51
|
Shah NA. Donor lymphocyte infusion in Acute Myeloid Leukemia. Best Pract Res Clin Haematol 2023; 36:101484. [PMID: 37612002 DOI: 10.1016/j.beha.2023.101484] [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: 04/17/2023] [Revised: 05/30/2023] [Accepted: 05/31/2023] [Indexed: 08/25/2023]
Abstract
Donor lymphocyte infusion (DLI) is an important treatment modality in the management of relapsed hematological malignancies after allogeneic hematopoietic cell transplantation (allo-HCT). Donor T lymphocytes can be used in a therapeutic, pre-emptive or prophylactic manner in an attempt to stimulate a graft versus leukemia (GVL) effect and eradicate residual disease or even prevent relapse in a high-risk setting. DLIs are not without complications, however, graft versus host disease (GVHD) in particular. Data to date is limited to retrospective and small prospective studies. This review summarizes the available literature on approaches to managing relapse, dosing and timing of DLI, complications and potential future therapies.
Collapse
|
52
|
Vallet N, Salmona M, Malet-Villemagne J, Bredel M, Bondeelle L, Tournier S, Mercier-Delarue S, Cassonnet S, Ingram B, Peffault de Latour R, Bergeron A, Socié G, Le Goff J, Lepage P, Michonneau D. Circulating T cell profiles associate with enterotype signatures underlying hematological malignancy relapses. Cell Host Microbe 2023; 31:1386-1403.e6. [PMID: 37463582 DOI: 10.1016/j.chom.2023.06.009] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 05/23/2023] [Accepted: 06/19/2023] [Indexed: 07/20/2023]
Abstract
Early administration of azithromycin after allogeneic hematopoietic stem cell transplantation was shown to increase the relapse of hematological malignancies. To determine the impact of azithromycin on the post-transplant gut ecosystem and its influence on relapse, we characterized overtime gut bacteriome, virome, and metabolome of 55 patients treated with azithromycin or a placebo. We describe four enterotypes and the network of associated bacteriophage species and metabolic pathways. One enterotype associates with sustained remission. One taxon from Bacteroides specifically associates with relapse, while two from Bacteroides and Prevotella correlate with complete remission. These taxa are associated with lipid, pentose, and branched-chain amino acid metabolic pathways and several bacteriophage species. Enterotypes and taxa associate with exhausted T cells and the functional status of circulating immune cells. These results illustrate how an antibiotic influences a complex network of gut bacteria, viruses, and metabolites and may promote cancer relapse through modifications of immune cells.
Collapse
Affiliation(s)
- Nicolas Vallet
- Université de Paris Cité, INSERM U976, 75010 Paris, France
| | - Maud Salmona
- Université de Paris Cité, INSERM U976, 75010 Paris, France; Virology Department, AP-HP, Saint-Louis Hospital, 75010 Paris, France
| | - Jeanne Malet-Villemagne
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, Domaine de Vilvert, 78350 Jouy-en-Josas, France
| | - Maxime Bredel
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, Domaine de Vilvert, 78350 Jouy-en-Josas, France
| | - Louise Bondeelle
- Pneumology Unit, AP-HP, Saint-Louis Hospital, 75010 Paris, France
| | - Simon Tournier
- Core Facilities, Saint-Louis Research Institute, Université de Paris Cité, UAR 2030/US 53, 75010 Paris, France
| | | | - Stéphane Cassonnet
- Service de Biostatistique et Information Médicale, AP-HP, Saint-Louis Hospital, 75010 Paris, France
| | | | - Régis Peffault de Latour
- Hematology Transplantation, AP-HP, Saint-Louis Hospital, 1 avenue Claude Vellefaux, 75010 Paris, France; Cryostem Consortium, 13382 Marseille, France
| | - Anne Bergeron
- Pneumology Department, Geneva University Hospitals, 1205 Geneva, Switzerland
| | - Gérard Socié
- Université de Paris Cité, INSERM U976, 75010 Paris, France; Hematology Transplantation, AP-HP, Saint-Louis Hospital, 1 avenue Claude Vellefaux, 75010 Paris, France
| | - Jérome Le Goff
- Université de Paris Cité, INSERM U976, 75010 Paris, France; Virology Department, AP-HP, Saint-Louis Hospital, 75010 Paris, France
| | - Patricia Lepage
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, Domaine de Vilvert, 78350 Jouy-en-Josas, France
| | - David Michonneau
- Université de Paris Cité, INSERM U976, 75010 Paris, France; Hematology Transplantation, AP-HP, Saint-Louis Hospital, 1 avenue Claude Vellefaux, 75010 Paris, France.
| |
Collapse
|
53
|
Zhou H, Jia B, Annageldiyev C, Minagawa K, Zhao C, Mineishi S, Ehmann WC, Naik SG, Cioccio J, Wirk B, Songdej N, Rakszawski KL, Nickolich MS, Shen J, Zheng H. CD26 lowPD-1 + CD8 T cells are terminally exhausted and associated with leukemia progression in acute myeloid leukemia. Front Immunol 2023; 14:1169144. [PMID: 37457737 PMCID: PMC10338956 DOI: 10.3389/fimmu.2023.1169144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2023] [Accepted: 06/15/2023] [Indexed: 07/18/2023] Open
Abstract
Acute myeloid leukemia (AML) is a devastating blood cancer with poor prognosis. Novel effective treatment is an urgent unmet need. Immunotherapy targeting T cell exhaustion by blocking inhibitory pathways, such as PD-1, is promising in cancer treatment. However, results from clinical studies applying PD-1 blockade to AML patients are largely disappointing. AML is highly heterogeneous. Identification of additional immune regulatory pathways and defining predictive biomarkers for treatment response are crucial to optimize the strategy. CD26 is a marker of T cell activation and involved in multiple immune processes. Here, we performed comprehensive phenotypic and functional analyses on the blood samples collected from AML patients and discovered that CD26lowPD-1+ CD8 T cells were associated with AML progression. Specifically, the percentage of this cell fraction was significantly higher in patients with newly diagnosed AML compared to that in patients achieved completed remission or healthy controls. Our subsequent studies on CD26lowPD-1+ CD8 T cells from AML patients at initial diagnosis demonstrated that this cell population highly expressed inhibitory receptors and displayed impaired cytokine production, indicating an exhaustion status. Importantly, CD26lowPD-1+ CD8 T cells carried features of terminal exhaustion, manifested by higher frequency of TEMRA differentiation, increased expression of transcription factors that are observed in terminally exhausted T cells, and high level of intracellular expression of granzyme B and perforin. Our findings suggest a prognostic and predictive value of CD26 in AML, providing pivotal information to optimize the immunotherapy for this devastating cancer.
Collapse
Affiliation(s)
- Huarong Zhou
- Penn State Cancer Institute, Penn State University College of Medicine, Hershey, PA, United States
- Fujian Institute of Hematology, Fujian Provincial Key Laboratory of Hematology, Fujian Medical University Union Hospital, Fujian Medical Center of Hematology, Fuzhou, China
| | - Bei Jia
- Penn State Cancer Institute, Penn State University College of Medicine, Hershey, PA, United States
| | - Charyguly Annageldiyev
- Penn State Cancer Institute, Penn State University College of Medicine, Hershey, PA, United States
| | - Kentaro Minagawa
- Penn State Cancer Institute, Penn State University College of Medicine, Hershey, PA, United States
| | - Chenchen Zhao
- Penn State Cancer Institute, Penn State University College of Medicine, Hershey, PA, United States
| | - Shin Mineishi
- Penn State Cancer Institute, Penn State University College of Medicine, Hershey, PA, United States
| | - W Christopher Ehmann
- Penn State Cancer Institute, Penn State University College of Medicine, Hershey, PA, United States
| | - Seema G. Naik
- Penn State Cancer Institute, Penn State University College of Medicine, Hershey, PA, United States
| | - Joseph Cioccio
- Penn State Cancer Institute, Penn State University College of Medicine, Hershey, PA, United States
| | - Baldeep Wirk
- Penn State Cancer Institute, Penn State University College of Medicine, Hershey, PA, United States
| | - Natthapol Songdej
- Penn State Cancer Institute, Penn State University College of Medicine, Hershey, PA, United States
| | - Kevin L. Rakszawski
- Penn State Cancer Institute, Penn State University College of Medicine, Hershey, PA, United States
| | - Myles S. Nickolich
- Penn State Cancer Institute, Penn State University College of Medicine, Hershey, PA, United States
| | - Jianzhen Shen
- Fujian Institute of Hematology, Fujian Provincial Key Laboratory of Hematology, Fujian Medical University Union Hospital, Fujian Medical Center of Hematology, Fuzhou, China
| | - Hong Zheng
- Penn State Cancer Institute, Penn State University College of Medicine, Hershey, PA, United States
- Department of Microbiology and Immunology, Penn State University College of Medicine, Hershey, PA, United States
| |
Collapse
|
54
|
Damiani D, Tiribelli M. Checkpoint Inhibitors in Acute Myeloid Leukemia. Biomedicines 2023; 11:1724. [PMID: 37371818 PMCID: PMC10295997 DOI: 10.3390/biomedicines11061724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2023] [Revised: 06/09/2023] [Accepted: 06/13/2023] [Indexed: 06/29/2023] Open
Abstract
The prognosis of acute myeloid leukemia (AML) remains unsatisfactory. Among the reasons for the poor response to therapy and high incidence of relapse, there is tumor cell immune escape, as AML blasts can negatively influence various components of the immune system, mostly weakening T-cells. Since leukemic cells can dysregulate immune checkpoints (ICs), receptor-based signal transductors that lead to the negative regulation of T-cells and, eventually, to immune surveillance escape, the inhibition of ICs is a promising therapeutic strategy and has led to the development of so-called immune checkpoint inhibitors (ICIs). ICIs, in combination with conventional chemotherapy, hypomethylating agents or targeted therapies, are being increasingly tested in cases of AML, but the results reported are often conflicting. Here, we review the main issues concerning the immune system in AML, the main pathways leading to immune escape and the results obtained from clinical trials of ICIs, alone or in combination, in newly diagnosed or relapsed/refractory AML.
Collapse
Affiliation(s)
- Daniela Damiani
- Division of Hematology and Stem Cell Transplantation, Udine Hospital, 33100 Udine, Italy;
- Department of Medicine, Udine University, 33100 Udine, Italy
| | - Mario Tiribelli
- Division of Hematology and Stem Cell Transplantation, Udine Hospital, 33100 Udine, Italy;
- Department of Medicine, Udine University, 33100 Udine, Italy
| |
Collapse
|
55
|
Apostolova P. Allo-HCT: damaged T cells don't bite. Blood 2023; 141:1507-1508. [PMID: 36995707 DOI: 10.1182/blood.2022019242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/31/2023] Open
|
56
|
Bellone M, Brevi A, Bronte V, Dusi S, Ferrucci PF, Nisticò P, Rosato A, Russo V, Sica A, Toietta G, Colombo MP. Cancer immunity and immunotherapy beyond COVID-19. Cancer Immunol Immunother 2023:10.1007/s00262-023-03411-9. [PMID: 36995489 PMCID: PMC10061391 DOI: 10.1007/s00262-023-03411-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Accepted: 02/12/2023] [Indexed: 03/31/2023]
Affiliation(s)
- Matteo Bellone
- Unit of Cellular Immunology, I.R.C.C.S. Ospedale San Raffaele, Milan, Italy.
| | - Arianna Brevi
- Unit of Cellular Immunology, I.R.C.C.S. Ospedale San Raffaele, Milan, Italy
| | - Vincenzo Bronte
- Immunology Section, Department of Medicine, University and Hospital Trust of Verona, Verona, Italy
| | - Silvia Dusi
- Immunology Section, Department of Medicine, University and Hospital Trust of Verona, Verona, Italy
| | - Pier Francesco Ferrucci
- Unit of Tumor Biotherapy, Department of Experimental Oncology, I.R.C.C.S. European Institute of Oncology, Milan, Italy
| | - Paola Nisticò
- Unit Tumor Immunology and Immunotherapy, I.R.C.C.S. Regina Elena National Cancer Institute, Rome, Italy
| | - Antonio Rosato
- Department of Surgery, Oncology and Gastroenterology, University of Padova, Padua, Italy
- Veneto Institute of Oncology IOV-I.R.C.C.S, Padua, Italy
| | - Vincenzo Russo
- Unit of Immuno-Biotherapy of Melanoma and Solid Tumors, I.R.C.C.S. Ospedale San Raffaele, Milan, Italy
| | - Antonio Sica
- Molecular Immunology Lab, I.R.C.C.S. Humanitas Clinical and Research Center, Rozzano (Mi), Italy
- Department of Pharmaceutical Sciences, University of Piemonte Orientale "A. Avogadro", Novara, Italy
| | - Gabriele Toietta
- Unit Tumor Immunology and Immunotherapy, I.R.C.C.S. Regina Elena National Cancer Institute, Rome, Italy
| | - Mario Paolo Colombo
- Molecular Immunology Unit, Department of Research, Fondazione I.R.C.C.S. Istituto Nazionale Dei Tumori, Via Amadeo 42, 20068, Milan, Italy.
| |
Collapse
|
57
|
Romine KA, MacPherson K, Cho HJ, Kosaka Y, Flynn PA, Byrd KH, Coy JL, Newman MT, Pandita R, Loo CP, Scott J, Adey AC, Lind EF. BET inhibitors rescue anti-PD1 resistance by enhancing TCF7 accessibility in leukemia-derived terminally exhausted CD8 + T cells. Leukemia 2023; 37:580-592. [PMID: 36681742 PMCID: PMC9991923 DOI: 10.1038/s41375-023-01808-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 12/08/2022] [Accepted: 01/04/2023] [Indexed: 01/22/2023]
Abstract
Many acute myeloid leukemia (AML) patients exhibit hallmarks of immune exhaustion, such as increased myeloid-derived suppressor cells, suppressive regulatory T cells and dysfunctional T cells. Similarly, we have identified the same immune-related features, including exhausted CD8+ T cells (TEx) in a mouse model of AML. Here we show that inhibitors that target bromodomain and extra-terminal domain (BET) proteins affect tumor-intrinsic factors but also rescue T cell exhaustion and ICB resistance. Ex vivo treatment of cells from AML mice and AML patients with BET inhibitors (BETi) reversed CD8+ T cell exhaustion by restoring proliferative capacity and expansion of the more functional precursor-exhausted T cells. This reversal was enhanced by combined BETi and anti-PD1 treatment. BETi synergized with anti-PD1 in vivo, resulting in the reduction of circulating leukemia cells, enrichment of CD8+ T cells in the bone marrow, and increase in expression of Tcf7, Slamf6, and Cxcr5 in CD8+ T cells. Finally, we profiled the epigenomes of in vivo JQ1-treated AML-derived CD8+ T cells by single-cell ATAC-seq and found that JQ1 increases Tcf7 accessibility specifically in Tex cells, suggesting that BETi likely acts mechanistically by relieving repression of progenitor programs in Tex CD8+ T cells and maintaining a pool of anti-PD1 responsive CD8+ T cells.
Collapse
Affiliation(s)
- Kyle A Romine
- Department of Cell, Developmental & Cancer Biology, Oregon Health & Science University, Portland, OR, USA
| | - Kevin MacPherson
- Department of Molecular & Medical Genetics, Oregon Health & Science University, Portland, OR, USA
| | - Hyun-Jun Cho
- Department of Molecular Microbiology and Immunology, Oregon Health & Science University, Portland, OR, USA
| | - Yoko Kosaka
- Department of Molecular Microbiology and Immunology, Oregon Health & Science University, Portland, OR, USA
| | - Patrick A Flynn
- Department of Molecular Microbiology and Immunology, Oregon Health & Science University, Portland, OR, USA
| | - Kaelan H Byrd
- Department of Molecular Microbiology and Immunology, Oregon Health & Science University, Portland, OR, USA
| | - Jesse L Coy
- Department of Molecular Microbiology and Immunology, Oregon Health & Science University, Portland, OR, USA
| | - Matthew T Newman
- School of Medicine, Oregon Health & Science University, Portland, OR, USA
| | - Ravina Pandita
- Department of Cell, Developmental & Cancer Biology, Oregon Health & Science University, Portland, OR, USA
| | - Christopher P Loo
- Department of Molecular Microbiology and Immunology, Oregon Health & Science University, Portland, OR, USA
| | - Jaime Scott
- Department of Molecular Microbiology and Immunology, Oregon Health & Science University, Portland, OR, USA
| | - Andrew C Adey
- Department of Molecular & Medical Genetics, Oregon Health & Science University, Portland, OR, USA
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA
- Knight Cardiovascular Institute, Oregon Health & Science University, Portland, OR, USA
- Center for Early Detection Advanced Research, Oregon Health & Science University, Portland, OR, USA
| | - Evan F Lind
- Department of Cell, Developmental & Cancer Biology, Oregon Health & Science University, Portland, OR, USA.
- Department of Molecular Microbiology and Immunology, Oregon Health & Science University, Portland, OR, USA.
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA.
| |
Collapse
|
58
|
Xia Y, An J, Li J, Gu W, Zhang Y, Zhao S, Zhao C, Xu Y, Li B, Zhong Z, Meng F. Transferrin-guided intelligent nanovesicles augment the targetability and potency of clinical PLK1 inhibitor to acute myeloid leukemia. Bioact Mater 2023; 21:499-510. [PMID: 36185744 PMCID: PMC9494038 DOI: 10.1016/j.bioactmat.2022.08.032] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 08/24/2022] [Accepted: 08/28/2022] [Indexed: 11/28/2022] Open
Abstract
Acute myeloid leukemia (AML) remains a most lethal hematological malignancy, partly because of its slow development of targeted therapies compared with other cancers. PLK1 inhibitor, volasertib (Vol), is among the few molecular targeted drugs granted breakthrough therapy status for AML; however, its fast clearance and dose-limiting toxicity greatly restrain its clinical benefits. Here, we report that transferrin-guided polymersomes (TPs) markedly augment the targetability, potency and safety of Vol to AML. Vol-loaded TPs (TPVol) with 4% transferrin exhibited best cellular uptake, effective down-regulation of p-PLK1, p-PTEN and p-AKT and superior apoptotic activity to free Vol in MV-4-11 leukemic cells. Intravenous injection of TPVol gave 6-fold higher AUC than free Vol and notable accumulation in AML-residing bone marrow. The efficacy studies in orthotopic MV-4-11 leukemic model demonstrated that TPVol significantly reduced leukemic cell proportions in periphery blood, bone marrow, liver and spleen, effectively enhanced mouse survival rate, and impeded bone loss. This transferrin-guided nano-delivery of molecular targeted drugs appears to be an interesting strategy towards the development of novel treatments for AML.
Collapse
Affiliation(s)
- Yifeng Xia
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, 215123, PR China
- College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, PR China
| | - Jingnan An
- Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Institute of Blood and Marrow Transplantation of Soochow University, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, 215123, PR China
| | - Jiaying Li
- Orthopedic Institute, Soochow University, Suzhou, 215007, PR China
| | - Wenxing Gu
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, 215123, PR China
| | - Yifan Zhang
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, 215123, PR China
| | - Songsong Zhao
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, 215123, PR China
| | - Cenzhu Zhao
- Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Institute of Blood and Marrow Transplantation of Soochow University, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, 215123, PR China
| | - Yang Xu
- Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Institute of Blood and Marrow Transplantation of Soochow University, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, 215123, PR China
| | - Bin Li
- Orthopedic Institute, Soochow University, Suzhou, 215007, PR China
| | - Zhiyuan Zhong
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, 215123, PR China
- College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, PR China
| | - Fenghua Meng
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, 215123, PR China
- College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, PR China
| |
Collapse
|
59
|
Immune Checkpoints and targeted agents in relapse and graft-versus-host disease after hematopoietic stem cell transplantation. Mol Biol Rep 2023; 50:2909-2917. [PMID: 36572760 DOI: 10.1007/s11033-022-08220-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Accepted: 12/16/2022] [Indexed: 12/27/2022]
Abstract
BACKGROUND Allogeneic hematopoietic stem cell transplantation (HSCT) is a potentially curative therapy for malignant hematologic disorders. Novel anti-infection agents have successfully decreased the risk of fatal infections post-HSCT in recent years, but the relapse of primary disease and graft-versus-host disease (GVHD) remain the major causes of death for transplant recipients, and significantly deteriorate the quality of life. Thus, it is crucial to maintain the immune homeostasis in transplant recipients and balance the graft-versus-leukemia (GVL) effect and GVHD. METHODS We reviewed the recently published literatures on immune checkpoint (IC) and targeted agents in relapse and GVHD after allogeneic HSCT RESULTS: Emerging data suggest that IC is an attractive target to modulate immune responses, and accumulating evidences of IC-targeted agents have been published for the treatment of malignancies and autoimmune disorders. The unique mechanism of IC-targeted agents, which affects the immune homeostasis of the transplant recipient by modulating alloreactivity, minimizes the risk of organ toxicity and immunosuppression associated with conventional therapy CONCLUSION: There is an increase in literature reporting the application of immune checkpoint-targeted agents in HSCT settings, and an overview will benefit further exploration in this field.
Collapse
|
60
|
Minnie SA, Waltner OG, Ensbey KS, Olver SD, Collinge AD, Sester DP, Schmidt CR, Legg SR, Takahashi S, Nemychenkov NS, Sekiguchi T, Driessens G, Zhang P, Koyama M, Spencer A, Holmberg LA, Furlan SN, Varelias A, Hill GR. TIGIT inhibition and lenalidomide synergistically promote antimyeloma immune responses after stem cell transplantation in mice. J Clin Invest 2023; 133:e157907. [PMID: 36512425 PMCID: PMC9927935 DOI: 10.1172/jci157907] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Accepted: 12/08/2022] [Indexed: 12/15/2022] Open
Abstract
Autologous stem cell transplantation (ASCT) with subsequent lenalidomide maintenance is standard consolidation therapy for multiple myeloma, and a subset of patients achieve durable progression-free survival that is suggestive of long-term immune control. Nonetheless, most patients ultimately relapse, suggesting immune escape. TIGIT appears to be a potent inhibitor of myeloma-specific immunity and represents a promising new checkpoint target. Here we demonstrate high expression of TIGIT on activated CD8+ T cells in mobilized peripheral blood stem cell grafts from patients with myeloma. To guide clinical application of TIGIT inhibition, we evaluated identical anti-TIGIT antibodies that do or do not engage FcγR and demonstrated that anti-TIGIT activity is dependent on FcγR binding. We subsequently used CRBN mice to investigate the efficacy of anti-TIGIT in combination with lenalidomide maintenance after transplantation. Notably, the combination of anti-TIGIT with lenalidomide provided synergistic, CD8+ T cell-dependent, antimyeloma efficacy. Analysis of bone marrow (BM) CD8+ T cells demonstrated that combination therapy suppressed T cell exhaustion, enhanced effector function, and expanded central memory subsets. Importantly, these immune phenotypes were specific to the BM tumor microenvironment. Collectively, these data provide a logical rationale for combining TIGIT inhibition with immunomodulatory drugs to prevent myeloma progression after ASCT.
Collapse
Affiliation(s)
- Simone A. Minnie
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Olivia G. Waltner
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Kathleen S. Ensbey
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Stuart D. Olver
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Alika D. Collinge
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - David P. Sester
- Translational Research Institute, Woolloongabba, Queensland, Australia
- Hugh Green Cytometry Centre, Malaghan Institute of Medical Research, Wellington, New Zealand
| | - Christine R. Schmidt
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Samuel R.W. Legg
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Shuichiro Takahashi
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | | | - Tomoko Sekiguchi
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | | | - Ping Zhang
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Motoko Koyama
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Andrew Spencer
- Australian Center for Blood Diseases, Monash University and
- Malignant Haematology and Stem Cell Transplantation, The Alfred Hospital, Melbourne, Victoria, Australia
- Department of Clinical Haematology, Monash University, Melbourne, Victoria, Australia
| | - Leona A. Holmberg
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
- Division of Medical Oncology and
| | - Scott N. Furlan
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
- Department of Pediatrics, University of Washington, Seattle, Washington, USA
| | - Antiopi Varelias
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
- Faculty of Medicine, The University of Queensland, St. Lucia, Queensland, Australia
| | - Geoffrey R. Hill
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
- Division of Medical Oncology and
| |
Collapse
|
61
|
Fuchs YF, Brunner J, Weigelt M, Schieferdecker A, Morgenstern R, Sturm A, Winter B, Jambor H, Stölzel F, Ruhnke L, von Bonin M, Rücker-Braun E, Heidenreich F, Fuchs A, Bonifacio E, Bornhäuser M, Poitz DM, Altmann H. Next Generation Biobanking: Employing a Robotic System for Automated Mononuclear Cell Isolation. Biopreserv Biobank 2023; 21:106-110. [PMID: 36251308 PMCID: PMC9963478 DOI: 10.1089/bio.2021.0181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
- Yannick F. Fuchs
- Center for Regenerative Therapies Dresden, Technische Universität Dresden, Dresden, Germany
| | - Jonathan Brunner
- Medical Department I, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Marc Weigelt
- Center for Regenerative Therapies Dresden, Technische Universität Dresden, Dresden, Germany
| | - Anja Schieferdecker
- National Center for Tumor Diseases (NCT), Partner Site Dresden, Germany and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Robert Morgenstern
- Center for Regenerative Therapies Dresden, Technische Universität Dresden, Dresden, Germany
| | - Andrea Sturm
- National Center for Tumor Diseases (NCT), Partner Site Dresden, Germany and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | | | - Helena Jambor
- Mildred Scheel Early Career Center, Medical Faculty, Technische Universität Dresden, Dresden, Germany
| | - Friedrich Stölzel
- Medical Department I, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Leo Ruhnke
- Medical Department I, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Malte von Bonin
- Medical Department I, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Elke Rücker-Braun
- Medical Department I, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany.,Clinical Trials Unit, DKMS gGmbH, Dresden, Germany
| | - Falk Heidenreich
- Medical Department I, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany.,Clinical Trials Unit, DKMS gGmbH, Dresden, Germany
| | - Anke Fuchs
- Center for Regenerative Therapies Dresden, Technische Universität Dresden, Dresden, Germany.,Mildred Scheel Early Career Center, Medical Faculty, Technische Universität Dresden, Dresden, Germany
| | - Ezio Bonifacio
- Center for Regenerative Therapies Dresden, Technische Universität Dresden, Dresden, Germany
| | - Martin Bornhäuser
- Medical Department I, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany.,National Center for Tumor Diseases (NCT), Partner Site Dresden, Germany and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - David M. Poitz
- Institute for Clinical Chemistry and Laboratory Medicine, University Hospital and Faculty of Medicine Carl Gustav Carus of TU Dresden, Dresden, Germany
| | - Heidi Altmann
- Medical Department I, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany.,National Center for Tumor Diseases (NCT), Partner Site Dresden, Germany and German Cancer Research Center (DKFZ), Heidelberg, Germany.,Address correspondence to: Heidi Altmann, PhD, Medical Department I, University Hospital Carl Gustav Carus, Technische Universität Dresden, Fetscherstr. 74, Dresden 01307, Germany
| |
Collapse
|
62
|
Aru B, Pehlivanoğlu C, Dal Z, Dereli-Çalışkan NN, Gürlü E, Yanıkkaya-Demirel G. A potential area of use for immune checkpoint inhibitors: Targeting bone marrow microenvironment in acute myeloid leukemia. Front Immunol 2023; 14:1108200. [PMID: 36742324 PMCID: PMC9895857 DOI: 10.3389/fimmu.2023.1108200] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Accepted: 01/03/2023] [Indexed: 01/22/2023] Open
Abstract
Acute myeloid leukemia (AML) arises from the cells of myeloid lineage and is the most frequent leukemia type in adulthood accounting for about 80% of all cases. The most common treatment strategy for the treatment of AML includes chemotherapy, in rare cases radiotherapy and stem cell and bone marrow transplantation are considered. Immune checkpoint proteins involve in the negative regulation of immune cells, leading to an escape from immune surveillance, in turn, causing failure of tumor cell elimination. Immune checkpoint inhibitors (ICIs) target the negative regulation of the immune cells and support the immune system in terms of anti-tumor immunity. Bone marrow microenvironment (BMM) bears various blood cell lineages and the interactions between these lineages and the noncellular components of BMM are considered important for AML development and progression. Administration of ICIs for the AML treatment may be a promising option by regulating BMM. In this review, we summarize the current treatment options in AML treatment and discuss the possible application of ICIs in AML treatment from the perspective of the regulation of BMM.
Collapse
Affiliation(s)
- Başak Aru
- Immunology Department, Faculty of Medicine, Yeditepe University, Istanbul, Türkiye
| | - Cemil Pehlivanoğlu
- Immunology Department, Faculty of Medicine, Yeditepe University, Istanbul, Türkiye
| | - Zeynep Dal
- School of Medicine, Yeditepe University, Istanbul, Türkiye
| | | | - Ege Gürlü
- School of Medicine, Yeditepe University, Istanbul, Türkiye
| | - Gülderen Yanıkkaya-Demirel
- Immunology Department, Faculty of Medicine, Yeditepe University, Istanbul, Türkiye,*Correspondence: Gülderen Yanıkkaya-Demirel,
| |
Collapse
|
63
|
Serroukh Y, Hébert J, Busque L, Mercier F, Rudd CE, Assouline S, Lachance S, Delisle JS. Blasts in context: the impact of the immune environment on acute myeloid leukemia prognosis and treatment. Blood Rev 2023; 57:100991. [PMID: 35941029 DOI: 10.1016/j.blre.2022.100991] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 06/22/2022] [Accepted: 07/13/2022] [Indexed: 01/28/2023]
Abstract
Acute myeloid leukemia (AML) is a cancer that originates from the bone marrow (BM). Under physiological conditions, the bone marrow supports the homeostasis of immune cells and hosts memory lymphoid cells. In this review, we summarize our present understanding of the role of the immune microenvironment on healthy bone marrow and on the development of AML, with a focus on T cells and other lymphoid cells. The types and function of different immune cells involved in the AML microenvironment as well as their putative role in the onset of disease and response to treatment are presented. We also describe how the immune context predicts the response to immunotherapy in AML and how these therapies modulate the immune status of the bone marrow. Finally, we focus on allogeneic stem cell transplantation and summarize the current understanding of the immune environment in the post-transplant bone marrow, the factors associated with immune escape and relevant strategies to prevent and treat relapse.
Collapse
Affiliation(s)
- Yasmina Serroukh
- Centre de recherche de l'Hôpital Maisonneuve-Rosemont, 5415 Boul. de L'Assomption, Montréal, Canada; Erasmus Medical center Cancer Institute, University Medical Center Rotterdam, Department of Hematology, Rotterdam, the Netherlands; Department of Medicine, Université de Montréal, Montreal, Canada; Institute for Hematology-Oncology, Transplantation, Cell and Gene Therapy, Hôpital Maisonneuve-Rosemont, Montreal, Canada.
| | - Josée Hébert
- Centre de recherche de l'Hôpital Maisonneuve-Rosemont, 5415 Boul. de L'Assomption, Montréal, Canada; Department of Medicine, Université de Montréal, Montreal, Canada; Institute for Hematology-Oncology, Transplantation, Cell and Gene Therapy, Hôpital Maisonneuve-Rosemont, Montreal, Canada; The Quebec Leukemia Cell Bank, Canada
| | - Lambert Busque
- Centre de recherche de l'Hôpital Maisonneuve-Rosemont, 5415 Boul. de L'Assomption, Montréal, Canada; Department of Medicine, Université de Montréal, Montreal, Canada; Institute for Hematology-Oncology, Transplantation, Cell and Gene Therapy, Hôpital Maisonneuve-Rosemont, Montreal, Canada
| | - François Mercier
- Division of Hematology and Experimental Medicine, Department of Medicine, McGill University, 3755 Côte-Sainte-Catherine Road, Montreal, Canada; Lady Davis Institute for Medical Research, Jewish General Hospital, 3755 Côte-Sainte-Catherine Road, Montreal, Canada
| | - Christopher E Rudd
- Centre de recherche de l'Hôpital Maisonneuve-Rosemont, 5415 Boul. de L'Assomption, Montréal, Canada; Department of Medicine, Université de Montréal, Montreal, Canada; Institute for Hematology-Oncology, Transplantation, Cell and Gene Therapy, Hôpital Maisonneuve-Rosemont, Montreal, Canada
| | - Sarit Assouline
- Division of Hematology and Experimental Medicine, Department of Medicine, McGill University, 3755 Côte-Sainte-Catherine Road, Montreal, Canada; Lady Davis Institute for Medical Research, Jewish General Hospital, 3755 Côte-Sainte-Catherine Road, Montreal, Canada
| | - Silvy Lachance
- Department of Medicine, Université de Montréal, Montreal, Canada; Institute for Hematology-Oncology, Transplantation, Cell and Gene Therapy, Hôpital Maisonneuve-Rosemont, Montreal, Canada
| | - Jean-Sébastien Delisle
- Centre de recherche de l'Hôpital Maisonneuve-Rosemont, 5415 Boul. de L'Assomption, Montréal, Canada; Department of Medicine, Université de Montréal, Montreal, Canada; Institute for Hematology-Oncology, Transplantation, Cell and Gene Therapy, Hôpital Maisonneuve-Rosemont, Montreal, Canada
| |
Collapse
|
64
|
Giannotta C, Autino F, Massaia M. Vγ9Vδ2 T-cell immunotherapy in blood cancers: ready for prime time? Front Immunol 2023; 14:1167443. [PMID: 37143664 PMCID: PMC10153673 DOI: 10.3389/fimmu.2023.1167443] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Accepted: 03/31/2023] [Indexed: 05/06/2023] Open
Abstract
In the last years, the tumor microenvironment (TME) has emerged as a promising target for therapeutic interventions in cancer. Cancer cells are highly dependent on the TME to growth and evade the immune system. Three major cell subpopulations are facing each other in the TME: cancer cells, immune suppressor cells, and immune effector cells. These interactions are influenced by the tumor stroma which is composed of extracellular matrix, bystander cells, cytokines, and soluble factors. The TME can be very different depending on the tissue where cancer arises as in solid tumors vs blood cancers. Several studies have shown correlations between the clinical outcome and specific patterns of TME immune cell infiltration. In the recent years, a growing body of evidence suggests that unconventional T cells like natural killer T (NKT) cells, mucosal-associated invariant T (MAIT) cells, and γδ T cells are key players in the protumor or antitumor TME commitment in solid tumors and blood cancers. In this review, we will focus on γδ T cells, especially Vγ9Vδ2 T cells, to discuss their peculiarities, pros, and cons as potential targets of therapeutic interventions in blood cancers.
Collapse
Affiliation(s)
- Claudia Giannotta
- Laboratorio di Immunologia dei Tumori del Sangue (LITS), Centro Interdipartimentale di Biotecnologie Molecolari “Guido Tarone”, Dipartimento di Biotecnologie Molecolari e Scienze per la Salute, Università Degli Studi di Torino, Torino, Italy
| | - Federica Autino
- Laboratorio di Immunologia dei Tumori del Sangue (LITS), Centro Interdipartimentale di Biotecnologie Molecolari “Guido Tarone”, Dipartimento di Biotecnologie Molecolari e Scienze per la Salute, Università Degli Studi di Torino, Torino, Italy
| | - Massimo Massaia
- Laboratorio di Immunologia dei Tumori del Sangue (LITS), Centro Interdipartimentale di Biotecnologie Molecolari “Guido Tarone”, Dipartimento di Biotecnologie Molecolari e Scienze per la Salute, Università Degli Studi di Torino, Torino, Italy
- Struttura Complessa (SC) Ematologia, Azienda Ospedaliera (AO) S. Croce e Carle, Cuneo, Italy
- *Correspondence: Massimo Massaia,
| |
Collapse
|
65
|
Yao D, Lai J, Lu Y, Zhong J, Zha X, Huang X, Liu L, Zeng X, Chen S, Weng J, Du X, Li Y, Xu L. Comprehensive analysis of the immune pattern of T cell subsets in chronic myeloid leukemia before and after TKI treatment. Front Immunol 2023; 14:1078118. [PMID: 36742315 PMCID: PMC9893006 DOI: 10.3389/fimmu.2023.1078118] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Accepted: 01/03/2023] [Indexed: 01/20/2023] Open
Abstract
Background Immunological phenotypes and differentiation statuses commonly decide the T cell function and anti-tumor ability. However, little is known about these alterations in CML patients. Method Here, we investigated the immunologic phenotypes (CD38/CD69/HLA-DR/CD28/CD57/BTLA/TIGIT/PD-1) of T subsets (TN, TCM, TEM, and TEMRA) in peripheral blood (PB) and bone marrow (BM) from de novo CML patients (DN-CML), patients who achieved a molecular response (MR) and those who failed to achieve an MR (TKI-F) after tyrosine kinase inhibitor (TKI) treatment using multicolor flow cytometry. Results CD38 or HLA-DR positive PB CD8+TN and TCM cells decreased in the DN-CML patients and this was further decreased in TKI-F patients. Meanwhile, the level of PD-1 elevated in CD8+ TEM and TEMRA cells from PB in all groups. Among BM sample, the level of HLA-DR+CD8+TCM cells significantly decreased in all groups and CD8+TEMRA cells from TKI-F patients exhibited increased level of TIGIT and CD8+ tissue-residual T cells (TRM) from DN-CML patients expressed a higher level of PD-1 and TIGIT. Lastly, we found a significantly decreased proportion of CD86+ dendritic cells (DCs) and an imbalanced CD80/CD86 in the PB and BM of DN-CML patients, which may impair the activation of T cells. Conclusion In summary, early differentiated TN and TCM cells from CML patients may remain in an inadequate activation state, particularly for TKI-F patients. And effector T cells (TEM, TEMRA and TRM) may be dysfunctional due to the expression of PD-1 and TIGIT in CML patients. Meanwhile, DCs cells exhibited the impairment of costimulatory molecule expression in DN-CML patients. Those factors may jointly contribute to the immune escape in CML patients.
Collapse
Affiliation(s)
- Danlin Yao
- Key Laboratory for Regenerative Medicine of Ministry of Education, Institute of Hematology, School of Medicine, Jinan University, Guangzhou, China.,Department of Hematology, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Jing Lai
- Department of Hematology, First Affiliated Hospital, Jinan University, Guangzhou, China
| | - Yuhong Lu
- Department of Hematology, First Affiliated Hospital, Jinan University, Guangzhou, China
| | - Jun Zhong
- Department of Hematology, First Affiliated Hospital, Jinan University, Guangzhou, China
| | - Xianfeng Zha
- Department of Clinical Laboratory, First Affiliated Hospital, Jinan University, Guangzhou, China
| | - Xin Huang
- Department of Hematology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Lian Liu
- Key Laboratory for Regenerative Medicine of Ministry of Education, Institute of Hematology, School of Medicine, Jinan University, Guangzhou, China
| | - Xiangbo Zeng
- Key Laboratory for Regenerative Medicine of Ministry of Education, Institute of Hematology, School of Medicine, Jinan University, Guangzhou, China
| | - Shaohua Chen
- Key Laboratory for Regenerative Medicine of Ministry of Education, Institute of Hematology, School of Medicine, Jinan University, Guangzhou, China
| | - Jianyu Weng
- Department of Hematology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Xin Du
- Department of Hematology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Yangqiu Li
- Key Laboratory for Regenerative Medicine of Ministry of Education, Institute of Hematology, School of Medicine, Jinan University, Guangzhou, China
| | - Ling Xu
- Key Laboratory for Regenerative Medicine of Ministry of Education, Institute of Hematology, School of Medicine, Jinan University, Guangzhou, China
| |
Collapse
|
66
|
Present and Future Role of Immune Targets in Acute Myeloid Leukemia. Cancers (Basel) 2022; 15:cancers15010253. [PMID: 36612249 PMCID: PMC9818182 DOI: 10.3390/cancers15010253] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 12/20/2022] [Accepted: 12/28/2022] [Indexed: 01/03/2023] Open
Abstract
It is now well known that the bone marrow (BM) cell niche contributes to leukemogenesis, but emerging data support the role of the complex crosstalk between AML cells and the BM microenvironment to induce a permissive immune setting that protects leukemic stem cells (LSCs) from therapy-induced death, thus favoring disease persistence and eventual relapse. The identification of potential immune targets on AML cells and the modulation of the BM environment could lead to enhanced anti-leukemic effects of drugs, immune system reactivation, and the restoration of AML surveillance. Potential targets and effectors of this immune-based therapy could be monoclonal antibodies directed against LSC antigens such as CD33, CD123, and CLL-1 (either as direct targets or via several bispecific T-cell engagers), immune checkpoint inhibitors acting on different co-inhibitory axes (alone or in combination with conventional AML drugs), and novel cellular therapies such as chimeric antigen receptor (CAR) T-cells designed against AML-specific antigens. Though dozens of clinical trials, mostly in phases I and II, are ongoing worldwide, results have still been negatively affected by difficulties in the identification of the optimal targets on LSCs.
Collapse
|
67
|
PD-1 Checkpoint Blockade in Patients for Acute Myeloid Leukemia after HSCT Relapse Resulted in Severe GVHD and sHLH. Case Rep Hematol 2022; 2022:1705905. [PMID: 36590870 PMCID: PMC9800091 DOI: 10.1155/2022/1705905] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Revised: 11/08/2022] [Accepted: 11/28/2022] [Indexed: 12/24/2022] Open
Abstract
Treatment with immune checkpoint inhibitors (ICI) such as carrizumab leads to immune-mediated adverse effects including severe acute graft versus host disease (aGVHD) and secondary hemophagocytic syndrome (sHLH). Herein, we present two cases where aGVHD and sHLH developed after ICI administration, which was treated using methylprednisolone (MP). They developed high-grade fever complicated with liver dysfunction and diarrhea 1 day after ICI administration. Treatment with MP does not alleviate symptoms because of steroid resistance. Hyperbilirubinemia, rash with blisters, and watery diarrhea showed severe aGVHD. Hyperferritinemia, hypertriglyceridemia, and cytopenias suggested the diagnosis of HLH and met the criteria for sHLH diagnosis. They were thus administered intravenous high-dose MP, methotrexate (MTX), basiliximab, ruxolitinib, etc, which resolved these symptoms.
Collapse
|
68
|
Giannotta C, Castella B, Tripoli E, Grimaldi D, Avonto I, D’Agostino M, Larocca A, Kopecka J, Grasso M, Riganti C, Massaia M. Immune dysfunctions affecting bone marrow Vγ9Vδ2 T cells in multiple myeloma: Role of immune checkpoints and disease status. Front Immunol 2022; 13:1073227. [PMID: 36605214 PMCID: PMC9808386 DOI: 10.3389/fimmu.2022.1073227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Accepted: 11/29/2022] [Indexed: 12/24/2022] Open
Abstract
Introduction Bone marrow (BM) Vγ9Vδ2 T cells are intrinsically predisposed to sense the immune fitness of the tumor microenvironment (TME) in multiple myeloma (MM) and monoclonal gammopathy of undetermined significance (MGUS). Methods In this work, we have used BM Vγ9Vδ2 T cells to interrogate the role of the immune checkpoint/immune checkpoint-ligand (ICP/ICP-L) network in the immune suppressive TME of MM patients. Results PD-1+ BM MM Vγ9Vδ2 T cells combine phenotypic, functional, and TCR-associated alterations consistent with chronic exhaustion and immune senescence. When challenged by zoledronic acid (ZA) as a surrogate assay to interrogate the reactivity to their natural ligands, BM MM Vγ9Vδ2 T cells further up-regulate PD-1 and TIM-3 and worsen TCR-associated alterations. BM MM Vγ9Vδ2 T cells up-regulate TIM-3 after stimulation with ZA in combination with αPD-1, whereas PD-1 is not up-regulated after ZA stimulation with αTIM-3, indicating a hierarchical regulation of inducible ICP expression. Dual αPD-1/αTIM-3 blockade improves the immune functions of BM Vγ9Vδ2 T cells in MM at diagnosis (MM-dia), whereas single PD-1 blockade is sufficient to rescue BM Vγ9Vδ2 T cells in MM in remission (MM-rem). By contrast, ZA stimulation induces LAG-3 up-regulation in BM Vγ9Vδ2 T cells from MM in relapse (MM-rel) and dual PD-1/LAG-3 blockade is the most effective combination in this setting. Discussion These data indicate that: 1) inappropriate immune interventions can exacerbate Vγ9Vδ2 T-cell dysfunction 2) ICP blockade should be tailored to the disease status to get the most of its beneficial effect.
Collapse
Affiliation(s)
- Claudia Giannotta
- Laboratorio di Immunologia dei Tumori del Sangue (LITS), Centro Interdipartimentale di Biotecnologie Molecolari “Guido Tarone”, Dipartimento di Biotecnologie Molecolari e Scienze della Salute, Università degli Studi di Torino, Torino, Italy
| | - Barbara Castella
- Laboratorio di Immunologia dei Tumori del Sangue (LITS), Centro Interdipartimentale di Biotecnologie Molecolari “Guido Tarone”, Dipartimento di Biotecnologie Molecolari e Scienze della Salute, Università degli Studi di Torino, Torino, Italy,Struttura Complessa (SC) Ematologia, Azienda Ospedaliera (AO) S.Croce e Carle, Cuneo, Italy
| | - Ezio Tripoli
- Laboratorio di Immunologia dei Tumori del Sangue (LITS), Centro Interdipartimentale di Biotecnologie Molecolari “Guido Tarone”, Dipartimento di Biotecnologie Molecolari e Scienze della Salute, Università degli Studi di Torino, Torino, Italy,Struttura Complessa (SC) Ematologia, Azienda Ospedaliera (AO) S.Croce e Carle, Cuneo, Italy
| | - Daniele Grimaldi
- Struttura Complessa (SC) Ematologia, Azienda Ospedaliera (AO) S.Croce e Carle, Cuneo, Italy
| | - Ilaria Avonto
- Servizio Interdipartimentale di Immunoematologia e Medicina Trasfusionale, Azienda Ospedaliera (AO) S.Croce e Carle, Cuneo, Italy
| | - Mattia D’Agostino
- Struttura Complessa (SC) Ematologia, Azienda Ospedaliero-Universitaria (AOU) Città della Salute e della Scienza di Torino, Torino, Italy
| | - Alessandra Larocca
- Struttura Complessa (SC) Ematologia, Azienda Ospedaliero-Universitaria (AOU) Città della Salute e della Scienza di Torino, Torino, Italy
| | - Joanna Kopecka
- Dipartimento di Oncologia, Università degli Studi di Torino, Torino, Italy
| | - Mariella Grasso
- Struttura Complessa (SC) Ematologia, Azienda Ospedaliera (AO) S.Croce e Carle, Cuneo, Italy
| | - Chiara Riganti
- Dipartimento di Oncologia, Università degli Studi di Torino, Torino, Italy
| | - Massimo Massaia
- Laboratorio di Immunologia dei Tumori del Sangue (LITS), Centro Interdipartimentale di Biotecnologie Molecolari “Guido Tarone”, Dipartimento di Biotecnologie Molecolari e Scienze della Salute, Università degli Studi di Torino, Torino, Italy,Struttura Complessa (SC) Ematologia, Azienda Ospedaliera (AO) S.Croce e Carle, Cuneo, Italy,*Correspondence: Massimo Massaia,
| |
Collapse
|
69
|
Azithromycin promotes relapse by disrupting immune and metabolic networks after allogeneic stem cell transplantation. Blood 2022; 140:2500-2513. [PMID: 35984904 DOI: 10.1182/blood.2022016926] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 07/28/2022] [Accepted: 08/16/2022] [Indexed: 12/13/2022] Open
Abstract
Administration of azithromycin after allogeneic hematopoietic stem cell transplantation for hematologic malignancies has been associated with relapse in a randomized phase 3 controlled clinical trial. Studying 240 samples from patients randomized in this trial is a unique opportunity to better understand the mechanisms underlying relapse, the first cause of mortality after transplantation. We used multi-omics on patients' samples to decipher immune alterations associated with azithromycin intake and post-transplantation relapsed malignancies. Azithromycin was associated with a network of altered energy metabolism pathways and immune subsets, including T cells biased toward immunomodulatory and exhausted profiles. In vitro, azithromycin exposure inhibited T-cell cytotoxicity against tumor cells and impaired T-cell metabolism through glycolysis inhibition, down-regulation of mitochondrial genes, and up-regulation of immunomodulatory genes, notably SOCS1. These results highlight that azithromycin directly affects immune cells that favor relapse, which raises caution about long-term use of azithromycin treatment in patients at high risk of malignancies. The ALLOZITHRO trial was registered at www.clinicaltrials.gov as #NCT01959100.
Collapse
|
70
|
Maffini E, Ursi M, Barbato F, Dicataldo M, Roberto M, Campanini E, Dan E, De Felice F, De Matteis S, Storci G, Bonafè M, Arpinati M, Bonifazi F. The prevention of disease relapse after allogeneic hematopoietic cell transplantation in acute myeloid leukemia. Front Oncol 2022; 12:1066285. [DOI: 10.3389/fonc.2022.1066285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Accepted: 11/11/2022] [Indexed: 12/02/2022] Open
Abstract
Disease relapse represents by far the most frequent cause of hematopoietic cell transplantation (HCT) failure. Patients with acute leukemia suffering relapse after HCT have limited conventional treatment options with little possibility of cure and represent, de facto, suitable candidates for the evaluation of novel cellular and biological-based therapies. Donor lymphocyte infusions (DLI) has been one of the first cellular therapies adopted to treat post HCT relapse of acute leukemia patients and still now, it is widely adopted in preemptive and prophylactic settings, with renewed interest for manipulated cellular products such as NK-DLI. The acquisition of novel biological insights into pathobiology of leukemia relapse are translating into the clinic, with novel combinations of target therapies and novel agents, helping delineate new therapeutical landscapes. Hypomethylating agents alone or in combination with novel drugs demonstrated their efficacy in pre-clinical models and controlled trials. FLT3 inhibitors represent an essential therapeutical instrument incorporated in post-transplant maintenance strategies. The Holy grail of allogeneic transplantation lies in the separation of graft-vs.-host disease from graft vs. tumor effects and after more than five decades, is still the most ambitious goal to reach and many ways to accomplish are on their way.
Collapse
|
71
|
Riether C. Regulation of hematopoietic and leukemia stem cells by regulatory T cells. Front Immunol 2022; 13:1049301. [PMID: 36405718 PMCID: PMC9666425 DOI: 10.3389/fimmu.2022.1049301] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Accepted: 10/20/2022] [Indexed: 01/25/2023] Open
Abstract
Adult bone marrow (BM) hematopoietic stem cells (HSCs) are maintained in a quiescent state and sustain the continuous production of all types of blood cells. HSCs reside in a specialized microenvironment the so-called HSC niche, which equally promotes HSC self-renewal and differentiation to ensure the integrity of the HSC pool throughout life and to replenish hematopoietic cells after acute injury, infection or anemia. The processes of HSC self-renewal and differentiation are tightly controlled and are in great part regulated through cellular interactions with classical (e.g. mesenchymal stromal cells) and non-classical niche cells (e.g. immune cells). In myeloid leukemia, some of these regulatory mechanisms that evolved to maintain HSCs, to protect them from exhaustion and immune destruction and to minimize the risk of malignant transformation are hijacked/disrupted by leukemia stem cells (LSCs), the malignant counterpart of HSCs, to promote disease progression as well as resistance to therapy and immune control. CD4+ regulatory T cells (Tregs) are substantially enriched in the BM compared to other secondary lymphoid organs and are crucially involved in the establishment of an immune privileged niche to maintain HSC quiescence and to protect HSC integrity. In leukemia, Tregs frequencies in the BM even increase. Studies in mice and humans identified the accumulation of Tregs as a major immune-regulatory mechanism. As cure of leukemia implies the elimination of LSCs, the understanding of these immune-regulatory processes may be of particular importance for the development of future treatments of leukemia as targeting major immune escape mechanisms which revolutionized the treatment of solid tumors such as the blockade of the inhibitory checkpoint receptor programmed cell death protein 1 (PD-1) seems less efficacious in the treatment of leukemia. This review will summarize recent findings on the mechanisms by which Tregs regulate stem cells and adaptive immune cells in the BM during homeostasis and in leukemia.
Collapse
Affiliation(s)
- Carsten Riether
- Department of Medical Oncology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland,Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland,*Correspondence: Carsten Riether,
| |
Collapse
|
72
|
Minnie SA, Waltner OG, Ensbey KS, Nemychenkov NS, Schmidt CR, Bhise SS, Legg SRW, Campoy G, Samson LD, Kuns RD, Zhou T, Huck JD, Vuckovic S, Zamora D, Yeh A, Spencer A, Koyama M, Markey KA, Lane SW, Boeckh M, Ring AM, Furlan SN, Hill GR. Depletion of exhausted alloreactive T cells enables targeting of stem-like memory T cells to generate tumor-specific immunity. Sci Immunol 2022; 7:eabo3420. [PMID: 36240285 PMCID: PMC10184646 DOI: 10.1126/sciimmunol.abo3420] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Some hematological malignancies such as multiple myeloma are inherently resistant to immune-mediated antitumor responses, the cause of which remains unknown. Allogeneic bone marrow transplantation (alloBMT) is the only curative immunotherapy for hematological malignancies due to profound graft-versus-tumor (GVT) effects, but relapse remains the major cause of death. We developed murine models of alloBMT where the hematological malignancy is either sensitive [acute myeloid leukemia (AML)] or resistant (myeloma) to GVT effects. We found that CD8+ T cell exhaustion in bone marrow was primarily alloantigen-driven, with expression of inhibitory ligands present on myeloma but not AML. Because of this tumor-independent exhaustion signature, immune checkpoint inhibition (ICI) in myeloma exacerbated graft-versus-host disease (GVHD) without promoting GVT effects. Administration of post-transplant cyclophosphamide (PT-Cy) depleted donor T cells with an exhausted phenotype and spared T cells displaying a stem-like memory phenotype with chromatin accessibility present in cytokine signaling genes, including the interleukin-18 (IL-18) receptor. Whereas ICI with anti-PD-1 or anti-TIM-3 remained ineffective after PT-Cy, administration of a decoy-resistant IL-18 (DR-18) strongly enhanced GVT effects in both myeloma and leukemia models, without exacerbation of GVHD. We thus defined mechanisms of resistance to T cell-mediated antitumor effects after alloBMT and described an immunotherapy approach targeting stem-like memory T cells to enhance antitumor immunity.
Collapse
Affiliation(s)
- Simone A. Minnie
- Clinical Research Division, Fred Hutchinson Cancer Center; Seattle, WA, 98109, UNITED STATES
| | - Olivia G. Waltner
- Clinical Research Division, Fred Hutchinson Cancer Center; Seattle, WA, 98109, UNITED STATES
| | - Kathleen S. Ensbey
- Clinical Research Division, Fred Hutchinson Cancer Center; Seattle, WA, 98109, UNITED STATES
| | - Nicole S. Nemychenkov
- Clinical Research Division, Fred Hutchinson Cancer Center; Seattle, WA, 98109, UNITED STATES
| | - Christine R. Schmidt
- Clinical Research Division, Fred Hutchinson Cancer Center; Seattle, WA, 98109, UNITED STATES
| | - Shruti S. Bhise
- Clinical Research Division, Fred Hutchinson Cancer Center; Seattle, WA, 98109, UNITED STATES
| | - Samuel RW. Legg
- Clinical Research Division, Fred Hutchinson Cancer Center; Seattle, WA, 98109, UNITED STATES
| | - Gabriela Campoy
- Clinical Research Division, Fred Hutchinson Cancer Center; Seattle, WA, 98109, UNITED STATES
| | - Luke D. Samson
- Clinical Research Division, Fred Hutchinson Cancer Center; Seattle, WA, 98109, UNITED STATES
| | - Rachel D. Kuns
- QIMR Berghofer Medical Research Institute; Brisbane, QLD, 4006, AUSTRALIA
| | - Ting Zhou
- Department of Immunobiology, Yale School of Medicine; New Haven, CT, 06519, UNITED STATES
| | - John D. Huck
- Department of Immunobiology, Yale School of Medicine; New Haven, CT, 06519, UNITED STATES
| | - Slavica Vuckovic
- QIMR Berghofer Medical Research Institute; Brisbane, QLD, 4006, AUSTRALIA
| | - Danniel Zamora
- Department of Medicine, University of Washington; Seattle, WA, 98109, UNITED STATES
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center; Seattle, WA, 98109, UNITED STATES
| | - Albert Yeh
- Clinical Research Division, Fred Hutchinson Cancer Center; Seattle, WA, 98109, UNITED STATES
- Department of Medicine, University of Washington; Seattle, WA, 98109, UNITED STATES
| | - Andrew Spencer
- Australian Center for Blood Diseases, Monash University/The Alfred Hospital; Melbourne, VIC, 3004, AUSTRALIA
- Malignant Haematology and Stem Cell Transplantation, The Alfred Hospital; Melbourne, VIC, 3004, AUSTRALIA
- Department of Clinical Haematology, Monash University; Melbourne, VIC, 3800, AUSTRALIA
| | - Motoko Koyama
- Clinical Research Division, Fred Hutchinson Cancer Center; Seattle, WA, 98109, UNITED STATES
| | - Kate A. Markey
- Clinical Research Division, Fred Hutchinson Cancer Center; Seattle, WA, 98109, UNITED STATES
- Department of Medicine, University of Washington; Seattle, WA, 98109, UNITED STATES
| | - Steven W. Lane
- QIMR Berghofer Medical Research Institute; Brisbane, QLD, 4006, AUSTRALIA
| | - Michael Boeckh
- Clinical Research Division, Fred Hutchinson Cancer Center; Seattle, WA, 98109, UNITED STATES
- Department of Medicine, University of Washington; Seattle, WA, 98109, UNITED STATES
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center; Seattle, WA, 98109, UNITED STATES
| | - Aaron M. Ring
- Department of Immunobiology, Yale School of Medicine; New Haven, CT, 06519, UNITED STATES
| | - Scott N. Furlan
- Clinical Research Division, Fred Hutchinson Cancer Center; Seattle, WA, 98109, UNITED STATES
- Department of Pediatrics, University of Washington; WA, 98105, UNITED STATES
| | - Geoffrey R. Hill
- Clinical Research Division, Fred Hutchinson Cancer Center; Seattle, WA, 98109, UNITED STATES
- Department of Medicine, University of Washington; Seattle, WA, 98109, UNITED STATES
| |
Collapse
|
73
|
Lak S, Janelle V, Djedid A, Boudreau G, Brasey A, Lisi V, Smaani A, Carli C, Busque L, Lavallée VP, Delisle JS. Combined PD-L1 and TIM3 blockade improves expansion of fit human CD8 + antigen-specific T cells for adoptive immunotherapy. Mol Ther Methods Clin Dev 2022; 27:230-245. [PMID: 36320412 PMCID: PMC9593254 DOI: 10.1016/j.omtm.2022.09.016] [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: 11/05/2021] [Accepted: 09/29/2022] [Indexed: 11/27/2022]
Abstract
Antigen-specific T cell expansion ex vivo followed by adoptive transfer enables targeting of a multitude of microbial and cancer antigens. However, clinical-scale T cell expansion from rare precursors requires repeated stimulation, which may lead to T cell dysfunction and limited therapeutic potential. We used a clinically compliant protocol to expand Epstein-Barr virus (EBV) and Wilms tumor 1 (WT1) antigen-specific CD8+ T cells, and leveraged T cell exhaustion-associated inhibitory receptor blockade to improve T cell expansion. Several inhibitory receptors were expressed early by ex vivo-expanded antigen-specific CD8+ T cells, including PD-1 and TIM3, with co-expression matching evidence of T cell dysfunction as the cultures progressed. Introduction of anti-PD-L1 and anti-TIM3 blockade in combination (but not individually) to the culture led to markedly improved antigen-specific T cell expansion without inducing T cell dysfunction. Single-cell RNA sequencing (RNA-seq) and T cell receptor (TCR) repertoire profiling revealed that double blockade does not impart specific transcriptional programs in T cells or alterations in TCR repertoires. However, combined blockade may affect gene expression in a minority of clonotypes in a donor-specific fashion. We conclude that antigen-specific CD8+ T cell manufacturing can be improved by using TIM3 and PD-L1/PD-1 axis blockade in combination. This approach is readily applicable to several adoptive immunotherapy strategies.
Collapse
Affiliation(s)
- Shirin Lak
- Centre de Recherche de L’Hôpital Maisonneuve-Rosemont, 5415 Boul. de L’Assomption, Montréal, QC H1T 2M4, Canada
| | - Valérie Janelle
- Centre de Recherche de L’Hôpital Maisonneuve-Rosemont, 5415 Boul. de L’Assomption, Montréal, QC H1T 2M4, Canada
| | - Anissa Djedid
- Centre de Recherche Du CHU Sainte-Justine, 3175 Chemin de la Côte-Sainte-Catherine, Montréal, QC H3T 1C5, Canada
| | - Gabrielle Boudreau
- Centre de Recherche de L’Hôpital Maisonneuve-Rosemont, 5415 Boul. de L’Assomption, Montréal, QC H1T 2M4, Canada
| | - Ann Brasey
- Centre de Recherche de L’Hôpital Maisonneuve-Rosemont, 5415 Boul. de L’Assomption, Montréal, QC H1T 2M4, Canada,Biomarker Unit, Centre C3i, 5415 Boul. de L’Assomption, Montréal, QC H1T 2M4, Canada
| | - Véronique Lisi
- Centre de Recherche Du CHU Sainte-Justine, 3175 Chemin de la Côte-Sainte-Catherine, Montréal, QC H3T 1C5, Canada
| | - Ali Smaani
- Centre de Recherche de L’Hôpital Maisonneuve-Rosemont, 5415 Boul. de L’Assomption, Montréal, QC H1T 2M4, Canada
| | - Cédric Carli
- Centre de Recherche de L’Hôpital Maisonneuve-Rosemont, 5415 Boul. de L’Assomption, Montréal, QC H1T 2M4, Canada
| | - Lambert Busque
- Centre de Recherche de L’Hôpital Maisonneuve-Rosemont, 5415 Boul. de L’Assomption, Montréal, QC H1T 2M4, Canada,Biomarker Unit, Centre C3i, 5415 Boul. de L’Assomption, Montréal, QC H1T 2M4, Canada,Department of Medicine, Université de Montréal, CP 6128, Succursale Centre-ville, Montréal, QC H3C 3J7, Canada,Hematology-Oncology and Cell Therapy Division, Hôpital Maisonneuve-Rosemont, Montréal, QC Canada
| | - Vincent-Philippe Lavallée
- Centre de Recherche Du CHU Sainte-Justine, 3175 Chemin de la Côte-Sainte-Catherine, Montréal, QC H3T 1C5, Canada,Department of Pediatrics, Université de Montréal, Montréal, QC, Canada,Hematology-Oncology Division, CHU Sainte-Justine, 3175 Chemin de la Côte-Sainte-Catherine, Montréal, QC H3T 1C5, Canada
| | - Jean-Sébastien Delisle
- Centre de Recherche de L’Hôpital Maisonneuve-Rosemont, 5415 Boul. de L’Assomption, Montréal, QC H1T 2M4, Canada,Department of Medicine, Université de Montréal, CP 6128, Succursale Centre-ville, Montréal, QC H3C 3J7, Canada,Hematology-Oncology and Cell Therapy Division, Hôpital Maisonneuve-Rosemont, Montréal, QC Canada,Corresponding author Jean-Sébastien Delisle, MD, FRCPC, PhD, Centre de recherche de l’Hôpital Maisonneuve-Rosemont 5415, Boul de L’Assomption, Montréal, QC, H1T 2M4, Canada.
| |
Collapse
|
74
|
Hao L, Chen Q, Chen X, Zhou Q. The Role of Gender-Related Immune Genes in Childhood Acute Myeloid Leukemia. BIOMED RESEARCH INTERNATIONAL 2022; 2022:3235238. [PMID: 36193320 PMCID: PMC9525781 DOI: 10.1155/2022/3235238] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 08/17/2022] [Accepted: 09/06/2022] [Indexed: 11/18/2022]
Abstract
The study of immune genes and immune cells is highly focused in recent years. To find immunological genes with prognostic value, the current study examines childhood acute myeloid leukemia according to gender. The TARGET database was used to gather the "mRNA expression profile data" and relevant clinical data of children with AML. To normalize processing and find differentially expressed genes (DEG) between male and female subgroups, the limma software package is utilized. We identified prognostic-related genes and built models using LASSO, multivariate Cox, and univariate Cox analysis. The prognostic significance of prognostic genes was then examined through the processing of survival analysis and risk score (RS) calculation. We investigated the connections between immune cells and prognostic genes as well as the connections between prognostic genes and medications. Finally, five immune genes from the TARGET database have been identified. These immune genes are considerably correlated to the prognosis of male patients.
Collapse
Affiliation(s)
- Lu Hao
- Science and Education Department, Shenzhen Baoan Shiyan People's Hospital, Shenzhen, China
| | - Qiuyan Chen
- Science and Education Department, Shenzhen Baoan Shiyan People's Hospital, Shenzhen, China
| | - Xi Chen
- Central Laboratory, The People's Hospital of Baoan Shenzhen, The Second Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Qing Zhou
- Central Laboratory, The People's Hospital of Baoan Shenzhen, The Second Affiliated Hospital of Shenzhen University, Shenzhen, China
| |
Collapse
|
75
|
How I Treat TP53-Mutated Acute Myeloid Leukemia and Myelodysplastic Syndromes. Cancers (Basel) 2022; 14:cancers14184519. [PMID: 36139679 PMCID: PMC9496940 DOI: 10.3390/cancers14184519] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 09/13/2022] [Accepted: 09/14/2022] [Indexed: 12/19/2022] Open
Abstract
TP53-mutated acute myeloid leukemia (AML) and myelodysplastic syndromes (MDS) are among the myeloid malignancies with the poorest prognosis. In this review, we analyze the prognosis of these two diseases, focussing particularly on the extent of the mono or biallelic mutation status of TP53 mutation, which is largely correlated with cytogenetic complexity. We discuss the possible/potential improvement in outcome based on recent results obtained with new drugs (especially eprenetapopt and magrolimab). We also focus on the impact of allogeneic hematopoietic stem cell transplantation (aHSCT) including post aHSCT treatment.
Collapse
|
76
|
Immune landscape after allo-HSCT: TIGIT- and CD161-expressing CD4 T cells are associated with subsequent leukemia relapse. Blood 2022; 140:1305-1321. [PMID: 35820057 DOI: 10.1182/blood.2022015522] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Accepted: 06/27/2022] [Indexed: 11/20/2022] Open
Abstract
Allogeneic hematopoietic stem cell transplantation (allo-HSCT) is the most effective treatment for selected patients with acute myeloid leukemia (AML) and relies on a "graft-versus-leukemia" effect (GVL) where donor T lymphocytes mediate control of malignant cell growth. However, relapse remains the major cause of death after allo-HSCT. In various malignancies, several immunoregulatory mechanisms have been shown to restrain antitumor immunity, including ligand-mediated engagement of inhibitory receptors (IRs) on effector cells, and induction of immunosuppressive cell subsets, such as regulatory T cells (Tregs) or myeloid-derived suppressor cells (MDSCs). Relapse after HSCT remains a major therapeutic challenge, but immunoregulatory mechanisms involved in restraining the GVL effect must be better deciphered in humans. We used mass cytometry to comprehensively characterize circulating leukocytes in 2 cohorts of patients after allo-HSCT. We first longitudinally assessed various immunoregulatory parameters highlighting specific trends, such as opposite dynamics between MDSCs and Tregs. More generally, the immune landscape was stable from months 3 to 6, whereas many variations occurred from months 6 to 12 after HSCT. Comparison with healthy individuals revealed that profound alterations in the immune equilibrium persisted 1 year after HSCT. Importantly, we found that high levels of TIGIT and CD161 expression on CD4 T cells at month 3 after HSCT were distinct features significantly associated with subsequent AML relapse in a second cross-sectional cohort. Altogether, these data provide global insights into the reconstitution of the immunoregulatory landscape after HSCT and highlight non-canonical IRs associated with relapse, which could open the path to new prognostic tools or therapeutic targets to restore subverted anti-AML immunity.
Collapse
|
77
|
Sánchez Martínez D, Tirado N, Mensurado S, Martínez-Moreno A, Romecín P, Gutiérrez Agüera F, Correia DV, Silva-Santos B, Menéndez P. Generation and proof-of-concept for allogeneic CD123 CAR-Delta One T (DOT) cells in acute myeloid leukemia. J Immunother Cancer 2022; 10:jitc-2022-005400. [PMID: 36162920 PMCID: PMC9516293 DOI: 10.1136/jitc-2022-005400] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/26/2022] [Indexed: 12/04/2022] Open
Abstract
Background Chimeric antigen receptor (CAR)-T cells have emerged as a breakthrough treatment for relapse/refractory hematological tumors, showing impressive complete remission rates. However, around 50% of the patients relapse before 1-year post-treatment. T-cell ‘fitness’ is critical to prolong CAR-T persistence and activity. Allogeneic T cells from healthy donors are less dysfunctional or exhausted than autologous patient-derived T cells; in this context, Delta One T cells (DOTs), a recently described cellular product based on MHC/HLA-independent Vδ1+γδ T cells, represent a promising allogeneic platform. Methods Here we generated and preclinically validated, for the first time, 4-1BB-based CAR-DOTs directed against the interleukin-3α chain receptor (CD123), a target antigen widely expressed on acute myeloid leukemia (AML) blasts. Results CD123CAR-DOTs showed vigorous, superior to control DOTs, cytotoxicity against AML cell lines and primary samples both in vitro and in vivo, even on tumor rechallenge. Conclusions Our results provide the proof-of-concept for a DOT-based next-generation allogeneic CAR-T therapy for AML.
Collapse
Affiliation(s)
- Diego Sánchez Martínez
- Josep Carreras Leukaemia Research Institute, Department of Biomedicine, School of Medicine, University of Barcelona, Barcelona, Spain .,Red Española de Terapias Avanzadas (TERAV) - Instituto de Salud Carlos III (ISCII) (RICORS, RD21/0017/0029)
| | - Néstor Tirado
- Josep Carreras Leukaemia Research Institute, Department of Biomedicine, School of Medicine, University of Barcelona, Barcelona, Spain.,Red Española de Terapias Avanzadas (TERAV) - Instituto de Salud Carlos III (ISCII) (RICORS, RD21/0017/0029)
| | - Sofia Mensurado
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Portugal
| | - Alba Martínez-Moreno
- Josep Carreras Leukaemia Research Institute, Department of Biomedicine, School of Medicine, University of Barcelona, Barcelona, Spain.,Red Española de Terapias Avanzadas (TERAV) - Instituto de Salud Carlos III (ISCII) (RICORS, RD21/0017/0029)
| | - Paola Romecín
- Josep Carreras Leukaemia Research Institute, Department of Biomedicine, School of Medicine, University of Barcelona, Barcelona, Spain.,Red Española de Terapias Avanzadas (TERAV) - Instituto de Salud Carlos III (ISCII) (RICORS, RD21/0017/0029)
| | - Francisco Gutiérrez Agüera
- Josep Carreras Leukaemia Research Institute, Department of Biomedicine, School of Medicine, University of Barcelona, Barcelona, Spain.,Red Española de Terapias Avanzadas (TERAV) - Instituto de Salud Carlos III (ISCII) (RICORS, RD21/0017/0029)
| | - Daniel V Correia
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Portugal
| | - Bruno Silva-Santos
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Portugal
| | - Pablo Menéndez
- Josep Carreras Leukaemia Research Institute, Department of Biomedicine, School of Medicine, University of Barcelona, Barcelona, Spain .,Red Española de Terapias Avanzadas (TERAV) - Instituto de Salud Carlos III (ISCII) (RICORS, RD21/0017/0029).,Centro de Investigación Biomédica en Red-Oncología (CIBERONC), Instituto de Salud Carlos III, Barcelona, Spain.,Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain
| |
Collapse
|
78
|
Xu ZJ, Zhang XL, Jin Y, Wang SS, Gu Y, Ma JC, Wen XM, Leng JY, Mao ZW, Lin J, Qian J. Pan-cancer analysis reveals distinct clinical, genomic, and immunological features of the LILRB immune checkpoint family in acute myeloid leukemia. Mol Ther Oncolytics 2022; 26:88-104. [PMID: 35795094 PMCID: PMC9233190 DOI: 10.1016/j.omto.2022.05.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Accepted: 05/27/2022] [Indexed: 10/28/2022] Open
|
79
|
Ouyang X, Gong Y. One Stone, Two Birds: N6-Methyladenosine RNA Modification in Leukemia Stem Cells and the Tumor Immune Microenvironment in Acute Myeloid Leukemia. Front Immunol 2022; 13:912526. [PMID: 35720276 PMCID: PMC9201081 DOI: 10.3389/fimmu.2022.912526] [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: 04/04/2022] [Accepted: 05/09/2022] [Indexed: 02/05/2023] Open
Abstract
Acute myeloid leukemia is the most common acute leukemia in adults, with accumulation of abundant blasts and impairment of hematogenic function. Despite great advances in diagnosis and therapy, the overall survival of patients with acute myeloid leukemia remains poor. Leukemia stem cells are the root cause of relapse and chemoresistance in acute myeloid leukemia. The tumor immune microenvironment is another trigger to induce recurrence and drug resistance. Understanding the underlying factors influencing leukemia stem cells and the tumor immune microenvironment is an urgent and unmet need. Intriguingly, N6-methyladenosine, the most widespread internal mRNA modification in eukaryotes, is found to regulate both leukemia stem cells and the tumor immune microenvironment. Methyltransferases and demethylases cooperatively make N6-methyladenosine modification reversible and dynamic. Increasing evidence demonstrates that N6-methyladenosine modification extensively participates in tumorigenesis and progression in various cancers, including acute myeloid leukemia. In this review, we summarize the current progress in studies on the functions of N6-methyladenosine modification in acute myeloid leukemia, especially in leukemia stem cells and the tumor immune microenvironment. We generalize the landscape of N6-methyladenosine modification in self-renewal of leukemia stem cells and immune microenvironment regulation, as well as in the initiation, growth, proliferation, differentiation, and apoptosis of leukemia cells. In addition, we further explore the clinical application of N6-methyladenosine modification in diagnosis, prognostic stratification, and effect evaluation. Considering the roles of N6-methyladenosine modification in leukemia stem cells and the tumor immune microenvironment, we propose targeting N6-methyladenosine regulators as one stone to kill two birds for acute myeloid leukemia treatment.
Collapse
Affiliation(s)
- Xianfeng Ouyang
- Department of Hematology, West China Hospital, Sichuan University, Chengdu, China.,Department of Hematology, Jiujiang First People's Hospital, Jiujiang, China
| | - Yuping Gong
- Department of Hematology, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
80
|
Faccani C, Rotta G, Clemente F, Fedeli M, Abbati D, Manfredi F, Potenza A, Anselmo A, Pedica F, Fiorentini G, Villa C, Protti MP, Doglioni C, Aldrighetti L, Bonini C, Casorati G, Dellabona P, de Lalla C. Workflow for high-dimensional flow cytometry analysis of T cells from tumor metastases. Life Sci Alliance 2022; 5:5/10/e202101316. [PMID: 35724271 PMCID: PMC9166301 DOI: 10.26508/lsa.202101316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 05/24/2022] [Accepted: 05/25/2022] [Indexed: 11/24/2022] Open
Abstract
We describe a multi-step high-dimensional (HD) flow cytometry workflow for the deep phenotypic characterization of T cells infiltrating metastatic tumor lesions in the liver, particularly derived from colorectal cancer (CRC-LM). First, we applied a novel flow cytometer setting approach based on single positive cells rather than fluorescent beads, resulting in optimal sensitivity when compared with previously published protocols. Second, we set up a 26-color based antibody panel designed to assess the functional state of both conventional T-cell subsets and unconventional invariant natural killer T, mucosal associated invariant T, and gamma delta T (γδT)-cell populations, which are abundant in the liver. Third, the dissociation of the CRC-LM samples was accurately tuned to preserve both the viability and antigenic integrity of the stained cells. This combined procedure permitted the optimal capturing of the phenotypic complexity of T cells infiltrating CRC-LM. Hence, this study provides a robust tool for high-dimensional flow cytometry analysis of complex T-cell populations, which could be adapted to characterize other relevant pathological tissues.
Collapse
Affiliation(s)
- Cristina Faccani
- Experimental Immunology Unit, Ospedale San Raffaele Scientific Institute, Milan, Italy
- School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | | | - Francesca Clemente
- Tumor Immunology Unit, Ospedale San Raffaele Scientific Institute, Milan, Italy
| | - Maya Fedeli
- Experimental Immunology Unit, Ospedale San Raffaele Scientific Institute, Milan, Italy
- Università Vita-Salute San Raffaele, Milan, Italy
| | - Danilo Abbati
- Experimental Hematology Unit, Ospedale San Raffaele Scientific Institute, Milan, Italy
| | - Francesco Manfredi
- Experimental Hematology Unit, Ospedale San Raffaele Scientific Institute, Milan, Italy
| | - Alessia Potenza
- Experimental Hematology Unit, Ospedale San Raffaele Scientific Institute, Milan, Italy
| | - Achille Anselmo
- Flow Cytometry Resource, Advanced Cytometry Technical Applications Laboratory (FRACTAL) Ospedale San Raffaele Scientific Institute, Milan, Italy
| | - Federica Pedica
- Department of Experimental Oncology, Pathology Unit, Ospedale San Raffaele Scientific Institute, Milan, Italy
| | - Guido Fiorentini
- Hepatobiliary Surgery, Ospedale San Raffaele Scientific Institute, Milan, Italy
| | - Chiara Villa
- Flow Cytometry Resource, Advanced Cytometry Technical Applications Laboratory (FRACTAL) Ospedale San Raffaele Scientific Institute, Milan, Italy
| | - Maria P Protti
- Tumor Immunology Unit, Ospedale San Raffaele Scientific Institute, Milan, Italy
| | - Claudio Doglioni
- Department of Experimental Oncology, Pathology Unit, Ospedale San Raffaele Scientific Institute, Milan, Italy
- Università Vita-Salute San Raffaele, Milan, Italy
| | - Luca Aldrighetti
- Hepatobiliary Surgery, Ospedale San Raffaele Scientific Institute, Milan, Italy
| | - Chiara Bonini
- Experimental Hematology Unit, Ospedale San Raffaele Scientific Institute, Milan, Italy
- Università Vita-Salute San Raffaele, Milan, Italy
| | - Giulia Casorati
- Experimental Immunology Unit, Ospedale San Raffaele Scientific Institute, Milan, Italy
| | - Paolo Dellabona
- Experimental Immunology Unit, Ospedale San Raffaele Scientific Institute, Milan, Italy
| | - Claudia de Lalla
- Experimental Immunology Unit, Ospedale San Raffaele Scientific Institute, Milan, Italy
| |
Collapse
|
81
|
Gambacorta V, Beretta S, Ciccimarra M, Zito L, Giannetti K, Andrisani A, Gnani D, Zanotti L, Oliveira G, Carrabba MG, Cittaro D, Merelli I, Ciceri F, Di Micco R, Vago L. Integrated Multiomic Profiling Identifies the Epigenetic Regulator PRC2 as a Therapeutic Target to Counteract Leukemia Immune Escape and Relapse. Cancer Discov 2022; 12:1449-1461. [PMID: 35255120 PMCID: PMC9394393 DOI: 10.1158/2159-8290.cd-21-0980] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 02/07/2022] [Accepted: 02/28/2022] [Indexed: 01/07/2023]
Abstract
Immune escape represents a major driver of acute myeloid leukemia (AML) reemergence after allogeneic hematopoietic cell transplantation (allo-HCT), with up to 40% of relapses prompted by nongenomic loss of HLA class II expression in leukemia cells. By integrative analysis of gene expression, DNA methylation, and chromatin accessibility in paired diagnosis/relapse primary samples and in the respective patient-derived xenografts (PDX), we identify the polycomb repressive complex 2 (PRC2) as a key epigenetic driver of this immune escape modality. We report that loss of expression of HLA class II molecules is accompanied by a PRC2-dependent reduction in chromatin accessibility. Pharmacologic inhibition of PRC2 subunits rescues HLA class II expression in AML relapses in vitro and in vivo, with consequent recovery of leukemia recognition by CD4+ T cells. Our results uncover a novel link between epigenetics and leukemia immune escape, which may rapidly translate into innovative strategies to cure or prevent AML posttransplantation relapse. SIGNIFICANCE Loss of HLA class II expression represents a frequent mechanism of leukemia posttransplantation relapse. Here we identify PRC2 as the main epigenetic driver of this immune escape modality and show that its chemical inhibition can reinstate a proficient graft-versus-leukemia effect, providing an innovative rationale for personalized epigenetic immunotherapies. See related commentary by Köhler and Zeiser, p. 1410. This article is highlighted in the In This Issue feature, p. 1397.
Collapse
Affiliation(s)
- Valentina Gambacorta
- Unit of Immunogenetics, Leukemia Genomics and Immunobiology, Division of Immunology, Transplantation and Infectious Disease, IRCCS San Raffaele Scientific Institute, Milano, Italy.,Unit of Senescence in Stem Cell Aging, Differentiation and Cancer, San Raffaele Telethon Institute for Gene Therapy (SR-TIGET), IRCCS San Raffaele Scientific Institute, Milano, Italy.,University of Milano Bicocca, Milano, Italy
| | - Stefano Beretta
- Institute for Biomedical Technologies, National Research Council, Segrate, Italy.,San Raffaele Telethon Institute for Gene Therapy (SR-TIGET), IRCCS San Raffaele Scientific Institute, Milano, Italy
| | - Martina Ciccimarra
- Unit of Immunogenetics, Leukemia Genomics and Immunobiology, Division of Immunology, Transplantation and Infectious Disease, IRCCS San Raffaele Scientific Institute, Milano, Italy
| | - Laura Zito
- Unit of Immunogenetics, Leukemia Genomics and Immunobiology, Division of Immunology, Transplantation and Infectious Disease, IRCCS San Raffaele Scientific Institute, Milano, Italy
| | - Kety Giannetti
- Unit of Senescence in Stem Cell Aging, Differentiation and Cancer, San Raffaele Telethon Institute for Gene Therapy (SR-TIGET), IRCCS San Raffaele Scientific Institute, Milano, Italy
| | - Angela Andrisani
- Unit of Immunogenetics, Leukemia Genomics and Immunobiology, Division of Immunology, Transplantation and Infectious Disease, IRCCS San Raffaele Scientific Institute, Milano, Italy
| | - Daniela Gnani
- Unit of Senescence in Stem Cell Aging, Differentiation and Cancer, San Raffaele Telethon Institute for Gene Therapy (SR-TIGET), IRCCS San Raffaele Scientific Institute, Milano, Italy
| | - Lucia Zanotti
- Unit of Immunogenetics, Leukemia Genomics and Immunobiology, Division of Immunology, Transplantation and Infectious Disease, IRCCS San Raffaele Scientific Institute, Milano, Italy
| | - Giacomo Oliveira
- Unit of Immunogenetics, Leukemia Genomics and Immunobiology, Division of Immunology, Transplantation and Infectious Disease, IRCCS San Raffaele Scientific Institute, Milano, Italy
| | - Matteo Giovanni Carrabba
- Hematology and Bone Marrow Transplantation Unit, IRCCS San Raffaele Scientific Institute, Milano, Italy
| | - Davide Cittaro
- Center for Omics Sciences at the IRCCS Ospedale San Raffaele (COSR), IRCCS San Raffaele Scientific Institute, Milano, Italy
| | - Ivan Merelli
- Institute for Biomedical Technologies, National Research Council, Segrate, Italy.,San Raffaele Telethon Institute for Gene Therapy (SR-TIGET), IRCCS San Raffaele Scientific Institute, Milano, Italy
| | - Fabio Ciceri
- Hematology and Bone Marrow Transplantation Unit, IRCCS San Raffaele Scientific Institute, Milano, Italy.,San Raffaele Vita-Salute University, Milano, Italy
| | - Raffaella Di Micco
- Unit of Senescence in Stem Cell Aging, Differentiation and Cancer, San Raffaele Telethon Institute for Gene Therapy (SR-TIGET), IRCCS San Raffaele Scientific Institute, Milano, Italy.,Corresponding Authors: Luca Vago, Unit of Immunogenetics, Leukemia Genomics and Immunobiology, IRCCS San Raffaele Scientific Institute, via Olgettina 60, Milano, Italy. Phone: 39 02-2643-4341; E-mail: ; and Raffaella Di Micco, Unit of Senescence in Stem Cell Aging, Differentiation and Cancer, San Raffaele Telethon Institute for Gene Therapy (SR-TIGET), IRCCS San Raffaele Scientific Institute, via Olgettina 60, Milano, Italy. Phone: 39 02-2643-5024; E-mail:
| | - Luca Vago
- Unit of Immunogenetics, Leukemia Genomics and Immunobiology, Division of Immunology, Transplantation and Infectious Disease, IRCCS San Raffaele Scientific Institute, Milano, Italy.,Hematology and Bone Marrow Transplantation Unit, IRCCS San Raffaele Scientific Institute, Milano, Italy.,San Raffaele Vita-Salute University, Milano, Italy.,Corresponding Authors: Luca Vago, Unit of Immunogenetics, Leukemia Genomics and Immunobiology, IRCCS San Raffaele Scientific Institute, via Olgettina 60, Milano, Italy. Phone: 39 02-2643-4341; E-mail: ; and Raffaella Di Micco, Unit of Senescence in Stem Cell Aging, Differentiation and Cancer, San Raffaele Telethon Institute for Gene Therapy (SR-TIGET), IRCCS San Raffaele Scientific Institute, via Olgettina 60, Milano, Italy. Phone: 39 02-2643-5024; E-mail:
| |
Collapse
|
82
|
Abstract
SUMMARY Accumulating evidence supports that loss of HLA expression contributes to relapse after allogeneic hematopoietic cell transplantation (allo-HCT), but the mechanisms behind this evasion strategy are unclear. The groups of Luca Vago and Raffaella Di Micco identified the polycomb repressive complex 2 (PRC2) as a key epigenetic driver of immune escape after allo-HCT by reducing the chromatin accessibility of HLA class II molecules, which could be targeted by pharmacologic inhibition of PRC2 subunits. See related article by Gambacorta et al., p. 1449 (10).
Collapse
Affiliation(s)
- Natalie Köhler
- Department of Medicine I, Faculty of Medicine, Medical Center, University of Freiburg, Freiburg, Germany.,CIBSS-Centre for Integrative Biological Signalling Studies, University of Freiburg, Freiburg, Germany
| | - Robert Zeiser
- Department of Medicine I, Faculty of Medicine, Medical Center, University of Freiburg, Freiburg, Germany.,CIBSS-Centre for Integrative Biological Signalling Studies, University of Freiburg, Freiburg, Germany
| |
Collapse
|
83
|
Krakow EF, Walter RB, Nathe JM, Perez T, Ahmed A, Polissar N, Miljacic L, Halpern AB, Flowers MED, Estey E. Intensive chemotherapy for acute myeloid leukemia relapse after allogeneic hematopoietic cell transplantation. Am J Hematol 2022; 97:E220-E223. [PMID: 35303371 DOI: 10.1002/ajh.26540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 03/14/2022] [Accepted: 03/15/2022] [Indexed: 11/09/2022]
Affiliation(s)
- Elizabeth F. Krakow
- Clinical Research Division of the Fred Hutchinson Cancer Research Center Seattle WA USA
- Division of Medical Oncology University of Washington Seattle WA USA
| | - Roland B. Walter
- Clinical Research Division of the Fred Hutchinson Cancer Research Center Seattle WA USA
- Division of Hematology University of Washington Seattle WA USA
| | - Julia M. Nathe
- Clinical Research Division of the Fred Hutchinson Cancer Research Center Seattle WA USA
| | - Tess Perez
- Clinical Research Division of the Fred Hutchinson Cancer Research Center Seattle WA USA
| | - Ali Ahmed
- Clinical Research Division of the Fred Hutchinson Cancer Research Center Seattle WA USA
| | - Nayak Polissar
- The Mountain‐Whisper‐Light: Statistics & Data Science Seattle WA USA
| | - Ljubomir Miljacic
- The Mountain‐Whisper‐Light: Statistics & Data Science Seattle WA USA
| | - Anna B. Halpern
- Clinical Research Division of the Fred Hutchinson Cancer Research Center Seattle WA USA
- Division of Hematology University of Washington Seattle WA USA
| | - Mary E. D. Flowers
- Clinical Research Division of the Fred Hutchinson Cancer Research Center Seattle WA USA
- Division of Medical Oncology University of Washington Seattle WA USA
| | - Eli Estey
- Clinical Research Division of the Fred Hutchinson Cancer Research Center Seattle WA USA
- Division of Hematology University of Washington Seattle WA USA
| |
Collapse
|
84
|
Liang Q, Zhang L, Wang W, Zhao J, Li Q, Pan H, Gao Z, Fang L, Shi J. High Expression of DC-STAMP Gene Predicts Adverse Outcomes in AML. Front Genet 2022; 13:876689. [PMID: 35571050 PMCID: PMC9091727 DOI: 10.3389/fgene.2022.876689] [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: 02/15/2022] [Accepted: 03/21/2022] [Indexed: 12/30/2022] Open
Abstract
Acute myeloid leukemia (AML) is a genetically heterogeneous hematological malignancy with poor prognosis. We explored the RNA sequence data and clinical information of AML patients from The Cancer Genome Atlas (TCGA) and Genotype-Tissue Expression (GTEx) database to search for the core molecule for prognosis. The DC-STAMP expression was significantly higher in AML patients, which was linked to old age, unfavorable cytogenetic risk, and death (all p < 0.05). Furthermore, it was revealed that high DC-STAMP expression was an independent unfavorable factor for overall survival (OS) by univariate analysis [hazard ratio (HR): 2.683; 95% confidence interval (CI): 1.723–4.178; p < 0.001] and multivariate analysis (HR: 1.733; 95% CI: 1.079–2.781; p = 0.023). The concordance index (C-index 0.734, 95% CI: 0.706–0.762), calibration curves, and decision curve analysis showed the certain predictive accuracy of a nomogram model based on multivariate analysis for OS. In addition, we found that the differentially expressed gene (DEG) enrichment pathways of high- and low-DC-STAMP expression group enrichment pathways were focused on channel activity and platelet alpha granule by the Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG), while gene set enrichment analysis (GSEA) pathways were mainly involved in mTORC1 signaling and TNF-α signaling via the NF-kB pathway. Moreover, a protein–protein interaction (PPI) network demonstrated that DC-STAMP interacted with two hub genes (PPBP and PF4), which were highly regulated and associated with poor survival. Finally, high DC-STAMP expression showed a significantly positive correlation with four immune cell [NK CD56 (dim) cells, macrophages, cytotoxic cells, and CD8 (+) T cells] infiltration and high level of immune checkpoint genes (PDCD1, CD274, CTLA-4, and TIGIT). Therefore, our results suggest that high expression of DC-STAMP predicts adverse outcomes for AML patients.
Collapse
Affiliation(s)
- Qian Liang
- Regenerative Medicine Clinic, State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| | - Lele Zhang
- Regenerative Medicine Clinic, State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| | - Wenjun Wang
- Regenerative Medicine Clinic, State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| | - Jingyu Zhao
- Regenerative Medicine Clinic, State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| | - Qiaoli Li
- Regenerative Medicine Clinic, State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| | - Hong Pan
- Regenerative Medicine Clinic, State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| | - Zhen Gao
- Regenerative Medicine Clinic, State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| | - Liwei Fang
- Regenerative Medicine Clinic, State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| | - Jun Shi
- Regenerative Medicine Clinic, State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| |
Collapse
|
85
|
Arcangeli S, Bove C, Mezzanotte C, Camisa B, Falcone L, Manfredi F, Bezzecchi E, El Khoury R, Norata R, Sanvito F, Ponzoni M, Greco B, Moresco MA, Carrabba MG, Ciceri F, Bonini C, Bondanza A, Casucci M. CAR T-cell manufacturing from naive/stem memory T-lymphocytes enhances antitumor responses while curtailing cytokine release syndrome. J Clin Invest 2022; 132:150807. [PMID: 35503659 PMCID: PMC9197529 DOI: 10.1172/jci150807] [Citation(s) in RCA: 131] [Impact Index Per Article: 43.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Accepted: 04/26/2022] [Indexed: 11/21/2022] Open
Abstract
Chimeric antigen receptor (CAR) T cell expansion and persistence represent key factors to achieve complete responses and prevent relapses. These features are typical of early memory T cells, which can be highly enriched through optimized manufacturing protocols. Here, we investigated the efficacy and safety profiles of CAR T cell products generated from preselected naive/stem memory T cells (TN/SCM), as compared with unselected T cells (TBULK). Notwithstanding their reduced effector signature in vitro, limiting CAR TN/SCM doses showed superior antitumor activity and the unique ability to counteract leukemia rechallenge in hematopoietic stem/precursor cell–humanized mice, featuring increased expansion rates and persistence together with an ameliorated exhaustion and memory phenotype. Most relevantly, CAR TN/SCM proved to be intrinsically less prone to inducing severe cytokine release syndrome, independently of the costimulatory endodomain employed. This safer profile was associated with milder T cell activation, which translated into reduced monocyte activation and cytokine release. These data suggest that CAR TN/SCM are endowed with a wider therapeutic index compared with CAR TBULK.
Collapse
Affiliation(s)
- Silvia Arcangeli
- Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Camilla Bove
- Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Claudia Mezzanotte
- Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Barbara Camisa
- Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Laura Falcone
- Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Francesco Manfredi
- Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Eugenia Bezzecchi
- Center for Omics Sciences, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Rita El Khoury
- Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Rossana Norata
- San Raffaele Telethon Institute for Gene Therapy (SR-TIGET), IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Francesca Sanvito
- Division of Experimental Oncology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Maurilio Ponzoni
- Division of Experimental Oncology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Beatrice Greco
- Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Marta Angiola Moresco
- Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Matteo G Carrabba
- Department of Hematology and Stem Cell Transplantation, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Fabio Ciceri
- Department of Hematology and Stem Cell Transplantation, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Chiara Bonini
- Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Attilio Bondanza
- Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Monica Casucci
- Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
| |
Collapse
|
86
|
Xu Q, Cao D, Fang B, Yan S, Hu Y, Guo T. Immune-related gene signature predicts clinical outcomes and immunotherapy response in acute myeloid leukemia. Cancer Med 2022; 11:3364-3380. [PMID: 35355427 PMCID: PMC9468431 DOI: 10.1002/cam4.4687] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 01/14/2022] [Accepted: 01/25/2022] [Indexed: 12/05/2022] Open
Abstract
Background The immune response in the bone marrow microenvironment has implications for progression and prognosis in acute myeloid leukemia (AML). However, few immune‐related biomarkers for AML prognosis and immunotherapy response have been identified. We aimed to establish a predictive gene signature and to explore the determinants of prognosis in AML. Methods Immune‐related genes with clinical significance were screened by a weighted gene co‐expression network analysis. Seven immune‐related genes were used to establish a gene signature by a multivariate Cox regression analysis. Based on the signature, low‐ and high‐risk groups were compared with respect to the immune microenvironment, immune checkpoints, pathway activities, and mutation frequencies. The tumor immune dysfunction and exclusion (TIDE) method was used to predict the response to immune checkpoint blockade (ICB) therapy. The Connectivity Map database was used to explore small‐molecule drugs expected to treat high‐risk populations. Results A seven‐gene prognostic signature was used to classify patients into high‐ and low‐risk groups. Prognosis was poorer for patients in the former than in the latter. The high‐risk group displayed higher levels of immune checkpoint molecules (LAG3, PD‐1, CTLA4, PD‐L2, and PD‐L1), immune cell infiltration (dendritic cells, T helper 1, and gamma delta T), and somatic mutations (NPM1 and RUNX1). Moreover, hematopoietic stem cell/leukemia stem cell pathways were enriched in the high‐risk phenotype. Compared with that in the low‐risk group, the lower TIDE score for the high‐risk group implied that this group is more likely to benefit from ICB therapy. Finally, some drugs (FLT3 inhibitors and BCL inhibitors) targeting the expression profiles associated with the high‐risk group were generated using Connectivity Map. Conclusion The newly developed immune‐related gene signature is an effective biomarker for predicting prognosis in AML and provides a basis, from an immunological perspective, for the development of comprehensive therapeutic strategies.
Collapse
Affiliation(s)
- Qiang Xu
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Collaborative Innovation Center of Hematology, Huazhong University of Science and Technology, Wuhan, China
| | - Dedong Cao
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Bin Fang
- Department of Nephrology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Siqi Yan
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yu Hu
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Collaborative Innovation Center of Hematology, Huazhong University of Science and Technology, Wuhan, China
| | - Tao Guo
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Collaborative Innovation Center of Hematology, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
87
|
Tang Y, Zhou Z, Yan H, You Y. Case Report: Preemptive Treatment With Low-Dose PD-1 Blockade and Azacitidine for Molecular Relapsed Acute Myeloid Leukemia With RUNX1-RUNX1T1 After Allogeneic Hematopoietic Stem Cell Transplantation. Front Immunol 2022; 13:810284. [PMID: 35185899 PMCID: PMC8847388 DOI: 10.3389/fimmu.2022.810284] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2021] [Accepted: 01/17/2022] [Indexed: 11/25/2022] Open
Abstract
Acute myeloid leukemia (AML) patients who develop hematological relapse (HR) after allogeneic hematopoietic stem cell transplantation (allo-HSCT) generally have dismal clinical outcomes. Measurable residual disease (MRD)-directed preemptive interventions are effective approaches to prevent disease progression and improve prognosis for molecular relapsed patients with warning signs of impending HR. In this situation, boosting the graft-vs-leukemia (GVL) effect with immune checkpoint inhibitors (ICIs) might be a promising prevention strategy, despite the potential for causing severe graft-vs-host disease (GVHD). In the present study, we reported for the first time an AML patient with RUNX1-RUNX1T1 who underwent preemptive treatment with the combined application of tislelizumab (an anti-PD-1 antibody) and azacitidine to avoid HR following allo-HSCT. On day +81, molecular relapse with MRD depicted by RUNX1-RUN1T1-positivity as well as mixed donor chimerism occurred in the patient. On day +95, with no signs of GVHD and an excellent eastern cooperative oncology group performance status (ECOG PS), the patient thus was administered with 100 mg of tislelizumab on day 1 and 100 mg of azacitidine on days 1-7. After the combination therapy, complete remission was successfully achieved with significant improvement in hematologic response, and the MRD marker RUNX1-RUNX1T1 turned negative, along with a complete donor chimerism in bone marrow. Meanwhile, the patient experienced moderate GVHD and immune-related adverse events (irAEs), successively involving the lung, liver, lower digestive tract and urinary system, which were well controlled by immunosuppressive therapies. As far as we know, this case is the first one to report the use of tislelizumab in combination with azacitidine to prevent post-transplant relapse in AML. In summary, the application of ICIs in MRD positive patients might be an attractive strategy for immune modulation in the future to reduce the incidence of HR in the post-transplant setting, but safer clinical application schedules need to be explored.
Collapse
Affiliation(s)
- Yutong Tang
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhenyang Zhou
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China
| | - Han Yan
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yong You
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
88
|
Xu J, Dong X, Wang R, Chen B. DOK2 Has Prognostic and Immunologic Significance in Adults With Acute Myeloid Leukemia: A Novel Immune-Related Therapeutic Target. Front Med (Lausanne) 2022; 9:842383. [PMID: 35321466 PMCID: PMC8935080 DOI: 10.3389/fmed.2022.842383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Accepted: 02/09/2022] [Indexed: 11/13/2022] Open
Abstract
BackgroundThe role of downstream tyrosine kinase 2 (DOK2), a major member of the DOK family, remains poorly defined in acute myeloid leukemia (AML). Herein, we investigated the expression levels, clinical outcomes, and biological functions of DOK2 in patients with AML.MethodsDatasets were obtained from the Cancer Genome Atlas (TCGA) database for transcriptomic and clinical information. Nomogram construction and assessment were conducted using Cox regression analysis, receiver operating characteristic (ROC) curves, and calibration plots. Public databases, including the Gene Expression Omnibus, Cancer Cell Line Encyclopedia, LinkedOmics, GeneMANIA, TISIDB, and Gene Set Cancer Analysis, were employed for relevant bioinformatic studies. Moreover, we utilized the CIBERSORT algorithm to evaluate the level of infiltration of immune cells into the bone marrow microenvironment.ResultsWe observed that DOK2 transcription levels were markedly upregulated in AML samples (P < 0.001), and its high expression was associated with inferior overall survival (OS) (HR = 2.17, P < 0.001) and disease-free survival (DFS) (HR = 2.50, P < 0.001). ROC curve analysis also showed the reliable diagnostic efficiency of DOK2 in AML. For treatment regimens, patients with high DOK2 expression could significantly prolong OS by receiving hematopoietic stem cell transplantation (HSCT) (P < 0.001), whereas those with low DOK2 expression were more likely to improve DFS by chemotherapy alone rather than HSCT. Nomograms constructed for predicting OS and DFS exhibited satisfactory discrimination and accuracy. Functional enrichment analysis identified that DOK2 was involved in important pathways associated with immune-related activities. Furthermore, CIBERSORT scores reflected negative correlations of DOK2 with activated mast cells and resting CD4+ memory T cells, which indicated its adverse immunomodulatory potential.ConclusionWe suggest that elevated DOK2 expression could be an unfavorable prognostic indicator of survival in patients with AML. Our findings provide new insights into the role of DOK2 in AML, with promising clinical implications.
Collapse
|
89
|
Yang X, Ma L, Zhang X, Huang L, Wei J. Targeting PD-1/PD-L1 pathway in myelodysplastic syndromes and acute myeloid leukemia. Exp Hematol Oncol 2022; 11:11. [PMID: 35236415 PMCID: PMC8889667 DOI: 10.1186/s40164-022-00263-4] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Accepted: 02/16/2022] [Indexed: 12/14/2022] Open
Abstract
Myelodysplastic syndromes (MDS) and acute myeloid leukemia (AML) are clonal hematopoietic stem cell diseases arising from the bone marrow (BM), and approximately 30% of MDS eventually progress to AML, associated with increasingly aggressive neoplastic hematopoietic clones and poor survival. Dysregulated immune microenvironment has been recognized as a key pathogenic driver of MDS and AML, causing high rate of intramedullary apoptosis in lower-risk MDS to immunosuppression in higher-risk MDS and AML. Immune checkpoint molecules, including programmed cell death-1 (PD-1) and programmed cell death ligand-1 (PD-L1), play important roles in oncogenesis by maintaining an immunosuppressive tumor microenvironment. Recently, both molecules have been examined in MDS and AML. Abnormal inflammatory signaling, genetic and/or epigenetic alterations, interactions between cells, and treatment of patients all have been involved in dysregulating PD-1/PD-L1 signaling in these two diseases. Furthermore, with the PD-1/PD-L1 pathway activated in immune microenvironment, the milieu of BM shift to immunosuppressive, contributing to a clonal evolution of blasts. Nevertheless, numerous preclinical studies have suggested a potential response of patients to PD-1/PD-L1 blocker. Current clinical trials employing these drugs in MDS and AML have reported mixed clinical responses. In this paper, we focus on the recent preclinical advances of the PD-1/PD-L1 signaling in MDS and AML, and available and ongoing outcomes of PD-1/PD-L1 inhibitor in patients. We also discuss the novel PD-1/PD-L1 blocker-based immunotherapeutic strategies and challenges, including identifying reliable biomarkers, determining settings, and exploring optimal combination therapies.
Collapse
Affiliation(s)
- Xingcheng Yang
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, Hubei, China.,Immunotherapy Research Center for Hematologic Diseases of Hubei Province, Wuhan, 430030, Hubei, China
| | - Ling Ma
- Department of Clinical Laboratory, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Xiaoying Zhang
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, Hubei, China.,Immunotherapy Research Center for Hematologic Diseases of Hubei Province, Wuhan, 430030, Hubei, China
| | - Liang Huang
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, Hubei, China. .,Immunotherapy Research Center for Hematologic Diseases of Hubei Province, Wuhan, 430030, Hubei, China.
| | - Jia Wei
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, Hubei, China. .,Immunotherapy Research Center for Hematologic Diseases of Hubei Province, Wuhan, 430030, Hubei, China.
| |
Collapse
|
90
|
Salhotra A, Stein AS. Role of Radiation Based Conditioning Regimens in Patients With High-Risk AML Undergoing Allogenic Transplantation in Remission or Active Disease and Mechanisms of Post-Transplant Relapse. Front Oncol 2022; 12:802648. [PMID: 35242706 PMCID: PMC8886676 DOI: 10.3389/fonc.2022.802648] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Accepted: 01/21/2022] [Indexed: 11/25/2022] Open
Abstract
In the two decades there has been a consistent improvement in the clinical outcomes of patients diagnosed with acute leukemia undergoing allogenic stem cell transplantation. These improvements have been made possible by advancements in supportive care practices, more precise risk stratification of leukemia patients by genetic testing at diagnosis, accurate disease assessment by measurable residual disease (MRD) in pretransplant marrow and attempts to clear residual disease clones prior to transplant. Availability of targeted therapies, immunotherapies, and approval of novel drug combinations with BCL-2 inhibitors has also improved remission rates for patients who are undergoing transplant. For patients who are unable to achieve a morphologic or MRD- remission prior to transplant, the risk of relapse post-transplant remains high. Total body irradiation (TBI) based intensification of transplant conditioning may be able to overcome risk of increased relapse rate in this clinical setting by improving clearance of leukemic clones. However, in the past increased nonrelapse mortality (NRM) associated with escalation of conditioning intensity has neutralized any potential benefit of decreasing relapse rate in HCT patient resulting in no significant improvement in overall survival. In this review we discuss incorporation of newer radiation techniques such as total marrow irradiation (TMI) to safely deliver targeted doses of radiation at higher doses to improve outcomes of patients with active leukemia. We also discuss the mechanisms associated with leukemia relapse and treatment options available in post allo-HCT relapse setting despite use of intensified conditioning regimens.
Collapse
Affiliation(s)
- Amandeep Salhotra
- Department of Hematology/Hematopoietic Cell Transplant (HCT), City of Hope National Cancer Center, Duarte, CA, United States
| | | |
Collapse
|
91
|
Ruggiero E, Carnevale E, Prodeus A, Magnani ZI, Camisa B, Merelli I, Politano C, Stasi L, Potenza A, Cianciotti BC, Manfredi F, Di Bono M, Vago L, Tassara M, Mastaglio S, Ponzoni M, Sanvito F, Liu D, Balwani I, Galli R, Genua M, Ostuni R, Doglio M, O'Connell D, Dutta I, Yazinski SA, McKee M, Arredouani MS, Schultes B, Ciceri F, Bonini C. CRISPR-based gene disruption and integration of high-avidity, WT1-specific T cell receptors improve antitumor T cell function. Sci Transl Med 2022; 14:eabg8027. [PMID: 35138911 DOI: 10.1126/scitranslmed.abg8027] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
T cell receptor (TCR)-based therapy has the potential to induce durable clinical responses in patients with cancer by targeting intracellular tumor antigens with high sensitivity and by promoting T cell survival. However, the need for TCRs specific for shared oncogenic antigens and the need for manufacturing protocols able to redirect T cell specificity while preserving T cell fitness remain limiting factors. By longitudinal monitoring of T cell functionality and dynamics in 15 healthy donors, we isolated 19 TCRs specific for Wilms' tumor antigen 1 (WT1), which is overexpressed by several tumor types. TCRs recognized several peptides restricted by common human leukocyte antigen (HLA) alleles and displayed a wide range of functional avidities. We selected five high-avidity HLA-A*02:01-restricted TCRs, three that were specific to the less explored immunodominant WT137-45 and two that were specific to the noncanonical WT1-78-64 epitopes, both naturally processed by primary acute myeloid leukemia (AML) blasts. With CRISPR-Cas9 genome editing tools, we combined TCR-targeted integration into the TCR α constant (TRAC) locus with TCR β constant (TRBC) knockout, thus avoiding TCRαβ mispairing and maximizing TCR expression and function. The engineered lymphocytes were enriched in memory stem T cells. A unique WT137-45-specific TCR showed antigen-specific responses and efficiently killed AML blasts, acute lymphoblastic leukemia blasts, and glioblastoma cells in vitro and in vivo in the absence of off-tumor toxicity. T cells engineered to express this receptor are being advanced into clinical development for AML immunotherapy and represent a candidate therapy for other WT1-expressing tumors.
Collapse
Affiliation(s)
- Eliana Ruggiero
- Experimental Hematology Unit, Division of Immunology, Transplantation and Infectious Diseases, Ospedale San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Erica Carnevale
- Experimental Hematology Unit, Division of Immunology, Transplantation and Infectious Diseases, Ospedale San Raffaele Scientific Institute, 20132 Milan, Italy
| | | | - Zulma Irene Magnani
- Experimental Hematology Unit, Division of Immunology, Transplantation and Infectious Diseases, Ospedale San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Barbara Camisa
- Experimental Hematology Unit, Division of Immunology, Transplantation and Infectious Diseases, Ospedale San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Ivan Merelli
- San Raffaele Telethon Institute for Gene Therapy (SR-TIGET), Ospedale San Raffaele Scientific Institute, 20132 Milan, Italy.,National Research Council, Institute for Biomedical Technologies, Segrate, Italy
| | - Claudia Politano
- Experimental Hematology Unit, Division of Immunology, Transplantation and Infectious Diseases, Ospedale San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Lorena Stasi
- Experimental Hematology Unit, Division of Immunology, Transplantation and Infectious Diseases, Ospedale San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Alessia Potenza
- Experimental Hematology Unit, Division of Immunology, Transplantation and Infectious Diseases, Ospedale San Raffaele Scientific Institute, 20132 Milan, Italy.,School of Medicine and Surgery, Milano-Bicocca University, 20126 Milan, Italy
| | - Beatrice Claudia Cianciotti
- Experimental Hematology Unit, Division of Immunology, Transplantation and Infectious Diseases, Ospedale San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Francesco Manfredi
- Experimental Hematology Unit, Division of Immunology, Transplantation and Infectious Diseases, Ospedale San Raffaele Scientific Institute, 20132 Milan, Italy.,Vita-Salute San Raffaele University, 20132 Milan, Italy
| | - Mattia Di Bono
- Experimental Hematology Unit, Division of Immunology, Transplantation and Infectious Diseases, Ospedale San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Luca Vago
- Immunogenetics, Leukemia Genomics and Immunobiology Unit, Division of Immunology, Transplantation and Infectious Diseases, Ospedale San Raffaele Scientific Institute, 20132 Milan, Italy.,Hematology and Bone Marrow Transplantation Unit, Ospedale San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Michela Tassara
- Immunohematology and Transfusion Medicine Unit, Ospedale San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Sara Mastaglio
- Hematology and Bone Marrow Transplantation Unit, Ospedale San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Maurilio Ponzoni
- Vita-Salute San Raffaele University, 20132 Milan, Italy.,Pathology Unit, Ospedale San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Francesca Sanvito
- Pathology Unit, Ospedale San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Dai Liu
- Intellia Therapeutics, Cambridge, MA 02139, USA
| | | | - Rossella Galli
- Neural Stem Cell Biology Unit, Division of Neurosciences, Ospedale San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Marco Genua
- Genomics of the Innate Immune System Unit, San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), Ospedale San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Renato Ostuni
- Vita-Salute San Raffaele University, 20132 Milan, Italy.,Genomics of the Innate Immune System Unit, San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), Ospedale San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Matteo Doglio
- Experimental Hematology Unit, Division of Immunology, Transplantation and Infectious Diseases, Ospedale San Raffaele Scientific Institute, 20132 Milan, Italy
| | | | - Ivy Dutta
- Intellia Therapeutics, Cambridge, MA 02139, USA
| | | | - Mark McKee
- Intellia Therapeutics, Cambridge, MA 02139, USA
| | | | | | - Fabio Ciceri
- Vita-Salute San Raffaele University, 20132 Milan, Italy.,Hematology and Bone Marrow Transplantation Unit, Ospedale San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Chiara Bonini
- Experimental Hematology Unit, Division of Immunology, Transplantation and Infectious Diseases, Ospedale San Raffaele Scientific Institute, 20132 Milan, Italy.,Vita-Salute San Raffaele University, 20132 Milan, Italy
| |
Collapse
|
92
|
Azacitidine-induced reconstitution of the bone marrow T cell repertoire is associated with superior survival in AML patients. Blood Cancer J 2022; 12:19. [PMID: 35091554 PMCID: PMC8799690 DOI: 10.1038/s41408-022-00615-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 01/09/2022] [Accepted: 01/13/2022] [Indexed: 12/31/2022] Open
Abstract
Hypomethylating agents (HMA) like azacitidine are licensed for the treatment of acute myeloid leukemia (AML) patients ineligible for allogeneic hematopoietic stem cell transplantation. Biomarker-driven identification of HMA-responsive patients may facilitate the choice of treatment, especially in the challenging subgroup above 60 years of age. Since HMA possesses immunomodulatory functions that constitute part of their anti-tumor effect, we set out to analyze the bone marrow (BM) immune environment by next-generation sequencing of T cell receptor beta (TRB) repertoires in 51 AML patients treated within the RAS-AZIC trial. Patients with elevated pretreatment T cell diversity (11 out of 41 patients) and those with a boost of TRB richness on day 15 after azacitidine treatment (12 out of 46 patients) had longer event-free and overall survival. Both pretreatment and dynamic BM T cell metrics proved to be better predictors of outcome than other established risk factors. The favorable broadening of the BM T cell space appeared to be driven by antigen since these patients showed significant skewing of TRBV gene usage. Our data suggest that one course of AZA can cause reconstitution to a more physiological T cell BM niche and that the T cell space plays an underestimated prognostic role in AML. Trial registration: DRKS identifier: DRKS00004519
Collapse
|
93
|
Greco B, Malacarne V, De Girardi F, Scotti GM, Manfredi F, Angelino E, Sirini C, Camisa B, Falcone L, Moresco MA, Paolella K, Di Bono M, Norata R, Sanvito F, Arcangeli S, Doglioni C, Ciceri F, Bonini C, Graziani A, Bondanza A, Casucci M. Disrupting N-glycan expression on tumor cells boosts chimeric antigen receptor T cell efficacy against solid malignancies. Sci Transl Med 2022; 14:eabg3072. [PMID: 35044789 DOI: 10.1126/scitranslmed.abg3072] [Citation(s) in RCA: 78] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Immunotherapy with chimeric antigen receptor (CAR)-engineered T cells showed exceptional successes in patients with refractory B cell malignancies. However, first-in-human studies in solid tumors revealed unique hurdles contributing to poor demonstration of efficacy. Understanding the determinants of tumor recognition by CAR T cells should translate into the design of strategies that can overcome resistance. Here, we show that multiple carcinomas express extracellular N-glycans, whose abundance negatively correlates with CAR T cell killing. By knocking out mannoside acetyl-glucosaminyltransferase 5 (MGAT5) in pancreatic adenocarcinoma (PAC), we showed that N-glycans protect tumors from CAR T cell killing by interfering with proper immunological synapse formation and reducing transcriptional activation, cytokine production, and cytotoxicity. To overcome this barrier, we exploited the high metabolic demand of tumors to safely inhibit N-glycans synthesis with the glucose/mannose analog 2-deoxy-d-glucose (2DG). Treatment with 2DG disrupts the N-glycan cover on tumor cells and results in enhanced CAR T cell activity in different xenograft mouse models of PAC. Moreover, 2DG treatment interferes with the PD-1-PD-L1 axis and results in a reduced exhaustion profile of tumor-infiltrating CAR T cells in vivo. The combined 2DG and CAR T cell therapy was successful against multiple carcinomas besides PAC, including those arising from the lung, ovary, and bladder, and with different clinically relevant CAR specificities, such as CD44v6 and CEA. Overall, our results indicate that tumor N-glycosylation regulates the quality and magnitude of CAR T cell responses, paving the way for the rational design of improved therapies against solid malignancies.
Collapse
Affiliation(s)
- Beatrice Greco
- Innovative Immunotherapies Unit, Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy.,Vita-Salute San Raffaele University, 20132 Milan, Italy
| | - Valeria Malacarne
- Lipid Signaling in Cancer and Metabolism Unit, Division of Experimental Oncology, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy.,Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Turin, 10124 Torino, Italy
| | - Federica De Girardi
- Innovative Immunotherapies Unit, Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Giulia Maria Scotti
- Center for Omics Sciences, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Francesco Manfredi
- Experimental Hematology Unit, Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Elia Angelino
- Lipid Signaling in Cancer and Metabolism Unit, Division of Experimental Oncology, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy.,Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Turin, 10124 Torino, Italy
| | - Camilla Sirini
- Innovative Immunotherapies Unit, Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy.,Vita-Salute San Raffaele University, 20132 Milan, Italy
| | - Barbara Camisa
- Innovative Immunotherapies Unit, Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy.,Experimental Hematology Unit, Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Laura Falcone
- Innovative Immunotherapies Unit, Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Marta Angiola Moresco
- Innovative Immunotherapies Unit, Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Katia Paolella
- Innovative Immunotherapies Unit, Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Mattia Di Bono
- Innovative Immunotherapies Unit, Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy.,Experimental Hematology Unit, Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Rossana Norata
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Francesca Sanvito
- Pathology Unit, Division of Experimental Oncology, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Silvia Arcangeli
- Innovative Immunotherapies Unit, Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Claudio Doglioni
- Vita-Salute San Raffaele University, 20132 Milan, Italy.,Pathology Unit, Division of Experimental Oncology, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Fabio Ciceri
- Vita-Salute San Raffaele University, 20132 Milan, Italy.,Hematology and Hematopoietic Stem Cell Transplantation Unit, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Chiara Bonini
- Vita-Salute San Raffaele University, 20132 Milan, Italy.,Experimental Hematology Unit, Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Andrea Graziani
- Lipid Signaling in Cancer and Metabolism Unit, Division of Experimental Oncology, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy.,Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Turin, 10124 Torino, Italy
| | - Attilio Bondanza
- Innovative Immunotherapies Unit, Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy.,Vita-Salute San Raffaele University, 20132 Milan, Italy
| | - Monica Casucci
- Innovative Immunotherapies Unit, Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| |
Collapse
|
94
|
Webster JA, Luznik L, Gojo I. Treatment of AML Relapse After Allo-HCT. Front Oncol 2022; 11:812207. [PMID: 34976845 PMCID: PMC8716583 DOI: 10.3389/fonc.2021.812207] [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: 11/09/2021] [Accepted: 11/25/2021] [Indexed: 01/02/2023] Open
Abstract
With advances in allogeneic hematopoietic stem cell transplant (allo-HCT), disease relapse has replaced transplant-related mortality as the primary cause of treatment failure for patients with acute myeloid leukemia (AML). The efficacy of allo-HCT in AML is a consequence of a graft-versus-leukemia (GVL) effect that is mediated by T lymphocytes, and unique mechanisms of immune evasion underlying post-allo-HCT AML relapses have recently been characterized. Relapsed AML following allo-HCT presents a particularly vexing clinical challenge because transplant-related toxicities, such as graft-versus-host (GVHD) and infections, increase the risk of treatment-related morbidity and mortality. In general, the prognosis of relapsed AML following allo-HCT is poor with most patients failing to achieve a subsequent remission and 2-year survival consistently <15%. The two factors that have been found to predict a better prognosis are a longer duration of post-transplant remission prior to relapse and a lower disease burden at the time of relapse. When considered in combination with a patient's age; co-morbidities; and performance status, these factors can help to inform the appropriate therapy for the treatment of post-transplant relapse. This review discusses the options for the treatment of post-transplant AML relapse with a focus on the options to achieve a subsequent remission and consolidation with cellular immunotherapy, such as a second transplant or donor lymphocyte infusion (DLI). While intensive reinduction therapy and less intensive approaches with hypomethylating agents have long represented the two primary options for the initial treatment of post-transplant relapse, molecularly targeted therapies and immunotherapy are emerging as potential alternative options to achieve remission. Herein, we highlight response and survival outcomes achieved specifically in the post-transplant setting using each of these approaches and discuss how some therapies may overcome the immunologic mechanisms that have been implicated in post-transplant relapse. As long-term survival in post-transplant relapse necessarily involves consolidation with cellular immunotherapy, we present data on the efficacy and toxicity of both DLI and second allo-HCT including when such therapies are integrated with reinduction. Finally, we provide our general approach to the treatment of post-transplant relapse, integrating both novel therapies and our improved understanding of the mechanisms underlying post-transplant relapse.
Collapse
Affiliation(s)
- Jonathan A Webster
- Hematologic Malignancies and Bone Marrow Transplantation Program, Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Leo Luznik
- Hematologic Malignancies and Bone Marrow Transplantation Program, Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Ivana Gojo
- Hematologic Malignancies and Bone Marrow Transplantation Program, Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| |
Collapse
|
95
|
Penter L, Gohil SH, Wu CJ. Natural Barcodes for Longitudinal Single Cell Tracking of Leukemic and Immune Cell Dynamics. Front Immunol 2022; 12:788891. [PMID: 35046946 PMCID: PMC8761982 DOI: 10.3389/fimmu.2021.788891] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2021] [Accepted: 12/08/2021] [Indexed: 11/26/2022] Open
Abstract
Blood malignancies provide unique opportunities for longitudinal tracking of disease evolution following therapeutic bottlenecks and for the monitoring of changes in anti-tumor immunity. The expanding development of multi-modal single-cell sequencing technologies affords newer platforms to elucidate the mechanisms underlying these processes at unprecedented resolution. Furthermore, the identification of molecular events that can serve as in-vivo barcodes now facilitate the tracking of the trajectories of malignant and of immune cell populations over time within primary human samples, as these permit unambiguous identification of the clonal lineage of cell populations within heterogeneous phenotypes. Here, we provide an overview of the potential for chromosomal copy number changes, somatic nuclear and mitochondrial DNA mutations, single nucleotide polymorphisms, and T and B cell receptor sequences to serve as personal natural barcodes and review technical implementations in single-cell analysis workflows. Applications of these methodologies include the study of acquired therapeutic resistance and the dissection of donor- and host cellular interactions in the context of allogeneic hematopoietic stem cell transplantation.
Collapse
Affiliation(s)
- Livius Penter
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, United States
- Broad Institute of Massachusetts Institute of Technology and Harvard University, Cambridge, MA, United States
- Harvard Medical School, Boston, MA, United States
- Department of Hematology, Oncology, and Tumorimmunology, Campus Virchow Klinikum, Berlin, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Satyen H. Gohil
- Department of Academic Haematology, University College London Cancer Institute, London, United Kingdom
- Department of Haematology, University College London Hospitals NHS Foundation Trust, London, United Kingdom
| | - Catherine J. Wu
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, United States
- Broad Institute of Massachusetts Institute of Technology and Harvard University, Cambridge, MA, United States
- Harvard Medical School, Boston, MA, United States
- Department of Medicine, Brigham and Women’s Hospital, Boston, MA, United States
| |
Collapse
|
96
|
Pradier A, Cordey S, Zanella MC, Melotti A, Wang S, Mamez AC, Chalandon Y, Masouridi-Levrat S, Kaiser L, Simonetta F, Vu DL. Human pegivirus-1 replication influences NK cell reconstitution after allogeneic haematopoietic stem cell transplantation. Front Immunol 2022; 13:1060886. [PMID: 36713419 PMCID: PMC9876574 DOI: 10.3389/fimmu.2022.1060886] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Accepted: 12/15/2022] [Indexed: 01/12/2023] Open
Abstract
Introduction Human pegivirus-1 (HPgV-1) is a so-called commensal virus for which no known associated organ disease has been found to date. Yet, it affects immune-reconstitution as previously studied in the HIV population, in whom active co-infection with HPgV-1 can modulate T and NK cell activation and differentiation leading to a protective effect against the evolution of the disease. Little is known on the effect of HPgV-1 on immune-reconstitution in allogeneic hematopoietic stem cell transplant (allo-HSCT) recipients, a patient population in which we and others have previously reported high prevalence of HPgV-1 replication. The aim of this study was to compare the immune reconstitution after allo-HSCT among HPgV-1-viremic and HPgV-1-non-viremic patients. Methods Within a cohort study of 40 allo-HSCT patients, 20 allo-HSCT recipients positive in plasma sample for HPgV-1 by rRT-PCR during the first year (1, 3, 6, 12 months) after transplantation were matched with 20 allo-HSCT recipients negative for HPgV-1. T and NK cell reconstitution was monitored by flow cytometry in peripheral blood samples from allo-HSCT recipients at the same time points. Results We observed no significant difference in the absolute number and subsets proportions of CD4 and CD8 T cells between patient groups at any analysed timepoint. We observed a significantly higher absolute number of NK cells at 3 months among HPgV-1-viremic patients. Immunophenotypic analysis showed a significantly higher proportion of CD56bright NK cells mirrored by a reduced percentage of CD56dim NK cells in HPgV-1-positive patients during the first 6 months after allo-HSCT. At 6 months post-allo-HSCT, NK cell phenotype significantly differed depending on HPgV-1, HPgV-1-viremic patients displaying NK cells with lower CD16 and CD57 expression compared with HPgV-1-negative patients. In accordance with their less differentiated phenotype, we detected a significantly reduced expression of granzyme B in NK cells in HPgV-1-viremic patients at 6 months. Discussion Our study shows that HPgV-1-viremic allo-HSCT recipients displayed an impaired NK cell, but not T cell, immune-reconstitution compared with HPgV-1-non-viremic patients, revealing for the first time a potential association between replication of the non-pathogenic HPgV-1 virus and immunomodulation after allo-HSCT.
Collapse
Affiliation(s)
- Amandine Pradier
- Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Division of Haematology, Department of Oncology, Geneva University Hospitals, Geneva, Switzerland
- Translational Research Center for Oncohematology, Department of Medicine and Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Samuel Cordey
- Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Laboratory of virology, Division of Laboratory Medicine, Geneva University Hospitals, Geneva, Switzerland
| | - Marie-Céline Zanella
- Laboratory of virology, Division of Laboratory Medicine, Geneva University Hospitals, Geneva, Switzerland
- Division of Infectious diseases, Geneva University Hospitals, Geneva, Switzerland
| | - Astrid Melotti
- Translational Research Center for Oncohematology, Department of Medicine and Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Sisi Wang
- Translational Research Center for Oncohematology, Department of Medicine and Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Anne-Claire Mamez
- Division of Haematology, Department of Oncology, Geneva University Hospitals, Geneva, Switzerland
| | - Yves Chalandon
- Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Division of Haematology, Department of Oncology, Geneva University Hospitals, Geneva, Switzerland
- Translational Research Center for Oncohematology, Department of Medicine and Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | | | - Laurent Kaiser
- Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Laboratory of virology, Division of Laboratory Medicine, Geneva University Hospitals, Geneva, Switzerland
- Division of Infectious diseases, Geneva University Hospitals, Geneva, Switzerland
- Center for emerging viruses, Geneva University Hospitals, Geneva, Switzerland
| | - Federico Simonetta
- Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Division of Haematology, Department of Oncology, Geneva University Hospitals, Geneva, Switzerland
- Translational Research Center for Oncohematology, Department of Medicine and Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Diem-Lan Vu
- Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Laboratory of virology, Division of Laboratory Medicine, Geneva University Hospitals, Geneva, Switzerland
- Division of Infectious diseases, Geneva University Hospitals, Geneva, Switzerland
- *Correspondence: Diem-Lan Vu, ;
| |
Collapse
|
97
|
Wen XM, Xu ZJ, Jin Y, Xia PH, Ma JC, Qian W, Lin J, Qian J. Association Analyses of TP53 Mutation With Prognosis, Tumor Mutational Burden, and Immunological Features in Acute Myeloid Leukemia. Front Immunol 2021; 12:717527. [PMID: 34745095 PMCID: PMC8566372 DOI: 10.3389/fimmu.2021.717527] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Accepted: 09/27/2021] [Indexed: 12/28/2022] Open
Abstract
Acute myeloid leukemia (AML) is a heterogeneous disease related to a broad spectrum of molecular alterations. The successes of immunotherapies treating solid tumors and a deeper understanding of the immune systems of patients with hematologic malignancies have promoted the development of immunotherapies for the treatment of AML. And high tumor mutational burden (TMB) is an emerging predictive biomarker for response to immunotherapy. However, the association of gene mutation in AML with TMB and immunological features still has not been clearly elucidated. In our study, based on The Cancer Genome Atlas (TCGA) and BeatAML cohorts, 20 frequently mutated genes were found to be covered by both datasets in AML. And TP53 mutation was associated with a poor prognosis, and its mutation displayed exclusiveness with other common mutated genes in both datasets. Moreover, TP53 mutation correlated with TMB and the immune microenvironment. Gene Set Enrichment Analysis (GSEA) showed that TP53 mutation upregulated signaling pathways involved in the immune system. In summary, TP53 mutation is frequently mutated in AML, and its mutation is associated with dismal outcome, TMB, and immunological features, which may serve as a biomarker to predict immune response in AML.
Collapse
Affiliation(s)
- Xiang-Mei Wen
- Laboratory Center, Affiliated People's Hospital of Jiangsu University, Zhenjiang, China.,Zhenjiang Clinical Research Center of Hematology, Affiliated People's Hospital of Jiangsu University, Zhenjiang, China.,The Key Lab of Precision Diagnosis and Treatment in Hematologic Malignancies of Zhenjiang City, Affiliated People's Hospital of Jiangsu University, Zhenjiang, China
| | - Zi-Jun Xu
- Laboratory Center, Affiliated People's Hospital of Jiangsu University, Zhenjiang, China.,Zhenjiang Clinical Research Center of Hematology, Affiliated People's Hospital of Jiangsu University, Zhenjiang, China.,The Key Lab of Precision Diagnosis and Treatment in Hematologic Malignancies of Zhenjiang City, Affiliated People's Hospital of Jiangsu University, Zhenjiang, China
| | - Ye Jin
- Zhenjiang Clinical Research Center of Hematology, Affiliated People's Hospital of Jiangsu University, Zhenjiang, China.,Department of Hematology, Affiliated People's Hospital of Jiangsu University, Zhenjiang, China
| | - Pei-Hui Xia
- Zhenjiang Clinical Research Center of Hematology, Affiliated People's Hospital of Jiangsu University, Zhenjiang, China.,Department of Hematology, Affiliated People's Hospital of Jiangsu University, Zhenjiang, China
| | - Ji-Chun Ma
- Laboratory Center, Affiliated People's Hospital of Jiangsu University, Zhenjiang, China.,Zhenjiang Clinical Research Center of Hematology, Affiliated People's Hospital of Jiangsu University, Zhenjiang, China.,The Key Lab of Precision Diagnosis and Treatment in Hematologic Malignancies of Zhenjiang City, Affiliated People's Hospital of Jiangsu University, Zhenjiang, China
| | - Wei Qian
- Department of Otolaryngology-Head and Neck Surgery, Affiliated People's Hospital of Jiangsu University, Zhenjiang, China
| | - Jiang Lin
- Laboratory Center, Affiliated People's Hospital of Jiangsu University, Zhenjiang, China.,Zhenjiang Clinical Research Center of Hematology, Affiliated People's Hospital of Jiangsu University, Zhenjiang, China.,The Key Lab of Precision Diagnosis and Treatment in Hematologic Malignancies of Zhenjiang City, Affiliated People's Hospital of Jiangsu University, Zhenjiang, China
| | - Jun Qian
- Zhenjiang Clinical Research Center of Hematology, Affiliated People's Hospital of Jiangsu University, Zhenjiang, China.,Department of Hematology, Affiliated People's Hospital of Jiangsu University, Zhenjiang, China
| |
Collapse
|
98
|
Li Y, Wu D, Yang X, Zhou S. Immunotherapeutic Potential of T Memory Stem Cells. Front Oncol 2021; 11:723888. [PMID: 34604060 PMCID: PMC8485052 DOI: 10.3389/fonc.2021.723888] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Accepted: 08/23/2021] [Indexed: 11/13/2022] Open
Abstract
Memory T cells include T memory stem cells (TSCM) and central memory T cells (TCM). Compared with effector memory T cells (TEM) and effector T cells (TEFF), they have better durability and anti-tumor immunity. Recent studies have shown that although TSCM has excellent self-renewal ability and versatility, if it is often exposed to antigens and inflammatory signals, TSCM will behave as a variety of inhibitory receptors such as PD-1, TIM-3 and LAG-3 expression, and metabolic changes from oxidative phosphorylation to glycolysis. These changes can lead to the exhaustion of T cells. Cumulative evidence in animal experiments shows that it is the least differentiated cell in the memory T lymphocyte system and is a central participant in many physiological and pathological processes in humans. It has a good clinical application prospect, so it is more and more important to study the factors affecting the formation of TSCM. This article summarizes and prospects the phenotypic and functional characteristics of TSCM, the regulation mechanism of formation, and its application in treatment of clinical diseases.
Collapse
Affiliation(s)
- Yujie Li
- Department of Biochemistry and Molecular Biology, School of Pre-Clinical Science, Guangxi Medical University, Nanning, China
| | - Dengqiang Wu
- National Center for International Research of Bio-targeting Theranostics, Guangxi Key Laboratory of Bio-targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, China
| | - Xuejia Yang
- National Center for International Research of Bio-targeting Theranostics, Guangxi Key Laboratory of Bio-targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, China
| | - Sufang Zhou
- Department of Biochemistry and Molecular Biology, School of Pre-Clinical Science, Guangxi Medical University, Nanning, China.,National Center for International Research of Bio-targeting Theranostics, Guangxi Key Laboratory of Bio-targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, China
| |
Collapse
|
99
|
Kunadt D, Stölzel F. Effective Immunosurveillance After Allogeneic Hematopoietic Stem Cell Transplantation in Acute Myeloid Leukemia. Cancer Manag Res 2021; 13:7411-7427. [PMID: 34594134 PMCID: PMC8478160 DOI: 10.2147/cmar.s261721] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Accepted: 09/15/2021] [Indexed: 12/25/2022] Open
Abstract
The number of patients receiving allogeneic hematopoietic stem cell transplantation (alloHCT) has increased constantly over the last years due to advances in transplant technology development, supportive care, transplant safety, and donor availability. Currently, acute myeloid leukemia (AML) is the most frequent indication for alloHCT. However, disease relapse remains the main cause of therapy failure. Therefore, concepts of maintaining and, if necessary, reinforcing a strong graft-versus-leukemia (GvL) effect is crucial for the prognosis and long-term survival of the patients. Over the last decades, it has become evident that effective immunosurveillance after alloHCT is an entangled complex of donor-specific characteristics, leukemia-associated geno- and phenotypes, and acquired resistance mechanisms. Furthermore, adoption of effector cells such as natural killer (NK) cells, alloreactive and regulatory T-cells with their accompanying receptor repertoire, and cell–cell interactions driven by messenger molecules within the stem cell and the bone marrow niche have important impact. In this review of pre- and posttransplant elements and mechanisms of immunosurveillance, we highlight the most important mechanisms after alloHCT.
Collapse
Affiliation(s)
- Desiree Kunadt
- Department of Internal Medicine I, University Hospital Carl Gustav Carus, Technical University of Dresden, Dresden, Germany
| | - Friedrich Stölzel
- Department of Internal Medicine I, University Hospital Carl Gustav Carus, Technical University of Dresden, Dresden, Germany
| |
Collapse
|
100
|
Paving the Way for Immunotherapy in Pediatric Acute Myeloid Leukemia: Current Knowledge and the Way Forward. Cancers (Basel) 2021; 13:cancers13174364. [PMID: 34503174 PMCID: PMC8431730 DOI: 10.3390/cancers13174364] [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: 07/26/2021] [Revised: 08/22/2021] [Accepted: 08/26/2021] [Indexed: 11/17/2022] Open
Abstract
Simple Summary Immunotherapy may be an attractive treatment option to increase survival, and to reduce treatment-related side effects, for children with acute myeloid leukemia (AML). While immunotherapies have shown successes in many cancer types, the development and subsequent clinical implementation have proven difficult in pediatric AML. To expedite the development of immunotherapy, it will be crucial to understand which pediatric AML patients are likely to respond to immunotherapies. Emerging research in solid malignancies has shown that the number and phenotype of immune cells in the tumor microenvironment is predictive of response to several types of immunotherapies. Such a predictive model may also be applicable for AML and, thus, knowledge on the immune cells infiltrating the bone marrow environment is needed. Here, we discuss the current state of knowledge on these infiltrating immune cells in pediatric AML, as well as ongoing immunotherapy trials, and provide suggestions concerning the way forward. Abstract Immunotherapeutic agents may be an attractive option to further improve outcomes and to reduce treatment-related toxicity for pediatric AML. While improvements in outcome have been observed with immunotherapy in many cancer types, immunotherapy development and implementation into patient care for both adult and pediatric AML has been hampered by an incomplete understanding of the bone marrow environment and a paucity of tumor-specific antigens. Since only a minority of patients respond in most immunotherapy trials across different cancer types, it will be crucial to understand which children with AML are likely to respond to or may benefit from immunotherapies. Immune cell profiling efforts hold promise to answer this question, as illustrated by the development of predictive scores in solid cancers. Such information on the number and phenotype of immune cells during current treatment regimens will be pivotal to generate hypotheses on how and when to intervene with immunotherapy in pediatric AML. In this review, we discuss the current understanding of the number and phenotype of immune cells in the bone marrow in pediatric AML, ongoing immunotherapy trials and how comprehensive immune profiling efforts may pave the way for successful clinical trials (and, ultimately, implementation into patient care).
Collapse
|