department of Pediatrics and Adolescent Medicine, Kepler University Clinic, Linz, Austria
| | | | - Matthias Wolfl
- Pediatric Oncology, Hematology and Stem Cell Transplantation Program, University Children's Hospital Würzburg, Würzburg, Germany
| | - Ronit Nirel
- Department of Statistics and Data Science, Hebrew University, Jerusalem, Israel
| | - Shai Izraeli
- Department of Pediatric Hematology and Oncology, Schneider Children's Medical Center and Sackler Faculty of Medicine, Tel-Aviv University, Tel Aviv, Israel
| | - Arndt Borkhardt
- Department of Paediatric Oncology, Haematology and Clinical Immunology, Medical Faculty, Heinrich-Heine University, Duesseldorf, Germany
| | - Kjeld Schmiegelow
- Department of Pediatrics and Adolescent Medicine, The University Hospital, Rigshospitalet, and Institute of Clinical Medicine, Faculty of Medicine, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
13
|
Hino C, Xu Y, Xiao J, Baylink DJ, Reeves ME, Cao H. The potential role of the thymus in immunotherapies for acute myeloid leukemia. Front Immunol 2023; 14:1102517. [PMID: 36814919 PMCID: PMC9940763 DOI: 10.3389/fimmu.2023.1102517] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2022] [Accepted: 01/20/2023] [Indexed: 02/09/2023] Open
Abstract
Understanding the factors which shape T-lymphocyte immunity is critical for the development and application of future immunotherapeutic strategies in treating hematological malignancies. The thymus, a specialized central lymphoid organ, plays important roles in generating a diverse T lymphocyte repertoire during the infantile and juvenile stages of humans. However, age-associated thymic involution and diseases or treatment associated injury result in a decline in its continuous role in the maintenance of T cell-mediated anti-tumor/virus immunity. Acute myeloid leukemia (AML) is an aggressive hematologic malignancy that mainly affects older adults, and the disease's progression is known to consist of an impaired immune surveillance including a reduction in naïve T cell output, a restriction in T cell receptor repertoire, and an increase in frequencies of regulatory T cells. As one of the most successful immunotherapies thus far developed for malignancy, T-cell-based adoptive cell therapies could be essential for the development of a durable effective treatment to eliminate residue leukemic cells (blasts) and prevent AML relapse. Thus, a detailed cellular and molecular landscape of how the adult thymus functions within the context of the AML microenvironment will provide new insights into both the immune-related pathogenesis and the regeneration of a functional immune system against leukemia in AML patients. Herein, we review the available evidence supporting the potential correlation between thymic dysfunction and T-lymphocyte impairment with the ontogeny of AML (II-VI). We then discuss how the thymus could impact current and future therapeutic approaches in AML (VII). Finally, we review various strategies to rejuvenate thymic function to improve the precision and efficacy of cancer immunotherapy (VIII).
Collapse
Affiliation(s)
- Christopher Hino
- Department of Internal Medicine, Loma Linda University, Loma Linda, CA, United States
| | - Yi Xu
- Division of Hematology and Oncology, Department of Medicine, Loma Linda University, Loma Linda, CA, United States.,Division of Regenerative Medicine, Department of Medicine, Loma Linda University, Loma Linda, CA, United States.,Loma Linda University Cancer Center, Loma Linda, CA, United States
| | - Jeffrey Xiao
- Division of Regenerative Medicine, Department of Medicine, Loma Linda University, Loma Linda, CA, United States
| | - David J Baylink
- Division of Regenerative Medicine, Department of Medicine, Loma Linda University, Loma Linda, CA, United States
| | - Mark E Reeves
- Division of Hematology and Oncology, Department of Medicine, Loma Linda University, Loma Linda, CA, United States.,Loma Linda University Cancer Center, Loma Linda, CA, United States
| | - Huynh Cao
- Division of Hematology and Oncology, Department of Medicine, Loma Linda University, Loma Linda, CA, United States.,Loma Linda University Cancer Center, Loma Linda, CA, United States
| |
Collapse
|
14
|
Tettamanti Boshier FA, Reeves DB, Duke ER, Swan DA, Prlic M, Cardozo-Ojeda EF, Schiffer JT. Substantial uneven proliferation of CD4 + T cells during recovery from acute HIV infection is sufficient to explain the observed expanded clones in the HIV reservoir. J Virus Erad 2022; 8:100091. [PMID: 36582473 PMCID: PMC9792356 DOI: 10.1016/j.jve.2022.100091] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Revised: 09/08/2022] [Accepted: 10/10/2022] [Indexed: 12/03/2022] Open
Abstract
The HIV reservoir is a population of 1-10 million anatomically dispersed, latently infected memory CD4+ T cells in which HIV DNA is quiescently integrated into human chromosomal DNA. When antiretroviral therapy (ART) is stopped and HIV replication initiates in one of these cells, systemic viral spread resumes, rekindling progression to AIDS. Therefore, HIV latency prevents cure. The detection of many populations of identical HIV sequences at unique integration sites implicates CD4+ T cell proliferation as the critical driver of reservoir sustainment after a prolonged period of effective ART. Initial reservoir formation occurs during the first week of primary infection usually before ART is started. While empirical data indicates that both de novo infection and cellular proliferation generate latently infected cells during early untreated infection, it is not known which of these mechanisms is predominant. We developed a mathematical model that recapitulates the profound depletion and brisk recovery of CD4+ T cells, reservoir creation, and viral load trajectory during primary HIV infection. We extended the model to stochastically simulate individual HIV reservoir clones. This model predicts the first detection of HIV infected clones approximately 5 weeks after infection as has recently been shown in vivo and suggests that substantial, uneven proliferation among clones during the recovery from CD4+ lymphopenia is the most plausible explanation for the observed clonal reservoir distribution during the first year of infection.
Collapse
Affiliation(s)
- Florencia A. Tettamanti Boshier
- Vaccine and Infectious Diseases Division, Fred Hutchinson Cancer Research Center, 1100 Fairview Ave., Seattle, WA, 98122, USA
| | - Daniel B. Reeves
- Vaccine and Infectious Diseases Division, Fred Hutchinson Cancer Research Center, 1100 Fairview Ave., Seattle, WA, 98122, USA
| | - Elizabeth R. Duke
- Vaccine and Infectious Diseases Division, Fred Hutchinson Cancer Research Center, 1100 Fairview Ave., Seattle, WA, 98122, USA
- Department of Medicine, University of Washington, 1959 NE Pacific St., Seattle, WA, 98195, USA
| | - David A. Swan
- Vaccine and Infectious Diseases Division, Fred Hutchinson Cancer Research Center, 1100 Fairview Ave., Seattle, WA, 98122, USA
| | - Martin Prlic
- Vaccine and Infectious Diseases Division, Fred Hutchinson Cancer Research Center, 1100 Fairview Ave., Seattle, WA, 98122, USA
- Department of Global Health, University of Washington, 1959 NE Pacific St., Seattle, WA, 98195, USA
| | - E. Fabian Cardozo-Ojeda
- Vaccine and Infectious Diseases Division, Fred Hutchinson Cancer Research Center, 1100 Fairview Ave., Seattle, WA, 98122, USA
| | - Joshua T. Schiffer
- Vaccine and Infectious Diseases Division, Fred Hutchinson Cancer Research Center, 1100 Fairview Ave., Seattle, WA, 98122, USA
- Department of Medicine, University of Washington, 1959 NE Pacific St., Seattle, WA, 98195, USA
- Clinical Research Division, University of Washington, 1959 NE Pacific St., Seattle, WA, USA
| |
Collapse
|
15
|
Krüger T, Wehner R, Herbig M, Kräter M, Kramer M, Middeke JM, Stölzel F, List C, Egger-Heidrich K, Teipel R, Oelschlägel U, Wermke M, Jambor H, Wobus M, Schetelig J, Jöhrens K, Tonn T, Subburayalu J, Schmitz M, Bornhauser M, von Bonin M. Perturbations of mesenchymal stromal cells after allogeneic hematopoietic cell transplantation predispose for bone marrow graft-versus-host-disease. Front Immunol 2022; 13:1005554. [PMID: 36311725 PMCID: PMC9599394 DOI: 10.3389/fimmu.2022.1005554] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Accepted: 09/27/2022] [Indexed: 12/04/2022] Open
Abstract
Functional impairment of the bone marrow (BM) niche has been suggested as a major reason for prolonged cytopenia and secondary graft failure after allogeneic hematopoietic cell transplantation (alloHCT). Because mesenchymal stromal cells (MSCs) serve as multipotent progenitors for several niche components in the BM, they might play a key role in this process. We used collagenase digested trephine biopsies to directly quantify MSCs in 73 patients before (n = 18) and/or after alloHCT (n = 65). For the first time, we demonstrate that acute graft-versus-host disease (aGvHD, n = 39) is associated with a significant decrease in MSC numbers. MSC reduction can be observed even before the clinical onset of aGvHD (n = 10). Assessing MSCs instantly after biopsy collection revealed phenotypic and functional differences depending on the occurrence of aGvHD. These differences vanished during ex vivo expansion. The MSC endotypes observed revealed an enhanced population of donor-derived classical dendritic cells type 1 and alloreactive T cells as the causing agent for compartmental inflammation and MSC damage before clinical onset of aGvHD was ascertained. In conclusion, MSCs endotypes may constitute a predisposing conductor of alloreactivity after alloHCT preceding the clinical diagnosis of aGvHD.
Collapse
Affiliation(s)
- Thomas Krüger
- Department of Internal Medicine I, University Hospital Carl Gustav Carus, Dresden, Germany
- German Cancer Consortium (DKTK), Partner Site Dresden, and German Cancer Research Center (DKFZ), Heidelberg, Germany
- *Correspondence: Thomas Krüger,
| | - Rebekka Wehner
- German Cancer Consortium (DKTK), Partner Site Dresden, and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Institute of Immunology, Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- National Center for Tumor Diseases (NCT), Dresden, Germany
| | - Maik Herbig
- Max Planck Institute for Science of Light and Max-Planck-Zentrum für Physik und Medizin, Erlangen, Germany
- Biotechnology Center, Center for Molecular and Cellular Bioengineering Technical University (TU) Dresden Tatzberg, Dresden, Germany
- Center for Regenerative Therapies (CRTD), Dresden, Germany
| | - Martin Kräter
- Max Planck Institute for Science of Light and Max-Planck-Zentrum für Physik und Medizin, Erlangen, Germany
- Biotechnology Center, Center for Molecular and Cellular Bioengineering Technical University (TU) Dresden Tatzberg, Dresden, Germany
| | - Michael Kramer
- Department of Internal Medicine I, University Hospital Carl Gustav Carus, Dresden, Germany
| | - Jan Moritz Middeke
- Department of Internal Medicine I, University Hospital Carl Gustav Carus, Dresden, Germany
| | - Friedrich Stölzel
- Department of Internal Medicine I, University Hospital Carl Gustav Carus, Dresden, Germany
| | - Catrin List
- Department of Internal Medicine I, University Hospital Carl Gustav Carus, Dresden, Germany
| | | | - Raphael Teipel
- Department of Internal Medicine I, University Hospital Carl Gustav Carus, Dresden, Germany
| | - Uta Oelschlägel
- Department of Internal Medicine I, University Hospital Carl Gustav Carus, Dresden, Germany
| | - Martin Wermke
- Department of Internal Medicine I, University Hospital Carl Gustav Carus, Dresden, Germany
- University Cancer Centrum (UCC), Early Clinical Trial Unit (ECTU), University Hospital Carl Gustav Carus, Dresden, Germany
| | - Helena Jambor
- Department of Internal Medicine I, University Hospital Carl Gustav Carus, Dresden, Germany
| | - Manja Wobus
- Department of Internal Medicine I, University Hospital Carl Gustav Carus, Dresden, Germany
| | - Johannes Schetelig
- Department of Internal Medicine I, University Hospital Carl Gustav Carus, Dresden, Germany
| | - Korinna Jöhrens
- Institute of Pathology, University Hospital Carl Gustav Carus, Dresden, Germany
| | - Torsten Tonn
- Institute of Transfusion Medicine, Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- German Red Cross Blood Donation Service North-East, Dresden, Germany
| | - Julien Subburayalu
- Department of Internal Medicine I, University Hospital Carl Gustav Carus, Dresden, Germany
- German Cancer Consortium (DKTK), Partner Site Dresden, and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Center for Regenerative Therapies (CRTD), Dresden, Germany
- Mildred Scheel Early Career Center, Medical Faculty, Technische Universität Dresden, Dresden, Germany
| | - Marc Schmitz
- German Cancer Consortium (DKTK), Partner Site Dresden, and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Institute of Immunology, Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- National Center for Tumor Diseases (NCT), Dresden, Germany
- Center for Regenerative Therapies (CRTD), Dresden, Germany
| | - Martin Bornhauser
- Department of Internal Medicine I, University Hospital Carl Gustav Carus, Dresden, Germany
- German Cancer Consortium (DKTK), Partner Site Dresden, and German Cancer Research Center (DKFZ), Heidelberg, Germany
- National Center for Tumor Diseases (NCT), Dresden, Germany
- Center for Regenerative Therapies (CRTD), Dresden, Germany
| | - Malte von Bonin
- Department of Internal Medicine I, University Hospital Carl Gustav Carus, Dresden, Germany
- German Cancer Consortium (DKTK), Partner Site Dresden, and German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
16
|
Tian Z, Yao W. PD-1/L1 inhibitor plus chemotherapy in the treatment of sarcomas. Front Immunol 2022; 13:898255. [PMID: 36072581 PMCID: PMC9441887 DOI: 10.3389/fimmu.2022.898255] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Accepted: 08/10/2022] [Indexed: 11/26/2022] Open
Abstract
There is an urgent clinical need for new therapeutic regimens for the effective treatment of advanced sarcomas. Accumulating evidence suggests that programmed death receptor-1/programmed death protein ligand-1 (PD-1/L1) inhibitors have synergistic effects with chemotherapy and have been approved for treatment of lung cancer, gastroesophageal cancer, and breast cancer. In this review, we reviewed the synergistic mechanism of PD-1/L1 inhibitors plus chemotherapy in the treatment of cancers, and the application of this combined regimen in several cancers, followed by a summary of the current evidence on the application of this combined regimen in the treatment of sarcomas as well as the main clinical trials currently underway. Based on the findings of this review, we believe that this combined approach will play an important role in the treatment of some subtypes of sarcomas in the future.
Collapse
|
17
|
Yi L, Yang L. Stem-like T cells and niches: Implications in human health and disease. Front Immunol 2022; 13:907172. [PMID: 36059484 PMCID: PMC9428355 DOI: 10.3389/fimmu.2022.907172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Accepted: 07/28/2022] [Indexed: 11/13/2022] Open
Abstract
Recently, accumulating evidence has elucidated the important role of T cells with stem-like characteristics in long-term maintenance of T cell responses and better patient outcomes after immunotherapy. The fate of TSL cells has been correlated with many physiological and pathological human processes. In this review, we described present advances demonstrating that stem-like T (TSL) cells are central players in human health and disease. We interpreted the evolutionary characteristics, mechanism and functions of TSL cells. Moreover, we discuss the import role of distinct niches and how they affect the stemness of TSL cells. Furthermore, we also outlined currently available strategies to generate TSL cells and associated affecting factors. Moreover, we summarized implication of TSL cells in therapies in two areas: stemness enhancement for vaccines, ICB, and adoptive T cell therapies, and stemness disruption for autoimmune disorders.
Collapse
|
18
|
Saidakova EV. Lymphopenia and Mechanisms of T-Cell Regeneration. CELL AND TISSUE BIOLOGY 2022; 16:302-311. [PMID: 35967247 PMCID: PMC9358362 DOI: 10.1134/s1990519x2204006x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Revised: 01/11/2022] [Accepted: 01/11/2022] [Indexed: 11/24/2022]
Abstract
Chronic lymphopenia, in particular, T-lymphocyte deficiency, increases the risk of death from cancer, cardiovascular and respiratory diseases and serves as a risk factor for a severe course and poor outcome of infectious diseases such as COVID-19. The regeneration of T-lymphocytes is a complex multilevel process, many questions of which still remain unanswered. The present review considers two main pathways of increasing the T-cell number in lymphopenia: production in the thymus and homeostatic proliferation in the periphery. Literature data on the signals that regulate each pathway are summarized. Their contribution to the quantitative and qualitative restoration of the immune cell pool is analyzed. The features of CD4+ and CD8+ T-lymphocytes’ regeneration are considered.
Collapse
Affiliation(s)
- E. V. Saidakova
- Institute of Ecology and Genetics of Microorganisms, Ural Branch, Russian Academy of Sciences—Branch of Perm Federal Research Center, Ural Branch, Russian Academy of Sciences, 614081 Perm, Russia
| |
Collapse
|
19
|
Caballero AC, Escribà-Garcia L, Alvarez-Fernández C, Briones J. CAR T-Cell Therapy Predictive Response Markers in Diffuse Large B-Cell Lymphoma and Therapeutic Options After CART19 Failure. Front Immunol 2022; 13:904497. [PMID: 35874685 PMCID: PMC9299440 DOI: 10.3389/fimmu.2022.904497] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Accepted: 06/07/2022] [Indexed: 12/02/2022] Open
Abstract
Immunotherapy with T cells genetically modified with chimeric antigen receptors (CARs) has shown significant clinical efficacy in patients with relapsed/refractory B-cell lymphoma. Nevertheless, more than 50% of treated patients do not benefit from such therapy due to either absence of response or further relapse. Elucidation of clinical and biological features that would predict clinical response to CART19 therapy is of paramount importance and eventually may allow for selection of those patients with greater chances of response. In the last 5 years, significant clinical experience has been obtained in the treatment of diffuse large B-cell lymphoma (DLBCL) patients with CAR19 T cells, and major advances have been made on the understanding of CART19 efficacy mechanisms. In this review, we discuss clinical and tumor features associated with response to CART19 in DLBCL patients as well as the impact of biological features of the infusion CART19 product on the clinical response. Prognosis of DLBCL patients that fail CART19 is poor and therapeutic approaches with new drugs are also discussed.
Collapse
Affiliation(s)
- Ana Carolina Caballero
- Hematology Service, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
- Laboratory of Experimental Hematology-IIB, Institut Recerca Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
- Campus Sant Pau, Josep Carreras Leukemia Research Institute, Barcelona, Spain
- Autonomous University of Barcelona, Barcelona, Spain
| | - Laura Escribà-Garcia
- Hematology Service, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
- Laboratory of Experimental Hematology-IIB, Institut Recerca Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
- Campus Sant Pau, Josep Carreras Leukemia Research Institute, Barcelona, Spain
| | - Carmen Alvarez-Fernández
- Hematology Service, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
- Laboratory of Experimental Hematology-IIB, Institut Recerca Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
- Campus Sant Pau, Josep Carreras Leukemia Research Institute, Barcelona, Spain
| | - Javier Briones
- Hematology Service, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
- Laboratory of Experimental Hematology-IIB, Institut Recerca Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
- Campus Sant Pau, Josep Carreras Leukemia Research Institute, Barcelona, Spain
- Autonomous University of Barcelona, Barcelona, Spain
| |
Collapse
|
20
|
Homeostatic cytokines tune naivety and stemness of cord blood-derived transgenic T cells. Cancer Gene Ther 2022; 29:961-972. [PMID: 34645974 DOI: 10.1038/s41417-021-00395-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 08/28/2021] [Accepted: 09/28/2021] [Indexed: 12/18/2022]
Abstract
Engineered T-cell therapies have proven to be successful in cancer and their clinical effectiveness is directly correlated with the infused T-cell differentiation profile. Indeed, stem cell memory and central memory T cells proliferate and persist longer in vivo compared with more-differentiated T cells, while conferring enhanced antitumor activity. Here, we propose an optimized process using cord blood (CB) to generate minimally differentiated T-cell products in terms of phenotype, function, gene expression, and metabolism, using peripheral blood (PB)-derived T cells cultured with IL-2 as a standard. Phenotypically, CB-derived T cells, particularly CD4 T cells, are less differentiated than their PB counterparts when cultured with IL-2 or with IL-7 and IL-15. Furthermore, culture with IL-7 and IL-15 enables better preservation of less-differentiated CB-derived T cells compared with IL-2. In addition, transcriptomic and metabolic assessments of CB-derived transgenic T cells cultured with IL-7 and IL-15 point out their naivety and stemness signature. These relatively quiescent transgenic T cells are nevertheless primed for secondary stimulation and cytokine production. In conclusion, our study indicates that CB may be used as a source of early differentiated T cells to develop more effective adoptive cancer immunotherapy.
Collapse
|
21
|
Sottoriva K, Paik NY, White Z, Bandara T, Shao L, Sano T, Pajcini KV. A Notch/IL-21 signaling axis primes bone marrow T cell progenitor expansion. JCI Insight 2022; 7:e157015. [PMID: 35349492 PMCID: PMC9090257 DOI: 10.1172/jci.insight.157015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Accepted: 03/24/2022] [Indexed: 11/17/2022] Open
Abstract
Long-term impairment in T cell-mediated adaptive immunity is a major clinical obstacle following treatment of blood disorders with hematopoietic stem cell transplantation. Although T cell development in the thymus has been extensively characterized, there are significant gaps in our understanding of prethymic processes that influence early T cell potential. We have uncovered a Notch/IL-21 signaling axis in bone marrow common lymphoid progenitor (CLP) cells. IL-21 receptor expression was driven by Notch activation in CLPs, and in vivo treatment with IL-21 induced Notch-dependent CLP proliferation. Taking advantage of this potentially novel signaling axis, we generated T cell progenitors ex vivo, which improved repopulation of the thymus and peripheral lymphoid organs of mice in an allogeneic transplant model. Importantly, Notch and IL-21 activation were equally effective in the priming and expansion of human cord blood cells toward the T cell fate, confirming the translational potential of the combined treatment.
Collapse
Affiliation(s)
| | - Na Yoon Paik
- Department of Pharmacology and Regenerative Medicine and
| | - Zachary White
- Department of Microbiology and Immunology, University of Illinois at Chicago College of Medicine, Chicago, Illinois, USA
| | | | - Lijian Shao
- Department of Pharmacology and Regenerative Medicine and
| | - Teruyuki Sano
- Department of Microbiology and Immunology, University of Illinois at Chicago College of Medicine, Chicago, Illinois, USA
| | | |
Collapse
|
22
|
Tang TCY, Xu N, Nordon R, Haber M, Micklethwaite K, Dolnikov A. Donor T cells for CAR T cell therapy. Biomark Res 2022; 10:14. [PMID: 35365224 PMCID: PMC8973942 DOI: 10.1186/s40364-022-00359-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Accepted: 02/26/2022] [Indexed: 01/01/2023] Open
Abstract
Adoptive cell therapy using patient-derived chimeric receptor antigen (CAR) T cells redirected against tumor cells has shown remarkable success in treating hematologic cancers. However, wider accessibility of cellular therapies for all patients is needed. Manufacture of patient-derived CAR T cells is limited by prolonged lymphopenia in heavily pre-treated patients and risk of contamination with tumor cells when isolating T cells from patient blood rich in malignant blasts. Donor T cells provide a good source of immune cells for adoptive immunotherapy and can be used to generate universal off-the-shelf CAR T cells that are readily available for administration into patients as required. Genome editing tools such as TALENs and CRISPR-Cas9 and non-gene editing methods such as short hairpin RNA and blockade of protein expression are currently used to enhance CAR T cell safety and efficacy by abrogating non-specific toxicity in the form of graft versus host disease (GVHD) and preventing CAR T cell rejection by the host.
Collapse
Affiliation(s)
- Tiffany C Y Tang
- Graduate School of Biomedical Engineering, Faculty of Engineering, UNSW Sydney, Sydney, NSW, Australia. .,Children's Cancer Institute, Lowy Cancer Research Center, UNSW Sydney, Sydney, NSW, Australia.
| | - Ning Xu
- Children's Cancer Institute, Lowy Cancer Research Center, UNSW Sydney, Sydney, NSW, Australia.,School of Women's and Children's Health, Faculty of Medicine, UNSW Sydney, Sydney, NSW, Australia
| | - Robert Nordon
- Graduate School of Biomedical Engineering, Faculty of Engineering, UNSW Sydney, Sydney, NSW, Australia
| | - Michelle Haber
- Children's Cancer Institute, Lowy Cancer Research Center, UNSW Sydney, Sydney, NSW, Australia.,School of Women's and Children's Health, Faculty of Medicine, UNSW Sydney, Sydney, NSW, Australia.,Kids Cancer Center, Sydney Children's Hospital, Sydney, NSW, Australia
| | - Kenneth Micklethwaite
- Blood Transplant and Cell Therapies Program, Department of Hematology, Westmead Hospital, Sydney, NSW, Australia.,Blood Transplant and Cell Therapies Laboratory, NSW Health Pathology, ICPMR Westmead, Sydney, NSW, Australia.,Westmead Institute for Medical Research, Sydney, NSW, Australia.,Sydney Medical School, The University of Sydney, Sydney, NSW, Australia
| | - Alla Dolnikov
- Children's Cancer Institute, Lowy Cancer Research Center, UNSW Sydney, Sydney, NSW, Australia.,School of Women's and Children's Health, Faculty of Medicine, UNSW Sydney, Sydney, NSW, Australia.,Kids Cancer Center, Sydney Children's Hospital, Sydney, NSW, Australia
| |
Collapse
|
23
|
Manouchehri N, Salinas VH, Rabi Yeganeh N, Pitt D, Hussain RZ, Stuve O. Efficacy of Disease Modifying Therapies in Progressive MS and How Immune Senescence May Explain Their Failure. Front Neurol 2022; 13:854390. [PMID: 35432156 PMCID: PMC9009145 DOI: 10.3389/fneur.2022.854390] [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: 01/13/2022] [Accepted: 02/18/2022] [Indexed: 12/11/2022] Open
Abstract
The advent of disease modifying therapies (DMT) in the past two decades has been the cornerstone of successful clinical management of multiple sclerosis (MS). Despite the great strides made in reducing the relapse frequency and occurrence of new signal changes on neuroimaging in patients with relapsing remitting MS (RRMS) by approved DMT, it has been challenging to demonstrate their effectiveness in non-active secondary progressive MS (SPMS) and primary progressive MS (PPMS) disease phenotypes. The dichotomy of DMT effectiveness between RRMS and progressive MS informs on distinct pathogeneses of the different MS phenotypes. Conversely, factors that render patients with progressive MS resistant to therapy are not understood. Thus far, age has emerged as the main correlate of the transition from RRMS to SPMS. Whether it is aging and age-related factors or the underlying immune senescence that qualitatively alter immune responses as the disease transitions to SPMS, that diminish DMT effectiveness, or both, is currently not known. Here, we will discuss the role of immune senescence on different arms of the immune system, and how it may explain relative DMT resistance.
Collapse
Affiliation(s)
- Navid Manouchehri
- Department of Neurology, The University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Victor H. Salinas
- Department of Neurology, The University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Negar Rabi Yeganeh
- Department of Radiopharmacy, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - David Pitt
- Department of Neurology, Yale University, New Haven, CT, United States
| | - Rehana Z. Hussain
- Department of Neurology, The University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Olaf Stuve
- Department of Neurology, The University of Texas Southwestern Medical Center, Dallas, TX, United States
- Neurology Section, VA North Texas Health Care System, Medical Service Dallas, Veterans Affairs Medical Center, Dallas, TX, United States
- *Correspondence: Olaf Stuve
| |
Collapse
|
24
|
Zhang C, Shi Y, Wu L, Wang C, Liao N, Wang F, Zhao B, Wang Y, Liu X. Far-Red Light Triggered Production of Bispecific T Cell Engagers (BiTEs) from Engineered Cells for Antitumor Application. ACS Synth Biol 2022; 11:888-899. [PMID: 35113526 DOI: 10.1021/acssynbio.1c00523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Bispecific T-cell engagers (BiTEs), which have shown potent antitumor activity in humans, are emerging as one of the most promising immunotherapeutic strategies for cancer treatment in recent years. However, the clinical application of BiTEs nowadays has been hampered by their short half-life in the circulatory system due to their low molecular weight and rapid renal clearance. Inevitable continuous infusion of BiTEs has become a routine operation in order to achieve effective treatment, although it is costly, inconvenient, time-consuming, and even painful for patients in some cases. To develop an on-demand, tunable, and reversible approach to overcome these limitations, we assembled a transcription-control device into mammalian cells based on a bacterial far-red light (FRL) responsive signaling pathway to drive the expression of a BiTE against Glypican 3 (GPC3), which is a highly tumor-specific antigen expressed in most hepatocellular carcinomas (HCC). As demonstrated in in vitro experiments, we proved that the FRL sensitive device spatiotemporally responded to the control of FRL illumination and produced a therapeutic level of BiTEs that recruited and activated human T cells to eliminate GPC3 positive tumor cells. By functionally harnessing the power of optogenetics to remotely regulate the production of BiTEs from bioengineered cells and demonstrating its effectiveness in treating tumor cells, this study provides a novel approach to achieve an in vivo supply of BiTEs, which could be potentially applied to other formats of bispecific antibodies and facilitate their clinical applications.
Collapse
Affiliation(s)
- Cuilin Zhang
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou 350025, PR China
- The Liver Center of Fujian Province, Fujian Medical University, Fuzhou 350025, PR China
| | - Yingjun Shi
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou 350025, PR China
- The Liver Center of Fujian Province, Fujian Medical University, Fuzhou 350025, PR China
| | - Lingjie Wu
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou 350025, PR China
- The Liver Center of Fujian Province, Fujian Medical University, Fuzhou 350025, PR China
| | - Chenyi Wang
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou 350025, PR China
- The Liver Center of Fujian Province, Fujian Medical University, Fuzhou 350025, PR China
| | - Naishun Liao
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou 350025, PR China
- The Liver Center of Fujian Province, Fujian Medical University, Fuzhou 350025, PR China
| | - Fei Wang
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou 350025, PR China
- The Liver Center of Fujian Province, Fujian Medical University, Fuzhou 350025, PR China
| | - Bixing Zhao
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou 350025, PR China
- The Liver Center of Fujian Province, Fujian Medical University, Fuzhou 350025, PR China
| | - Yingchao Wang
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou 350025, PR China
- The Liver Center of Fujian Province, Fujian Medical University, Fuzhou 350025, PR China
| | - Xiaolong Liu
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou 350025, PR China
- The Liver Center of Fujian Province, Fujian Medical University, Fuzhou 350025, PR China
| |
Collapse
|
25
|
Long YX, Sun Y, Liu RZ, Zhang MY, Zhao J, Wang YQ, Zhou YW, Cheng K, Chen Y, Zhu CR, Liu JY. Immune-Related Pneumonitis Was Decreased by Addition of Chemotherapy with PD-1/L1 Inhibitors: Systematic Review and Network Meta-Analysis of Randomized Controlled Trials (RCTs). Curr Oncol 2022; 29:267-282. [PMID: 35049699 PMCID: PMC8774972 DOI: 10.3390/curroncol29010025] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Revised: 12/09/2021] [Accepted: 12/21/2021] [Indexed: 12/11/2022] Open
Abstract
Purpose: Immune-related pneumonitis (IRP) has attracted extensive attention, owing to its increased mortality rate. Conventional chemotherapy (C) has been considered as an immunosuppressive agent and may thus reduce IRP’s risk when used in combination with PD-1/L1 inhibitors. This study aimed to assess the risk of IRP with PD-1/L1 inhibitors plus chemotherapy (I+C) versus PD-1/L1 inhibitors alone (I) in solid cancer treatment. Method: Multiple databases were searched for RCTs before January 2021. This NMA was performed among I+C, I, and C to investigate IRP’s risk. Subgroup analysis was carried out on the basis of different PD-1/L1 inhibitors and cancer types. Results: Thirty-one RCTs (19,624 patients) were included. The I+C group exhibited a lower risk of IRP in any grade (RR, 0.60; 95% CI, 0.38–0.95) and in grade 3–5 (RR, 0.44; 95% CI, 0.21–0.92) as opposed to the I group. The risk of any grade IRP with PD-1 plus chemotherapy was lower than that with PD-1 monotherapy (RR, 0.50; 95% CI, 0.28–0.89), although grade 3–5 IRP was similar. There was no statistically meaningful difference in the risk of any grade IRP between PD-L1 plus chemotherapy and PD-L1 inhibitors monotherapy (RR, 0.95; 95% CI, 0.43–2.09) or grade 3–5 IRP (RR, 0.71;95% CI, 0.24–2.07). In addition, compared with the I group, the I+C group was correlated with a decreased risk in IRP regardless of cancer type, while a substantial difference was only observed in NSCLC patients for grade 3–5 IRP (RR, 0.39; 95% CI, 0.15–0.98). Conclusion: In comparison to PD-1/L1 inhibitor treatment alone, combining chemotherapy with PD-1/L1 inhibitors might reduce the risk of IRP in the general population. Furthermore, PD-1 inhibitors in combination with chemotherapy were correlated with a decreased risk of IRP compared to PD-1 inhibitor treatment alone. In contrast to the I group, the I+C group exhibited a lower risk of IRP, especially for NSCLC patients.
Collapse
Affiliation(s)
- Yi-Xiu Long
- Department of Biotherapy, Cancer Center, West China Hospital of Sichuan University, Chengdu 610041, China; (Y.-X.L.); (M.-Y.Z.); (J.Z.); (Y.-Q.W.); (Y.-W.Z.); (K.C.); (Y.C.)
| | - Yue Sun
- Department of Epidemiology and Health Statistics, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu 610041, China; (Y.S.); (C.-R.Z.)
| | - Rui-Zhi Liu
- Department of Medicine and Life Science, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China;
| | - Ming-Yi Zhang
- Department of Biotherapy, Cancer Center, West China Hospital of Sichuan University, Chengdu 610041, China; (Y.-X.L.); (M.-Y.Z.); (J.Z.); (Y.-Q.W.); (Y.-W.Z.); (K.C.); (Y.C.)
| | - Jing Zhao
- Department of Biotherapy, Cancer Center, West China Hospital of Sichuan University, Chengdu 610041, China; (Y.-X.L.); (M.-Y.Z.); (J.Z.); (Y.-Q.W.); (Y.-W.Z.); (K.C.); (Y.C.)
| | - Yu-Qing Wang
- Department of Biotherapy, Cancer Center, West China Hospital of Sichuan University, Chengdu 610041, China; (Y.-X.L.); (M.-Y.Z.); (J.Z.); (Y.-Q.W.); (Y.-W.Z.); (K.C.); (Y.C.)
| | - Yu-Wen Zhou
- Department of Biotherapy, Cancer Center, West China Hospital of Sichuan University, Chengdu 610041, China; (Y.-X.L.); (M.-Y.Z.); (J.Z.); (Y.-Q.W.); (Y.-W.Z.); (K.C.); (Y.C.)
| | - Ke Cheng
- Department of Biotherapy, Cancer Center, West China Hospital of Sichuan University, Chengdu 610041, China; (Y.-X.L.); (M.-Y.Z.); (J.Z.); (Y.-Q.W.); (Y.-W.Z.); (K.C.); (Y.C.)
| | - Ye Chen
- Department of Biotherapy, Cancer Center, West China Hospital of Sichuan University, Chengdu 610041, China; (Y.-X.L.); (M.-Y.Z.); (J.Z.); (Y.-Q.W.); (Y.-W.Z.); (K.C.); (Y.C.)
| | - Cai-Rong Zhu
- Department of Epidemiology and Health Statistics, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu 610041, China; (Y.S.); (C.-R.Z.)
| | - Ji-Yan Liu
- Department of Biotherapy, Cancer Center, West China Hospital of Sichuan University, Chengdu 610041, China; (Y.-X.L.); (M.-Y.Z.); (J.Z.); (Y.-Q.W.); (Y.-W.Z.); (K.C.); (Y.C.)
- Sichuan Clinical Research Center of Biotherapy, Chengdu 610041, China
- Department of Oncology, The First People’s Hospital of Ziyang, Ziyang 641300, China
- Correspondence: ; Tel.: +86-28-542-3261; Fax: +86-28-8542-3609
| |
Collapse
|
26
|
Alonso-Álvarez S, Colado E, Moro-García MA, Alonso-Arias R. Cytomegalovirus in Haematological Tumours. Front Immunol 2021; 12:703256. [PMID: 34733270 PMCID: PMC8558552 DOI: 10.3389/fimmu.2021.703256] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Accepted: 09/14/2021] [Indexed: 12/11/2022] Open
Abstract
The exquisite coupling between herpesvirus and human beings is the result of millions of years of relationship, coexistence, adaptation, and divergence. It is probably based on the ability to generate a latency that keeps viral activity at a very low level, thereby apparently minimising harm to its host. However, this evolutionary success disappears in immunosuppressed patients, especially in haematological patients. The relevance of infection and reactivation in haematological patients has been a matter of interest, although one fundamentally focused on reactivation in the post-allogeneic stem cell transplant (SCT) patient cohort. Newer transplant modalities have been progressively introduced in clinical settings, with successively more drugs being used to manipulate graft composition and functionality. In addition, new antiviral drugs are available to treat CMV infection. We review the immunological architecture that is key to a favourable outcome in this subset of patients. Less is known about the effects of herpesvirus in terms of mortality or disease progression in patients with other malignant haematological diseases who are treated with immuno-chemotherapy or new molecules, or in patients who receive autologous SCT. The absence of serious consequences in these groups has probably limited the motivation to deepen our knowledge of this aspect. However, the introduction of new therapeutic agents for haematological malignancies has led to a better understanding of how natural killer (NK) cells, CD4+ and CD8+ T lymphocytes, and B lymphocytes interact, and of the role of CMV infection in the context of recently introduced drugs such as Bruton tyrosine kinase (BTK) inhibitors, phosphoinosytol-3-kinase inhibitors, anti-BCL2 drugs, and even CAR-T cells. We analyse the immunological basis and recommendations regarding these scenarios.
Collapse
Affiliation(s)
- Sara Alonso-Álvarez
- Haematology and Haemotherapy Department, Hospital Universitario Central de Asturias, Oviedo, Spain.,Laboratory Medicine Department, Hospital Universitario Central de Asturias, Oviedo, Spain.,Department of Hematologic Malignancies, Health Research Institute of the Principality of Asturias (ISPA), Oviedo, Spain
| | - Enrique Colado
- Haematology and Haemotherapy Department, Hospital Universitario Central de Asturias, Oviedo, Spain.,Laboratory Medicine Department, Hospital Universitario Central de Asturias, Oviedo, Spain.,Department of Hematologic Malignancies, Health Research Institute of the Principality of Asturias (ISPA), Oviedo, Spain
| | - Marco A Moro-García
- Laboratory Medicine Department, Hospital Universitario Central de Asturias, Oviedo, Spain.,Department of Cardiac Pathology, Health Research Institute of the Principality of Asturias (ISPA), Oviedo, Spain
| | - Rebeca Alonso-Arias
- Department of Cardiac Pathology, Health Research Institute of the Principality of Asturias (ISPA), Oviedo, Spain.,Immunology Department, Hospital Universitario Central de Asturias, Oviedo, Spain
| |
Collapse
|
27
|
Macintyre AN, French MJ, Sanders BR, Riebe KJ, Shterev ID, Wiehe K, Hora B, Evangelous T, Dugan G, Bourland JD, Cline JM, Sempowski GD. Long-Term Recovery of the Adaptive Immune System in Rhesus Macaques After Total Body Irradiation. Adv Radiat Oncol 2021; 6:100677. [PMID: 34646962 PMCID: PMC8498734 DOI: 10.1016/j.adro.2021.100677] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 12/14/2020] [Accepted: 01/30/2021] [Indexed: 12/15/2022] Open
Abstract
PURPOSE Ionizing radiation causes acute damage to hematopoietic and immune cells, but the long-term immunologic consequences of irradiation are poorly understood. We therefore performed a prospective study of the delayed immune effects of radiation using a rhesus macaque model. METHODS AND MATERIALS Ten macaques received 4 Gy high-energy x-ray total body irradiation (TBI) and 6 control animals received sham irradiation. TBI caused transient lymphopenia that resolved over several weeks. Once white blood cell counts recovered, flow cytometry was used to immunophenotype the circulating adaptive immune cell populations 4, 9, and 21 months after TBI. Data were fit using a mixed-effects model to determine age-dependent, radiation-dependent, and interacting effects. T cell receptor (TCR) sequencing and quantification of TCR Excision Circles were used to determine relative contributions of thymopoiesis and peripheral expansion to T cell repopulation. Two years after TBI, the cohort was vaccinated with a 23-valent pneumococcal polysaccharide vaccine and a tetravalent influenza hemagglutinin vaccine. RESULTS Aging, but not TBI, led to significant changes in the frequencies of dendritic cells, CD4 and CD8 T cells, and B cells. However, irradiated animals exhibited increased frequencies of central memory T cells and decreased frequencies of naïve T cells. These consequences of irradiation were time-dependent and more prolonged in the CD8 T cell population. Irradiation led to transient increases in CD8+ T cell TCR Excision Circles and had no significant effect on TCR sequence entropy, indicating T cell recovery was partially mediated by thymopoiesis. Animals that were irradiated and then vaccinated showed normal immunoglobulin G binding and influenza neutralization titers in response to the 4 protein antigens but weaker immunoglobulin G binding titers to 10 of the 23 polysaccharide antigens. CONCLUSIONS These findings indicate that TBI causes subtle but long-lasting immune defects that are evident years after recovery from lymphopenia.
Collapse
Affiliation(s)
- Andrew N. Macintyre
- Duke Human Vaccine Institute and Department of Medicine, Duke University School of Medicine, Durham, North Carolina
| | - Matthew J. French
- Duke Human Vaccine Institute and Department of Medicine, Duke University School of Medicine, Durham, North Carolina
| | - Brittany R. Sanders
- Duke Human Vaccine Institute and Department of Medicine, Duke University School of Medicine, Durham, North Carolina
| | - Kristina J. Riebe
- Duke Human Vaccine Institute and Department of Medicine, Duke University School of Medicine, Durham, North Carolina
| | - Ivo D. Shterev
- Duke Human Vaccine Institute and Department of Medicine, Duke University School of Medicine, Durham, North Carolina
| | - Kevin Wiehe
- Duke Human Vaccine Institute and Department of Medicine, Duke University School of Medicine, Durham, North Carolina
| | - Bhavna Hora
- Duke Human Vaccine Institute and Department of Medicine, Duke University School of Medicine, Durham, North Carolina
| | - Tyler Evangelous
- Duke Human Vaccine Institute and Department of Medicine, Duke University School of Medicine, Durham, North Carolina
| | - Greg Dugan
- Department of Pathology/Comparative Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina
| | - J. Daniel Bourland
- Department of Radiation Oncology, Wake Forest University School of Medicine, Winston-Salem, North Carolina
| | - J. Mark Cline
- Department of Pathology/Comparative Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina
| | - Gregory D. Sempowski
- Duke Human Vaccine Institute and Department of Medicine, Duke University School of Medicine, Durham, North Carolina
| |
Collapse
|
28
|
Lingering effects of chemotherapy on mature T cells impair proliferation. Blood Adv 2021; 4:4653-4664. [PMID: 33002133 DOI: 10.1182/bloodadvances.2020001797] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Accepted: 08/30/2020] [Indexed: 12/11/2022] Open
Abstract
Engineered T-cell therapies have demonstrated impressive clinical responses in patients with hematologic malignancies. Despite this efficacy, many patients have a transient persistence of T cells, which can be correlated with transient clinical response. Translational data on T cells from pediatric cancer patients shows a progressive decline in chimeric antigen receptor (CAR) suitability with cumulative chemotherapy regardless of regimen. We investigated the effects of chemotherapy on surviving T cells in vitro, describing residual deficits unique to each agent including mitochondrial damage and metabolic alterations. In the case of cyclophosphamide but not doxorubicin or cytarabine, these effects could be reversed with N-acetylcysteine. Specifically, we observed that surviving T cells could be stimulated, expanded, and transduced with CARs with preserved short-term cytolytic function but at far lower numbers and with residual metabolic deficits. These data have implications for understanding the effects of chemotherapy on mature T cells later collected for adoptive cell therapy, as chemotherapy-exposed T cells may have lingering dysfunction that affects ex vivo adoptive cell therapy manufacturing techniques and, ultimately, clinical efficacy.
Collapse
|
29
|
Immune reconstitution and infectious complications following axicabtagene ciloleucel therapy for large B-cell lymphoma. Blood Adv 2021; 5:143-155. [PMID: 33570626 DOI: 10.1182/bloodadvances.2020002732] [Citation(s) in RCA: 120] [Impact Index Per Article: 40.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Accepted: 10/30/2020] [Indexed: 12/12/2022] Open
Abstract
Chimeric antigen receptor (CAR) T-cell therapy targeting CD19 has significantly improved outcomes in the treatment of refractory or relapsed large B-cell lymphoma (LBCL). We evaluated the long-term course of hematologic recovery, immune reconstitution, and infectious complications in 41 patients with LBCL treated with axicabtagene ciloleucel (axi-cel) at a single center. Grade 3+ cytopenias occurred in 97.6% of patients within the first 28 days postinfusion, with most resolved by 6 months. Overall, 63.4% of patients received a red blood cell transfusion, 34.1% of patients received a platelet transfusion, 36.6% of patients received IV immunoglobulin, and 51.2% of patients received growth factor (granulocyte colony-stimulating factor) injections beyond the first 28 days postinfusion. Only 40% of patients had recovered detectable CD19+ B cells by 1 year, and 50% of patients had a CD4+ T-cell count <200 cells per μL by 18 months postinfusion. Patients with durable responses to axi-cel had significantly longer durations of B-cell aplasia, and this duration correlated strongly with the recovery of CD4+ T-cell counts. There were significantly more infections within the first 28 days compared with any other period of follow-up, with the majority being mild-moderate in severity. Receipt of corticosteroids was the only factor that predicted risk of infection in a multivariate analysis (hazard ratio, 3.69; 95% confidence interval, 1.18-16.5). Opportunistic infections due to Pneumocystis jirovecii and varicella-zoster virus occurred up to 18 months postinfusion in patients who prematurely discontinued prophylaxis. These results support the use of comprehensive supportive care, including long-term monitoring and antimicrobial prophylaxis, beyond 12 months after axi-cel treatment.
Collapse
|
30
|
Effects of Chemotherapy Agents on Circulating Leukocyte Populations: Potential Implications for the Success of CAR-T Cell Therapies. Cancers (Basel) 2021; 13:cancers13092225. [PMID: 34066414 PMCID: PMC8124952 DOI: 10.3390/cancers13092225] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 04/25/2021] [Accepted: 05/04/2021] [Indexed: 12/15/2022] Open
Abstract
Simple Summary CAR-T cell therapy is a new approach to cancer treatment that is based on manipulating a patient’s own T cells such that they become able to seek and destroy cancer cells in a highly specific manner. This approach is showing remarkable efficacy in treating some types of blood cancers but so far has been much less effective against solid cancers. Here, we review the diverse effects of chemotherapy agents on circulating leukocyte populations and find that, despite some negative effects over the short term, chemotherapy can favourably modulate the immune systems of cancer patients over the longer term. Since blood is the starting material for CAR-T cell production, we propose that these effects could significantly influence the success of manufacturing, and anti-cancer activity, of CAR-T cells. Thus, if timed correctly, chemotherapy-induced changes to circulating immune cells could allow CAR-T cells to unleash more effective anti-tumour responses. Abstract Adoptive T-cell therapy using autologous T cells genetically modified to express cancer-specific chimeric antigen receptors (CAR) has emerged as a novel approach for cancer treatment. CAR-T cell therapy has been approved in several major jurisdictions for treating refractory or relapsed cases of B-cell precursor acute lymphoblastic leukaemia and diffuse large B-cell lymphoma. However, in solid cancer patients, several clinical studies of CAR-T cell therapy have demonstrated minimal therapeutic effects, thus encouraging interest in better integrating CAR-T cells with other treatments such as conventional cytotoxic chemotherapy. Increasing evidence shows that not only do chemotherapy drugs have tumoricidal effects, but also significantly modulate the immune system. Here, we discuss immunomodulatory effects of chemotherapy drugs on circulating leukocyte populations, including their ability to enhance cytotoxic effects and preserve the frequency of CD8+ T cells and to deplete immunosuppressive populations including regulatory T cells and myeloid-derived suppressor cells. By modulating the abundance and phenotype of leukocytes in the blood (the ‘raw material’ for CAR-T cell manufacturing), we propose that prior chemotherapy could facilitate production of the most effective CAR-T cell products. Further research is required to directly test this concept and identify strategies for the optimal integration of CAR-T cell therapies with cytotoxic chemotherapy for solid cancers.
Collapse
|
31
|
Panoskaltsis N, McCarthy NE, Stagg AJ, Mummery CJ, Husni M, Arebi N, Greenstein D, Price CL, Al-Hassi HO, Koutinas M, Mantalaris A, Knight SC. Immune reconstitution and clinical recovery following anti-CD28 antibody (TGN1412)-induced cytokine storm. Cancer Immunol Immunother 2021; 70:1127-1142. [PMID: 33033851 PMCID: PMC7543968 DOI: 10.1007/s00262-020-02725-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Accepted: 09/11/2020] [Indexed: 12/21/2022]
Abstract
Cytokine storm can result from cancer immunotherapy or certain infections, including COVID-19. Though short-term immune-related adverse events are routinely described, longer-term immune consequences and sequential immune monitoring are not as well defined. In 2006, six healthy volunteers received TGN1412, a CD28 superagonist antibody, in a first-in-man clinical trial and suffered from cytokine storm. After the initial cytokine release, antibody effect-specific immune monitoring started on Day + 10 and consisted mainly of evaluation of dendritic cell and T-cell subsets and 15 serum cytokines at 21 time-points over 2 years. All patients developed problems with concentration and memory; three patients were diagnosed with mild-to-moderate depression. Mild neutropenia and autoantibody production was observed intermittently. One patient suffered from peripheral dry gangrene, required amputations, and had persistent Raynaud's phenomenon. Gastrointestinal irritability was noted in three patients and coincided with elevated γδT-cells. One had pruritus associated with elevated IgE levels, also found in three other asymptomatic patients. Dendritic cells, initially undetectable, rose to normal within a month. Naïve CD8+ T-cells were maintained at high levels, whereas naïve CD4+ and memory CD4+ and CD8+ T-cells started high but declined over 2 years. T-regulatory cells cycled circannually and were normal in number. Cytokine dysregulation was especially noted in one patient with systemic symptoms. Over a 2-year follow-up, cognitive deficits were observed in all patients following TGN1412 infusion. Some also had signs or symptoms of psychological, mucosal or immune dysregulation. These observations may discern immunopathology, treatment targets, and long-term monitoring strategies for other patients undergoing immunotherapy or with cytokine storm.
Collapse
Affiliation(s)
- Nicki Panoskaltsis
- Department of Haematology, Imperial College London, Northwick Park & St. Mark's Campus, London, UK.
- Antigen Presentation Research Group, Imperial College London, Northwick Park & St. Mark's Campus, London, UK.
- Biological Systems Engineering Laboratory, Centre for Process Systems Engineering, Department of Chemical Engineering, Imperial College London, London, UK.
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University School of Medicine, Atlanta, USA.
- BioMedical Systems Engineering Laboratory, Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, USA.
| | - Neil E McCarthy
- Antigen Presentation Research Group, Imperial College London, Northwick Park & St. Mark's Campus, London, UK
- Centre for Immunobiology, The Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Andrew J Stagg
- Antigen Presentation Research Group, Imperial College London, Northwick Park & St. Mark's Campus, London, UK
- Centre for Immunobiology, The Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Catherine J Mummery
- Dementia Research Centre, National Hospital for Neurology and Neurosurgery, Queen Square, Department of Neurology, Northwick Park Hospital, London, UK
- National Hospital for Neurology and Neurosurgery, University College London Hospital, University College London, London, UK
| | - Mariwan Husni
- Central and North West London Mental Health NHS Foundation Trust, Northwick Park Hospital, London, UK
- Psychiatry Department, Arabian Gulf University, Manama, Kingdom of Bahrain
| | - Naila Arebi
- Department of Gastroenterology and Intestinal Physiology, St. Mark's Hospital, London, UK
- Inflammatory Bowel Disease Clinical Service, St Mark's Hospital, London, UK
| | - David Greenstein
- Department of Vascular Surgery, North West London Hospitals NHS Trust, Northwick Park & St. Mark's Hospitals Site, London, UK
- Department of Vascular Surgery, Northwick Park Hospital and Imperial College London, London, UK
| | - Claire L Price
- Antigen Presentation Research Group, Imperial College London, Northwick Park & St. Mark's Campus, London, UK
- Lucid Group Communications, Buckinghamshire, UK
| | - Hafid O Al-Hassi
- Antigen Presentation Research Group, Imperial College London, Northwick Park & St. Mark's Campus, London, UK
- Research Institute in Healthcare Science, Faculty of Science and Engineering, University of Wolverhampton, Wolverhampton, UK
| | - Michalis Koutinas
- Biological Systems Engineering Laboratory, Centre for Process Systems Engineering, Department of Chemical Engineering, Imperial College London, London, UK
- Department of Chemical Engineering, Cyprus University of Technology, Limassol, Cyprus
| | - Athanasios Mantalaris
- Biological Systems Engineering Laboratory, Centre for Process Systems Engineering, Department of Chemical Engineering, Imperial College London, London, UK
- BioMedical Systems Engineering Laboratory, Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, USA
| | - Stella C Knight
- Antigen Presentation Research Group, Imperial College London, Northwick Park & St. Mark's Campus, London, UK
| |
Collapse
|
32
|
Lo Presti V, Cornel AM, Plantinga M, Dünnebach E, Kuball J, Boelens JJ, Nierkens S, van Til NP. Efficient lentiviral transduction method to gene modify cord blood CD8 + T cells for cancer therapy applications. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2021; 21:357-368. [PMID: 33898633 PMCID: PMC8056177 DOI: 10.1016/j.omtm.2021.03.015] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Accepted: 03/17/2021] [Indexed: 01/01/2023]
Abstract
Adoptive T cell therapy utilizing tumor-specific autologous T cells has shown promising results for cancer treatment. However, the limited numbers of autologous tumor-associated antigen (TAA)-specific T cells and the functional aberrancies, due to disease progression or treatment, remain factors that may significantly limit the success of the therapy. The use of allogeneic T cells, such as umbilical cord blood (CB) derived, overcomes these issues but requires gene modification to induce a robust and specific anti-tumor effect. CB T cells are readily available in CB banks and show low toxicity, high proliferation rates, and increased anti-leukemic effect upon transfer. However, the combination of anti-tumor gene modification and preservation of advantageous immunological traits of CB T cells represent major challenges for the harmonized production of T cell therapy products. In this manuscript, we optimized a protocol for expansion and lentiviral vector (LV) transduction of CB CD8+ T cells, achieving a transduction efficiency up to 83%. Timing of LV treatment, selection of culture media, and the use of different promoters were optimized in the transduction protocol. LentiBOOST was confirmed as a non-toxic transduction enhancer of CB CD8+ T cells, with minor effects on the proliferation capacity and cell viability of the T cells. Positively, the use of LentiBOOST does not affect the functionality of the cells, in the context of tumor cell recognition. Finally, CB CD8+ T cells were more amenable to LV transduction than peripheral blood (PB) CD8+ T cells and maintained a more naive phenotype. In conclusion, we show an efficient method to genetically modify CB CD8+ T cells using LV, which is especially useful for off-the-shelf adoptive cell therapy products for cancer treatment.
Collapse
Affiliation(s)
- Vania Lo Presti
- Center for Translational Immunology, UMC Utrecht, Utrecht, the Netherlands.,Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
| | - Annelisa M Cornel
- Center for Translational Immunology, UMC Utrecht, Utrecht, the Netherlands.,Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
| | - Maud Plantinga
- Center for Translational Immunology, UMC Utrecht, Utrecht, the Netherlands.,Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
| | - Ester Dünnebach
- Center for Translational Immunology, UMC Utrecht, Utrecht, the Netherlands.,Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
| | - Jurgen Kuball
- Center for Translational Immunology, UMC Utrecht, Utrecht, the Netherlands.,Department of Hematology, UMC Utrecht, Utrecht, the Netherlands
| | - Jaap Jan Boelens
- Stem Cell Transplant and Cellular Therapies, MSK Kids, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Stefan Nierkens
- Center for Translational Immunology, UMC Utrecht, Utrecht, the Netherlands.,Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
| | - Niek P van Til
- Center for Translational Immunology, UMC Utrecht, Utrecht, the Netherlands.,AVROBIO, Inc., Cambridge, MA, USA.,Child Neurology, Emma Children's Hospital, Amsterdam University Medical Centers, Vrije Universiteit and Amsterdam Neuroscience, Amsterdam, the Netherlands
| |
Collapse
|
33
|
Saghafian-Hedengren S, Sverremark-Ekström E, Nilsson A. T Cell Subsets During Early Life and Their Implication in the Treatment of Childhood Acute Lymphoblastic Leukemia. Front Immunol 2021; 12:582539. [PMID: 33763058 PMCID: PMC7982872 DOI: 10.3389/fimmu.2021.582539] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2020] [Accepted: 02/01/2021] [Indexed: 11/13/2022] Open
Abstract
The immune system plays a major role in recognizing and eliminating malignant cells, and this has been exploited in the development of immunotherapies aimed at either activating or reactivating the anti-tumor activity of a patient's immune system. A wide range of therapeutic approaches involving T lymphocytes, such as programmed cell death protein ligand-1 (PDL-1) inhibitors, cytotoxic T-lymphocyte-associated protein-4 (CTLA-4) blockers, and CD19-targeted T-cell therapy through chimeric antigen receptor (CAR)-T cells or CD19/CD3 bi-specific T-cell engagers, have been introduced to the field of oncology, leading to significant improvements in overall survival of adult cancer patients. During the past few years, the availability and approval of T-cell based immunotherapies have become a reality also for the treatment of childhood cancers. However, the distribution, ratio of regulatory to effector cells and the quality of T-cell responses early in life are distinct from those during adolescence and adulthood, raising the possibility that these differences impact the efficacy of immunotherapy. Herein we provide a brief overview of the properties of conventional T cell subsets during early life. Focusing on the most common cancer type during childhood, acute lymphoblastic leukemia (ALL), we describe how current conventional therapies used against ALL influence the T-cell compartment of small children. We describe early life T-cell responses in relation to immunotherapies engaging T-cell anticancer reactivity and present our opinion that it is not only immaturity of the adaptive immune system, but also the impact of an immunosuppressive environment that may prove disadvantageous in the setting of immunotherapies targeting pediatric cancer cells.
Collapse
Affiliation(s)
- Shanie Saghafian-Hedengren
- Division of Paediatric Oncology and Paediatric Surgery, Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
| | - Eva Sverremark-Ekström
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
| | - Anna Nilsson
- Division of Paediatric Oncology and Paediatric Surgery, Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
34
|
Perez-Diez A, Wong CS, Liu X, Mystakelis H, Song J, Lu Y, Sheikh V, Bourgeois JS, Lisco A, Laidlaw E, Cudrici C, Zhu C, Li QZ, Freeman AF, Williamson PR, Anderson M, Roby G, Tsang JS, Siegel R, Sereti I. Prevalence and pathogenicity of autoantibodies in patients with idiopathic CD4 lymphopenia. J Clin Invest 2021; 130:5326-5337. [PMID: 32634122 DOI: 10.1172/jci136254] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Accepted: 06/24/2020] [Indexed: 12/13/2022] Open
Abstract
BACKGROUNDIdiopathic CD4 lymphopenia (ICL) is defined by persistently low CD4+ cell counts (<300 cells/μL) in the absence of a causal infection or immune deficiency and can manifest with opportunistic infections. Approximately 30% of ICL patients develop autoimmune disease. The prevalence and breadth of their autoantibodies, however, and their potential contribution to pathogenesis of ICL remain unclear.METHODSWe hybridized 34 and 51 ICL patients' sera to a 9,000-human-proteome array and to a 128-known-autoantigen array, respectively. Using a flow-based method, we characterized the presence of anti-lymphocyte Abs in the whole cohort of 72 patients, as well as the Ab functional capability of inducing Ab-dependent cell-mediated cytotoxicity (ADCC), complement deposition, and complement-dependent cytotoxicity (CDC). We tested ex vivo the activation of the classical complement pathway on ICL CD4+ T cells.RESULTSAll ICL patients had a multitude of autoantibodies mostly directed against private (not shared) targets and unrelated quantitatively or qualitatively to the patients' autoimmune disease status. The targets included lymphocyte intracellular and membrane antigens, confirmed by the detection by flow of IgM and IgG (mostly IgG1 and IgG4) anti-CD4+ cell Abs in 50% of the patients, with half of these cases triggering lysis of CD4+ T cells. We also detected in vivo classical complement activation on CD4+ T cells in 14% of the whole cohort.CONCLUSIONOur data demonstrate that a high prevalence of autoantibodies in ICL, some of which are specific for CD4+ T cells, may contribute to pathogenesis, and may represent a potentially novel therapeutic target.TRIAL REGISTRATIONClinicalTrials.gov NCT00867269.FUNDINGNIAID and National Institute of Arthritis and Musculoskeletal and Skin Diseases of the NIH.
Collapse
Affiliation(s)
| | - Chun-Shu Wong
- HIV Pathogenesis Section, Laboratory of Immunoregulation, and
| | - Xiangdong Liu
- HIV Pathogenesis Section, Laboratory of Immunoregulation, and
| | | | - Jian Song
- Multiscale Systems Biology Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases (NIAID), and
| | - Yong Lu
- Multiscale Systems Biology Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases (NIAID), and
| | - Virginia Sheikh
- HIV Pathogenesis Section, Laboratory of Immunoregulation, and
| | | | - Andrea Lisco
- HIV Pathogenesis Section, Laboratory of Immunoregulation, and
| | | | - Cornelia Cudrici
- Immunoregulation Section, Autoimmunity Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, NIH, Bethesda, Maryland, USA
| | | | - Quan-Zhen Li
- Microarray Core Facility and.,Department of Immunology and Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | | | - Peter R Williamson
- Translational Mycology Section, Laboratory of Clinical and Molecular Immunology, NIAID, and
| | - Megan Anderson
- HIV Pathogenesis Section, Laboratory of Immunoregulation, and
| | - Gregg Roby
- HIV Pathogenesis Section, Laboratory of Immunoregulation, and
| | - John S Tsang
- Multiscale Systems Biology Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases (NIAID), and.,Trans-NIH Center for Human Immunology, NIH, Bethesda, Maryland, USA
| | - Richard Siegel
- Immunoregulation Section, Autoimmunity Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, NIH, Bethesda, Maryland, USA
| | - Irini Sereti
- HIV Pathogenesis Section, Laboratory of Immunoregulation, and
| |
Collapse
|
35
|
Xu X, Huang S, Xiao X, Sun Q, Liang X, Chen S, Zhao Z, Huo Z, Tu S, Li Y. Challenges and Clinical Strategies of CAR T-Cell Therapy for Acute Lymphoblastic Leukemia: Overview and Developments. Front Immunol 2021; 11:569117. [PMID: 33643279 PMCID: PMC7902522 DOI: 10.3389/fimmu.2020.569117] [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: 06/03/2020] [Accepted: 12/22/2020] [Indexed: 12/12/2022] Open
Abstract
Chimeric antigen receptor (CAR) T-cell therapy exhibits desirable and robust efficacy in patients with acute lymphoblastic leukemia (ALL). Stimulated by the revolutionized progress in the use of FDA-approved CD19 CAR T cells, novel agents with CAR designs and targets are being produced in pursuit of superior performance. However, on the path from bench to bedside, new challenges emerge. Accessibility is considered the initial barrier to the transformation of this patient-specific product into a commercially available product. To ensure infusion safety, profound comprehension of adverse events and proactive intervention are required. Additionally, resistance and relapse are the most critical and intractable issues in CAR T-cell therapy for ALL, thus precluding its further development. Understanding the limitations through up-to-date insights and characterizing multiple strategies will be critical to leverage CAR T-cell therapy flexibly for use in clinical situations. Herein, we provide an overview of the application of CAR T-cell therapy in ALL, emphasizing the main challenges and potential clinical strategies in an effort to promote a standardized set of treatment paradigms for ALL.
Collapse
Affiliation(s)
- Xinjie Xu
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, China.,The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China.,State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Shengkang Huang
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, China.,The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Xinyi Xiao
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, China.,The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Qihang Sun
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, China.,The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Xiaoqian Liang
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, China.,The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Sifei Chen
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, China.,The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Zijing Zhao
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, China.,The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Zhaochang Huo
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, China.,The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Sanfang Tu
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Yuhua Li
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
36
|
Shang L, Duah M, Xu Y, Liang Y, Wang D, Xia F, Li L, Sun Z, Yan Z, Xu K, Pan B. Dynamic of plasma IL-22 level is an indicator of thymic output after allogeneic hematopoietic cell transplantation. Life Sci 2021; 265:118849. [PMID: 33278390 DOI: 10.1016/j.lfs.2020.118849] [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: 09/11/2020] [Revised: 11/20/2020] [Accepted: 11/25/2020] [Indexed: 12/29/2022]
Abstract
AIMS Interleukin-22 (IL-22) promotes thymus recovery and improves T-cell recovery in preclinical allogeneic hematopoietic cell transplant models. However, the correlation between IL-22 and thymus recovery is unknown in human transplant. MATERIALS AND METHODS In this study, plasma IL-22 levels of transplanted humans were analyzed peri-transplant. Thymic output was assessed by detecting blood signal joint T-cell receptor excision circles (TRECs). Flow cytometry was applied to measure T-cell subsets. KEY FINDINGS Plasma IL-22 level positively correlated with blood TRECs level at days 14 and 28 posttransplant. Multiple linear regression analysis showed plasma IL-22 level, occurrence of acute graft-versus-host disease (aGVHD) and age were significantly associated with blood TRECs level at day 28 after allotransplant. An increase of plasma IL-22 level during day 14 and day 28 correlated with faster recovery of blood TRECs and naïve T-cell levels in allotransplant recipients. Recipients with high TRECs levels at day 28 had lower incidence of aGVHD comparing with those who with low TRECs levels according to a median split of their TRECs levels, an effect also seen in the high IL-22 level and low IL-22 level cohorts. Other factors such as age and infection had impacts on plasma IL-22 level in allotransplants. SIGNIFICANCE Our findings suggest that dynamic change of plasma IL-22 level is an indicator of thymic output and occurrence of aGVHD. Monitoring plasma IL-22 level might help to assess recovery of thymus function in human allotransplants.
Collapse
Affiliation(s)
- Longmei Shang
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou 221002, China
| | - Maxwell Duah
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou 221002, China
| | - Yan Xu
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou 221002, China
| | - Yiwen Liang
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou 221002, China
| | - Dong Wang
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou 221002, China
| | - Fan Xia
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou 221002, China; Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou Medical University, Xuzhou 221002, China
| | - Lingling Li
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou 221002, China
| | - Zengtian Sun
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou Medical University, Xuzhou 221002, China
| | - Zhiling Yan
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou 221002, China; Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou Medical University, Xuzhou 221002, China
| | - Kailin Xu
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou 221002, China; Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou Medical University, Xuzhou 221002, China.
| | - Bin Pan
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou 221002, China; Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou Medical University, Xuzhou 221002, China.
| |
Collapse
|
37
|
Jiang J, Ahuja S. Addressing Patient to Patient Variability for Autologous CAR T Therapies. J Pharm Sci 2021; 110:1871-1876. [PMID: 33340532 DOI: 10.1016/j.xphs.2020.12.015] [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: 09/23/2020] [Revised: 12/10/2020] [Accepted: 12/14/2020] [Indexed: 12/11/2022]
Abstract
Chimeric Antigen Receptor (CAR) T cell therapy clinical trials have had unprecedented success in the endeavors to cure cancer patients, particularly those having hematological cancers. As researchers learn more about the ways to make CAR T cells more effective to kill tumor cells, equally important will be understanding the differences between T cells from healthy donors and cancer patients and how these differences could affect ex vivo expansion of T cells during CAR T production. This undoubtedly could be a crucial factor in treating solid tumors, where CAR T cells are needed in significantly higher numbers. As the evidence for significant differences between the patients and healthy donors is compelling, an adaptable and robust production process should be designed to allow manufacture of the required CAR T cells for all cancer patients. Improving the fundamental understanding of the cellular metabolism and accompanying epigenetic and phenotypic changes during in vivo and ex vivo expansion of T cells will be just as important. Such discoveries will provide an invaluable tool box from which actionable knowledge could be drawn for designing an adaptable CAR T production process that is able to absorb the patient-to-patient variation.
Collapse
Affiliation(s)
- Jinlin Jiang
- Cell Culture and Fermentation Sciences, BioPharmaceutical Development, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD
| | - Sanjeev Ahuja
- Cell Culture and Fermentation Sciences, BioPharmaceutical Development, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD.
| |
Collapse
|
38
|
Bear AS, Vonderheide RH, O'Hara MH. Challenges and Opportunities for Pancreatic Cancer Immunotherapy. Cancer Cell 2020; 38:788-802. [PMID: 32946773 PMCID: PMC7738380 DOI: 10.1016/j.ccell.2020.08.004] [Citation(s) in RCA: 327] [Impact Index Per Article: 81.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Revised: 08/10/2020] [Accepted: 08/10/2020] [Indexed: 12/17/2022]
Abstract
Pancreatic ductal adenocarcinoma (PDA) is among the most immune-resistant tumor types. Its unique genomic landscape shaped by oncogenic drivers promotes immune suppression from the earliest stages of tumor inception to subvert adaptive T cell immunity. Single-agent immune modulators have thus far proven clinically ineffective, and multi-modal therapies targeting mechanisms of immunotherapy resistance are likely needed. Here, we review novel immunotherapy strategies currently under investigation to (1) confer antigen specificity, (2) enhance T cell effector function, and (3) neutralize immunosuppressive elements within the tumor microenvironment that may be rationally combined to untangle the web of immune resistance in PDA and other tumors.
Collapse
Affiliation(s)
- Adham S Bear
- Division of Hematology-Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Robert H Vonderheide
- Division of Hematology-Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA.
| | - Mark H O'Hara
- Division of Hematology-Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA. mark.o'
| |
Collapse
|
39
|
Mika T, Ladigan-Badura S, Maghnouj A, Mustafa B, Klein-Scory S, Baraniskin A, Döhring S, Fuchs I, Ehl S, Hahn SA, Schroers R. Altered T-Lymphocyte Biology Following High-Dose Melphalan and Autologous Stem Cell Transplantation With Implications for Adoptive T-Cell Therapy. Front Oncol 2020; 10:568056. [PMID: 33363008 PMCID: PMC7759611 DOI: 10.3389/fonc.2020.568056] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Accepted: 10/20/2020] [Indexed: 01/18/2023] Open
Abstract
In relapsed and refractory multiple myeloma (MM), adoptive cell therapies (ACT) including CAR-T-cells are under clinical investigation. However, relapse due to T-cell exhaustion or limited persistence is an obstacle. Before ACT are considered in MM, high-dose (HD) melphalan followed by autologous stem-cell transplantation (autoSCT) has been administered in most clinical situations. Yet, the impact of HD chemotherapy on T-cells in MM with respect to ACT is unclear. In this study, T-lymphocytes’ phenotypes, expansion properties, lentiviral transduction efficacy, and gene expression were examined with special respect to patients following HD melphalan. Significant impairment of T-cells’ expansion and transduction rates could be demonstrated. Expansion was diminished due to inherent disadvantages of the predominant T-cell phenotype but restored over time. The quantitative fraction of CD27−/CD28− T-cells before expansion was predictive of T-cell yield. Following autoSCT, the transduction efficacy was reduced by disturbed lentiviral genome integration. Moreover, an unfavorable T-cell phenotype after expansion was demonstrated. In initial analyses of CD107a degranulation impaired T-cell cytotoxicity was detected in one patient following melphalan and autoSCT. The findings of our study have potential implications regarding the time point of leukapheresis for CAR-T-cell manufacturing. Our results point to a preferred interval of more than 3 months until patients should undergo cell separation for CAR-T therapy in the specific situation post-HD melphalan/autoSCT. Monitoring of CD27−/CD28− T-cells, has the potential to influence clinical decision making before apheresis in MM.
Collapse
Affiliation(s)
- Thomas Mika
- Department of Medicine, Hematology and Oncology, Ruhr-University Bochum, Bochum, Germany.,Department of Molecular Gastrointestinal Oncology, Ruhr-University Bochum, Bochum, Germany
| | - Swetlana Ladigan-Badura
- Department of Medicine, Hematology and Oncology, Ruhr-University Bochum, Bochum, Germany.,Department of Molecular Gastrointestinal Oncology, Ruhr-University Bochum, Bochum, Germany
| | - Abdelouahid Maghnouj
- Department of Molecular Gastrointestinal Oncology, Ruhr-University Bochum, Bochum, Germany
| | - Bakr Mustafa
- Department of Molecular Gastrointestinal Oncology, Ruhr-University Bochum, Bochum, Germany
| | - Susanne Klein-Scory
- Department of Medicine, Hematology and Oncology, Ruhr-University Bochum, Bochum, Germany
| | - Alexander Baraniskin
- Department of Medicine, Hematology and Oncology, Ruhr-University Bochum, Bochum, Germany
| | - Sascha Döhring
- Department of Molecular Gastrointestinal Oncology, Ruhr-University Bochum, Bochum, Germany
| | - Ilka Fuchs
- Center for Chronic Immunodeficiency, Faculty of Medicine, University of Freiburg, Freiburg im Breisgau, Germany
| | - Stephan Ehl
- Center for Chronic Immunodeficiency, Faculty of Medicine, University of Freiburg, Freiburg im Breisgau, Germany
| | - Stephan A Hahn
- Department of Molecular Gastrointestinal Oncology, Ruhr-University Bochum, Bochum, Germany
| | - Roland Schroers
- Department of Medicine, Hematology and Oncology, Ruhr-University Bochum, Bochum, Germany
| |
Collapse
|
40
|
Wang M, Liang H, Wang W, Zhao S, Cai X, Zhao Y, Li C, Cheng B, Xiong S, Li J, He J, Liang W. Immune-related adverse events of a PD-L1 inhibitor plus chemotherapy versus a PD-L1 inhibitor alone in first-line treatment for advanced non-small cell lung cancer: A meta-analysis of randomized control trials. Cancer 2020; 127:777-786. [PMID: 33119182 DOI: 10.1002/cncr.33270] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Revised: 07/02/2020] [Accepted: 07/23/2020] [Indexed: 12/19/2022]
Abstract
BACKGROUND The addition of chemotherapy to a programmed death 1/programmed death ligand 1 (PD-L1) inhibitor is a more effective option as a first-line treatment for advanced non-small cell lung cancer (NSCLC). It might also inhibit an overactive immune response and thereby reduce immune-related adverse events (irAEs). This meta-analysis assessed the rate of irAEs with a PD-(L)1 inhibitor plus chemotherapy (I+C) versus a PD-(L)1 inhibitor alone (I) and evaluated the indirect relative risk (RR) of I+C versus I. METHODS The protocol of this study was registered with PROSPERO (CRD42020139923). The pooled rates of irAEs at different grades were calculated by a single-arm meta-analysis weighted by sample size, and RRs were determined by direct meta-analysis and indirect treatment comparison. RESULTS Overall, I+C had a lower rate of grade 3 or higher irAEs than I (7.1% vs 10.6%; indirect RR, 0.516; 95% confidence interval [CI], 0.291-0.916), although irAEs of any grade were similar. The rate of pneumonitis with I+C was lower than the rate with I for any grade (5.9% vs 7.1%; indirect RR, 0.217; 95% CI, 0.080-0.588) and for grade 3 or higher. In the endocrine system, I+C was associated with a lower overall ratein comparison with I (16.1% vs 20.1%; indirect RR, 0.260; 95% CI, 0.120-0.564), whereas irAEs of the digestive system were similar with I+C and I. In other systems, I+C decreased the rate of skin reactions, including rash, in comparison with I (10.4% vs 12.9%; indirect RR, 0.474; 95% CI, 0.299-0.751). The rate of grade 3 or higher skin reactions (excluding rash) also decreased with I+C versus I (1.1% vs 2.0%) with an indirect RR of 0.158 (95% CI, 0.032-0.765), whereas other included irAEs were similar. CONCLUSIONS In comparison with a PD-(L)1 inhibitor alone, a combination with chemotherapy for the first-line treatment of NSCLC decreased the rates of most irAEs, such as pneumonitis and endocrine and skin reactions, and the overall rate. LAY SUMMARY In the first-line treatment of advanced non-small cell lung cancer (NSCLC), the addition of chemotherapy to a programmed death 1/programmed death ligand 1 (PD-(L)1) inhibitor is a more effective option. Adding chemotherapy might reduce immune-related adverse events (irAEs). Thus, this article assesses the rate of irAEs with a PD-(L)1 inhibitor plus chemotherapy (I+C) in comparison with a PD-(L)1 inhibitor alone (I) and evaluates the indirect relative risk (RR) with I+C versus I. The key finding is that in comparison with a PD-(L)1 inhibitor alone, a combination with chemotherapy for the first-line treatment of NSCLC decreases the rates of most irAEs, such as pneumonitis and endocrine and skin reactions, and the overall rate.
Collapse
Affiliation(s)
- Manting Wang
- Department of Thoracic Surgery and Oncology, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.,State Key Laboratory of Respiratory Disease, Guangzhou, China.,National Clinical Research Center for Respiratory Disease, Guangzhou, China.,Guangzhou Institute of Respiratory Health, Guangzhou, China.,Nanshan School, Clinical Medicine Department, Guangzhou Medical University, Guangzhou, China
| | - Hengrui Liang
- Department of Thoracic Surgery and Oncology, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.,State Key Laboratory of Respiratory Disease, Guangzhou, China.,National Clinical Research Center for Respiratory Disease, Guangzhou, China.,Guangzhou Institute of Respiratory Health, Guangzhou, China
| | - Wei Wang
- Department of Thoracic Surgery and Oncology, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.,State Key Laboratory of Respiratory Disease, Guangzhou, China.,National Clinical Research Center for Respiratory Disease, Guangzhou, China.,Guangzhou Institute of Respiratory Health, Guangzhou, China
| | - Shen Zhao
- Department of Medical Oncology, Sun Yat-Sen University Cancer Center, Guangzhou, China.,State Key Laboratory of Oncology in South China, Guangzhou, China.,Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Xiuyu Cai
- Department of Medical Oncology, Sun Yat-Sen University Cancer Center, Guangzhou, China.,State Key Laboratory of Oncology in South China, Guangzhou, China.,Collaborative Innovation Center for Cancer Medicine, Guangzhou, China.,Department of General Internal Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Yi Zhao
- Department of Thoracic Surgery and Oncology, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.,State Key Laboratory of Respiratory Disease, Guangzhou, China.,National Clinical Research Center for Respiratory Disease, Guangzhou, China.,Guangzhou Institute of Respiratory Health, Guangzhou, China
| | - Caichen Li
- Department of Thoracic Surgery and Oncology, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.,State Key Laboratory of Respiratory Disease, Guangzhou, China.,National Clinical Research Center for Respiratory Disease, Guangzhou, China.,Guangzhou Institute of Respiratory Health, Guangzhou, China
| | - Bo Cheng
- Department of Thoracic Surgery and Oncology, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.,State Key Laboratory of Respiratory Disease, Guangzhou, China.,National Clinical Research Center for Respiratory Disease, Guangzhou, China.,Guangzhou Institute of Respiratory Health, Guangzhou, China
| | - Shan Xiong
- Department of Thoracic Surgery and Oncology, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.,State Key Laboratory of Respiratory Disease, Guangzhou, China.,National Clinical Research Center for Respiratory Disease, Guangzhou, China.,Guangzhou Institute of Respiratory Health, Guangzhou, China
| | - Jianfu Li
- Department of Thoracic Surgery and Oncology, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.,State Key Laboratory of Respiratory Disease, Guangzhou, China.,National Clinical Research Center for Respiratory Disease, Guangzhou, China.,Guangzhou Institute of Respiratory Health, Guangzhou, China
| | - Jianxing He
- Department of Thoracic Surgery and Oncology, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.,State Key Laboratory of Respiratory Disease, Guangzhou, China.,National Clinical Research Center for Respiratory Disease, Guangzhou, China.,Guangzhou Institute of Respiratory Health, Guangzhou, China
| | - Wenhua Liang
- Department of Thoracic Surgery and Oncology, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.,State Key Laboratory of Respiratory Disease, Guangzhou, China.,National Clinical Research Center for Respiratory Disease, Guangzhou, China.,Guangzhou Institute of Respiratory Health, Guangzhou, China
| |
Collapse
|
41
|
Abstract
Following periods of haematopoietic cell stress, such as after chemotherapy, radiotherapy, infection and transplantation, patient outcomes are linked to the degree of immune reconstitution, specifically of T cells. Delayed or defective recovery of the T cell pool has significant clinical consequences, including prolonged immunosuppression, poor vaccine responses and increased risks of infections and malignancies. Thus, strategies that restore thymic function and enhance T cell reconstitution can provide considerable benefit to individuals whose immune system has been decimated in various settings. In this Review, we focus on the causes and consequences of impaired adaptive immunity and discuss therapeutic strategies that can recover immune function, with a particular emphasis on approaches that can promote a diverse repertoire of T cells through de novo T cell formation.
Collapse
|
42
|
Cappell KM, Sherry RM, Yang JC, Goff SL, Vanasse DA, McIntyre L, Rosenberg SA, Kochenderfer JN. Long-Term Follow-Up of Anti-CD19 Chimeric Antigen Receptor T-Cell Therapy. J Clin Oncol 2020; 38:3805-3815. [PMID: 33021872 DOI: 10.1200/jco.20.01467] [Citation(s) in RCA: 165] [Impact Index Per Article: 41.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
PURPOSE Anti-CD19 chimeric antigen receptors (CARs) are artificial fusion proteins that cause CD19-specific T-cell activation. Durability of remissions and incidence of long-term adverse events are critical factors determining the utility of anti-CD19 CAR T-cell therapy, but long-term follow-up of patients treated with anti-CD19 CAR T cells is limited. This work provides the longest follow-up of patients in remission after anti-CD19 CAR T-cell therapy. METHODS Between 2009 and 2015, we administered 46 CAR T-cell treatments to 43 patients (ClinicalTrials.gov identifier: NCT00924326). Patients had relapsed B-cell malignancies of the following types: diffuse large B-cell lymphoma or primary mediastinal B-cell lymphoma (DLBCL/PMBCL; n = 28), low-grade B-cell lymphoma (n = 8), or chronic lymphocytic leukemia (CLL; n = 7). This report focuses on long-term outcomes of these patients. The CAR used was FMC63-28Z; axicabtagene ciloleucel uses the same CAR. Cyclophosphamide plus fludarabine conditioning chemotherapy was administered before CAR T cells. RESULTS The percentages of CAR T-cell treatments resulting in a > 3-year duration of response (DOR) were 51% (95% CI, 35% to 67%) for all evaluable treatments, 48% (95% CI, 28% to 69%) for DLBCL/PMBCL, 63% (95% CI, 25% to 92%) for low-grade lymphoma, and 50% (95% CI, 16% to 84%) for CLL. The median event-free survival of all 45 evaluable treatments was 55 months. Long-term adverse effects were rare, except for B-cell depletion and hypogammaglobulinemia. Median peak blood CAR-positive cell levels were higher among patients with a DOR of > 3 years (98/µL; range, 9-1,217/µL) than among patients with a DOR of < 3 years (18/µL; range, 0-308/μL, P = .0051). CONCLUSION Complete remissions of a variety of B-cell malignancies lasting ≥ 3 years occurred after 51% of evaluable anti-CD19 CAR T-cell treatments. Remissions of up to 9 years are ongoing. Late adverse events were rare.
Collapse
Affiliation(s)
- Kathryn M Cappell
- National Institutes of Health (NIH)/National Heart, Lung, and Blood Institute Medical Oncology/Hematology Fellowship Program, Bethesda, MD
| | - Richard M Sherry
- Surgery Branch, Center for Cancer Research, National Cancer Istitute, NIH, Bethesda, MD
| | - James C Yang
- Surgery Branch, Center for Cancer Research, National Cancer Istitute, NIH, Bethesda, MD
| | - Stephanie L Goff
- Surgery Branch, Center for Cancer Research, National Cancer Istitute, NIH, Bethesda, MD
| | - Danielle A Vanasse
- Surgery Branch, Center for Cancer Research, National Cancer Istitute, NIH, Bethesda, MD
| | - Lori McIntyre
- Surgery Branch, Center for Cancer Research, National Cancer Istitute, NIH, Bethesda, MD
| | - Steven A Rosenberg
- Surgery Branch, Center for Cancer Research, National Cancer Istitute, NIH, Bethesda, MD
| | - James N Kochenderfer
- Surgery Branch, Center for Cancer Research, National Cancer Istitute, NIH, Bethesda, MD
| |
Collapse
|
43
|
Wang JH. Why the Outcome of Anti-Tumor Immune Responses is Heterogeneous: A Novel Idea in the Context of Immunological Heterogeneity in Cancers. Bioessays 2020; 42:e2000024. [PMID: 32767371 DOI: 10.1002/bies.202000024] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Revised: 07/12/2020] [Indexed: 12/12/2022]
Abstract
The question as to why some hosts can eradicate their tumors while others succumb to tumor-progression remains unanswered. Here, a provocative concept is proposed that intrinsic differences in the T cell receptor (TCR) repertoire of individuals may influence the outcome of anti-tumor immunity by affecting the frequency and/or variety of tumor-reactive CD8 and/or CD4 tumor-infiltrating lymphocytes. This idea implicates that the TCR repertoire in a given patient might not provide sufficiently different TCR clones that can recognize tumor antigens, namely, "a hole in the TCR repertoire" might exist. This idea may provide a novel perspective to further dissect the mechanisms underlying heterogeneous anti-tumor immune responses in different hosts. Besides tumor-intrinsic heterogeneity and host microbiome, the various factors that may constantly shape the dynamic TCR repertoire are also discussed. Elucidating mechanistic differences in different individuals' immune systems will allow to better harness immune system to design new personalized cancer immunotherapy.
Collapse
Affiliation(s)
- Jing H Wang
- Department of Immunology and Microbiology, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| |
Collapse
|
44
|
Quinn KM, Kartikasari AER, Cooke RE, Koldej RM, Ritchie DS, Plebanski M. Impact of age-, cancer-, and treatment-driven inflammation on T cell function and immunotherapy. J Leukoc Biol 2020; 108:953-965. [PMID: 32678927 DOI: 10.1002/jlb.5mr0520-466r] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Revised: 06/16/2020] [Accepted: 06/03/2020] [Indexed: 12/11/2022] Open
Abstract
Many cancers are predominantly diagnosed in older individuals and chronic inflammation has a major impact on the overall health and immune function of older cancer patients. Chronic inflammation is a feature of aging, it can accelerate disease in many cancers and it is often exacerbated during conventional treatments for cancer. This review will provide an overview of the factors that lead to increased inflammation in older individuals and/or individuals with cancer, as well as those that result from conventional treatments for cancer, using ovarian cancer (OC) and multiple myeloma (MM) as key examples. We will also consider the impact of chronic inflammation on immune function, with a particular focus on T cells as they are key targets for novel cancer immunotherapies. Overall, this review aims to highlight specific pathways for potential interventions that may be able to mitigate the impact of chronic inflammation in older cancer patients.
Collapse
Affiliation(s)
- Kylie M Quinn
- School of Health and Biomedical Sciences, RMIT University, Bundoora, Victoria, Australia.,Department of Biochemistry, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| | | | - Rachel E Cooke
- Australian Cancer Research Foundation (ACRF) Translational Laboratory, Royal Melbourne Hospital, Melbourne, Australia.,Department of Medicine, University of Melbourne, Melbourne, Australia
| | - Rachel M Koldej
- Australian Cancer Research Foundation (ACRF) Translational Laboratory, Royal Melbourne Hospital, Melbourne, Australia.,Department of Medicine, University of Melbourne, Melbourne, Australia
| | - David S Ritchie
- Australian Cancer Research Foundation (ACRF) Translational Laboratory, Royal Melbourne Hospital, Melbourne, Australia.,Department of Medicine, University of Melbourne, Melbourne, Australia
| | - Magdalena Plebanski
- School of Health and Biomedical Sciences, RMIT University, Bundoora, Victoria, Australia
| |
Collapse
|
45
|
Park J, Lim SH, Kim SH, Yun J, Kim CK, Lee SC, Won JH, Hong DS, Park SK. Is immunological recovery clinically relevant at 100 days after allogeneic transplantation? Korean J Intern Med 2020; 35:957-969. [PMID: 32306712 PMCID: PMC7373953 DOI: 10.3904/kjim.2018.414] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Accepted: 03/23/2019] [Indexed: 01/26/2023] Open
Abstract
BACKGROUND/AIMS Immune reconstitution following allogeneic hematopoietic stem cell transplantation (HSCT) is affected by multiple variables during the transplantation. METHODS We assessed the clinical factors contributing to immune function reconstitution at 100 days post-allogeneic HSCT in 114 patients receiving fludarabine-based conditioning. Immunophenotypic analysis using flow cytometry was performed to evaluate the percentage and the absolute numbers of T-cell subsets, natural killer cells, and B-cells as clinical outcomes. RESULTS Tacrolimus-based graft-versus-host disease (GVHD) prophylaxis, T-cell depletion, and acute GVHD were significantly associated with delayed immune reconstitution of T-cell subsets. The incidence of chronic GVHD was significantly increased in the normal recovery group compared to the abnormal group (p = 0.01). Epstein-Barr virus reactivation was more frequently observed in the abnormal group of T-cell subsets (p = 0.045). All viral reactivation events including cytomegalovirus reactivation appeared to be more frequent in the abnormal group of T-cell subsets. CONCLUSION The immune recovery status post-allogeneic HSCT was affected by GVHD prophylactic regimens, especially in cases receiving tacrolimus-based GVHD prophylaxis, T-cell depletion, and possibly those manifesting acute GVHD. Delayed immune reconstitution might increase the morbidity due to viral reactivation. Treatment strategies are needed to prevent infectious complications and enhance immune reconstitution based on the immune recovery status following allogeneic HSCT with fludarabine-based conditioning.
Collapse
Affiliation(s)
- Jin Park
- Division of Hemato-oncology, Department of Internal Medicine, Soonchunhyang University Bucheon Hospital, Bucheon, Korea
| | - Sung Hee Lim
- Division of Hemato-oncology, Department of Internal Medicine, Soonchunhyang University Bucheon Hospital, Bucheon, Korea
| | - Se Hyung Kim
- Division of Hemato-oncology, Department of Internal Medicine, Soonchunhyang University Bucheon Hospital, Bucheon, Korea
| | - Jina Yun
- Division of Hemato-oncology, Department of Internal Medicine, Soonchunhyang University Bucheon Hospital, Bucheon, Korea
| | - Chan Kyu Kim
- Division of Hemato-oncology, Department of Internal Medicine, Soonchunhyang University Bucheon Hospital, Bucheon, Korea
| | - Sang Cheol Lee
- Division of Hemato-oncology, Department of Internal Medicine, Soonchunhyang University Cheonan Hospital, Cheonan, Korea
| | - Jong Ho Won
- Division of Hemato-oncology, Department of Internal Medicine, Soonchunhyang University Seoul Hospital, Seoul, Korea
| | - Dae Sik Hong
- Division of Hemato-oncology, Department of Internal Medicine, Soonchunhyang University Bucheon Hospital, Bucheon, Korea
| | - Seong Kyu Park
- Division of Hemato-oncology, Department of Internal Medicine, Soonchunhyang University Bucheon Hospital, Bucheon, Korea
- Correspondence to Seong Kyu Park, M.D. Division of Hemato-oncology, Department of Internal Medicine, Soonchunhyang University Bucheon, Hospital, 170 Jomaru-ro, Wonmi-gu, Bucheon 14584, Korea Tel: +82-32-621-5185 Fax: +82-32-621-5018 E-mail:
| |
Collapse
|
46
|
Lycke J, Lenhoff S. Intensive immunosuppression followed by autologous hematopoietic stem cell transplantation for the treatment of multiple sclerosis. Ther Adv Neurol Disord 2020; 13:1756286420929467. [PMID: 32636931 PMCID: PMC7315665 DOI: 10.1177/1756286420929467] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Accepted: 03/22/2020] [Indexed: 12/20/2022] Open
Abstract
Autologous hematopoietic stem cell transplantation (AHSCT) to treat multiple sclerosis (MS) has mostly been used in devastating cases as the last option to stop further neurological deterioration. However, evidence from several retrospective clinical trials indicates that young, less disabled patients with highly inflammatory active MS are the most likely to benefit from AHSCT, and after moving from high-intensity to nonmyeloablative procedures the tolerability of AHSCT has increased and its associated risk and mortality have declined considerably. Recent meta-analyses and randomized clinical trials show that AHSCT is more effective than currently approved disease-modifying therapies (DMTs), with suppression of disease activity in 70-90% of patients and long-term cessation of disease activity in two-thirds of treated patients. The rationale for AHSCT is to eliminate autoimmunity and achieve immune resetting by intense immunosuppression followed by infusion of autologous hematopoietic stem cells. Similar effects on the immune system have been suggested for cladribine and alemtuzumab treatment and, together with AHSCT, they constitute the induction or immune-reconstitution therapies for MS. Although, further randomized controlled trials of AHSCT for MS are needed, it has become clear that improved patient selection and lower intensity conditioning regimens have reduced AHSCT associated risks and mortality and strengthened the position of AHSCT among other DMTs. Do we have enough experience and scientific support for AHSCT in MS to move from an exclusive treatment for aggressive, treatment-resistant MS and acquire broader indications, similar to other effective DMTs?
Collapse
Affiliation(s)
- Jan Lycke
- Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gröna stråket 11, 3 tr, Sahlgrenska University Hospital, Gothenburg, 415 45, Sweden
| | - Stig Lenhoff
- Department of Hematology, Oncology and Radiophysics, Skane University Hospital, Lund, Sweden
| |
Collapse
|
47
|
Laws HJ, Baumann U, Bogdan C, Burchard G, Christopeit M, Hecht J, Heininger U, Hilgendorf I, Kern W, Kling K, Kobbe G, Külper W, Lehrnbecher T, Meisel R, Simon A, Ullmann A, de Wit M, Zepp F. Impfen bei Immundefizienz. Bundesgesundheitsblatt Gesundheitsforschung Gesundheitsschutz 2020; 63:588-644. [PMID: 32350583 PMCID: PMC7223132 DOI: 10.1007/s00103-020-03123-w] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Hans-Jürgen Laws
- Klinik für Kinder-Onkologie, -Hämatologie und Klinische Immunologie, Universitätsklinikum Düsseldorf, Düsseldorf, Deutschland
| | - Ulrich Baumann
- Klinik für Pädiatrische Pneumologie, Allergologie und Neonatologie, Medizinische Hochschule Hannover, Hannover, Deutschland
| | - Christian Bogdan
- Mikrobiologisches Institut - Klinische Mikrobiologie, Immunologie und Hygiene, Universitätsklinikum Erlangen, Friedrich-Alexander Universität FAU Erlangen-Nürnberg, Erlangen, Deutschland
- Ständige Impfkommission (STIKO), Robert Koch-Institut, Berlin, Deutschland
| | - Gerd Burchard
- Ständige Impfkommission (STIKO), Robert Koch-Institut, Berlin, Deutschland
- Bernhard-Nocht-Institut für Tropenmedizin, Hamburg, Deutschland
| | - Maximilian Christopeit
- Interdisziplinäre Klinik für Stammzelltransplantation, Universitätsklinikum Eppendorf, Hamburg, Deutschland
| | - Jane Hecht
- Abteilung für Infektionsepidemiologie, Fachgebiet Nosokomiale Infektionen, Surveillance von Antibiotikaresistenz und -verbrauch, Robert Koch-Institut, Berlin, Deutschland
| | - Ulrich Heininger
- Ständige Impfkommission (STIKO), Robert Koch-Institut, Berlin, Deutschland
- Universitäts-Kinderspital beider Basel, Basel, Schweiz
| | - Inken Hilgendorf
- Klinik für Innere Medizin II, Abteilung für Hämatologie und Internistische Onkologie, Universitätsklinikum Jena, Jena, Deutschland
| | - Winfried Kern
- Klinik für Innere Medizin II, Abteilung Infektiologie, Universitätsklinikum Freiburg, Freiburg, Deutschland
| | - Kerstin Kling
- Abteilung für Infektionsepidemiologie, Fachgebiet Impfprävention, Robert Koch-Institut, Berlin, Deutschland.
| | - Guido Kobbe
- Klinik für Hämatologie, Onkologie und Klinische Immunologie, Universitätsklinikum Düsseldorf, Düsseldorf, Deutschland
| | - Wiebe Külper
- Abteilung für Infektionsepidemiologie, Fachgebiet Impfprävention, Robert Koch-Institut, Berlin, Deutschland
| | - Thomas Lehrnbecher
- Klinik für Kinder- und Jugendmedizin, Universitätsklinikum Frankfurt, Frankfurt am Main, Deutschland
| | - Roland Meisel
- Klinik für Kinder-Onkologie, -Hämatologie und Klinische Immunologie, Universitätsklinikum Düsseldorf, Düsseldorf, Deutschland
| | - Arne Simon
- Klinik für Pädiatrische Onkologie und Hämatologie, Universitätsklinikum des Saarlandes, Homburg/Saar, Deutschland
| | - Andrew Ullmann
- Medizinische Klinik und Poliklinik II, Universitätsklinikum Würzburg, Würzburg, Deutschland
| | - Maike de Wit
- Klinik für Innere Medizin - Hämatologie, Onkologie und Palliativmedizin, Vivantes Klinikum Neukölln, Berlin, Deutschland
- Klinik für Innere Medizin - Onkologie, Vivantes Auguste-Viktoria-Klinikum, Berlin, Deutschland
| | - Fred Zepp
- Ständige Impfkommission (STIKO), Robert Koch-Institut, Berlin, Deutschland
- Zentrum für Kinder- und Jugendmedizin, Universitätsmedizin Mainz, Mainz, Deutschland
| |
Collapse
|
48
|
Serrano-Del Valle A, Naval J, Anel A, Marzo I. Novel Forms of Immunomodulation for Cancer Therapy. Trends Cancer 2020; 6:518-532. [PMID: 32460005 DOI: 10.1016/j.trecan.2020.02.015] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Revised: 02/07/2020] [Accepted: 02/19/2020] [Indexed: 02/07/2023]
Abstract
In recent years immunotherapy has provided new hope for cancer patients. However, some patients eventually relapse. Immunological responses are thought to underlie the long-term effects of conventional or targeted therapies. Whether this influence emerges from direct effects on cancer cells through immunogenic cell death (ICD) or by modulating the immune environment requires further clarification. ICD-related molecular mechanisms are also shared by cell-intrinsic defense responses that combat foreign intrusions. Indeed, we could potentially mimic and harness these processes to improve cancer immunogenicity. In addition, the microbiome is materializing as a missing factor in the cancer-immune therapy axis. The emerging idea of manipulating the gut microbiota to improve responses to anticancer therapy is becoming increasingly popular, but further clinical authentication is needed.
Collapse
Affiliation(s)
- Alfonso Serrano-Del Valle
- Apoptosis, Immunity, and Cancer Group, Department of Biochemistry and Molecular and Cell Biology, University of Zaragoza, and Aragon Health Research Institute (IIS-Aragon), Zaragoza 50009, Spain.
| | - Javier Naval
- Apoptosis, Immunity, and Cancer Group, Department of Biochemistry and Molecular and Cell Biology, University of Zaragoza, and Aragon Health Research Institute (IIS-Aragon), Zaragoza 50009, Spain
| | - Alberto Anel
- Apoptosis, Immunity, and Cancer Group, Department of Biochemistry and Molecular and Cell Biology, University of Zaragoza, and Aragon Health Research Institute (IIS-Aragon), Zaragoza 50009, Spain
| | - Isabel Marzo
- Apoptosis, Immunity, and Cancer Group, Department of Biochemistry and Molecular and Cell Biology, University of Zaragoza, and Aragon Health Research Institute (IIS-Aragon), Zaragoza 50009, Spain
| |
Collapse
|
49
|
Ramakrishna S, Barsan V, Mackall C. Prospects and challenges for use of CAR T cell therapies in solid tumors. Expert Opin Biol Ther 2020; 20:503-516. [DOI: 10.1080/14712598.2020.1738378] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Sneha Ramakrishna
- Department of Pediatrics, Bass Center for Childhood Cancer and Blood Disorders, Center for Cancer Cell Therapy, Stanford, USA
| | - Valentin Barsan
- Department of Pediatrics, Bass Center for Childhood Cancer and Blood Disorders, Center for Cancer Cell Therapy, Stanford, USA
| | - Crystal Mackall
- Department of Pediatrics, Bass Center for Childhood Cancer and Blood Disorders, Center for Cancer Cell Therapy, Stanford, USA
- Stanford Cancer Institute, Stanford University, Stanford, USA
- Department of Medicine, Stanford University, Stanford, USA
| |
Collapse
|
50
|
Gustafson CE, Jadhav R, Cao W, Qi Q, Pegram M, Tian L, Weyand CM, Goronzy JJ. Immune cell repertoires in breast cancer patients after adjuvant chemotherapy. JCI Insight 2020; 5:134569. [PMID: 32102986 PMCID: PMC7101137 DOI: 10.1172/jci.insight.134569] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Accepted: 01/29/2020] [Indexed: 12/24/2022] Open
Abstract
Adjuvant chemotherapy in breast cancer patients causes immune cell depletion at an age when the regenerative capacity is compromised. Successful regeneration requires the recovery of both quantity and quality of immune cell subsets. Although immune cell numbers rebound within a year after treatment, it is unclear whether overall compositional diversity is recovered. We investigated the regeneration of immune cell complexity by comparing peripheral blood mononuclear cells from breast cancer patients ranging from 1-5 years after chemotherapy with those of age-matched healthy controls using mass cytometry and T cell receptor sequencing. These data reveal universal changes in patients' CD4+ T cells that persisted for years and consisted of expansion of Th17-like CD4 memory populations with incomplete recovery of CD4+ naive T cells. Conversely, CD8+ T cells fully recovered within a year. Mechanisms of T cell regeneration, however, were unbiased, as CD4+ and CD8+ T cell receptor diversity remained high. Likewise, terminal differentiated effector memory cells were not expanded, indicating that regeneration was not driven by recognition of latent viruses. These data suggest that, while CD8+ T cell immunity is successfully regenerated, the CD4 compartment may be irreversibly affected. Moreover, the bias of CD4 memory toward inflammatory effector cells may impact responses to vaccination and infection.
Collapse
Affiliation(s)
- Claire E Gustafson
- Division of Immunology and Rheumatology, Department of Medicine, Stanford University School of Medicine, Stanford, California, USA.,Department of Medicine, Veterans Administration Healthcare System, Palo Alto, California, USA
| | - Rohit Jadhav
- Division of Immunology and Rheumatology, Department of Medicine, Stanford University School of Medicine, Stanford, California, USA.,Department of Medicine, Veterans Administration Healthcare System, Palo Alto, California, USA
| | - Wenqiang Cao
- Division of Immunology and Rheumatology, Department of Medicine, Stanford University School of Medicine, Stanford, California, USA.,Department of Medicine, Veterans Administration Healthcare System, Palo Alto, California, USA
| | - Qian Qi
- Division of Immunology and Rheumatology, Department of Medicine, Stanford University School of Medicine, Stanford, California, USA.,Department of Medicine, Veterans Administration Healthcare System, Palo Alto, California, USA
| | | | - Lu Tian
- Department of Biomedical Data Science, Stanford University School of Medicine, Stanford, California, USA
| | - Cornelia M Weyand
- Division of Immunology and Rheumatology, Department of Medicine, Stanford University School of Medicine, Stanford, California, USA.,Department of Medicine, Veterans Administration Healthcare System, Palo Alto, California, USA
| | - Jorg J Goronzy
- Division of Immunology and Rheumatology, Department of Medicine, Stanford University School of Medicine, Stanford, California, USA.,Department of Medicine, Veterans Administration Healthcare System, Palo Alto, California, USA
| |
Collapse
|