1
|
Tassaneetrithep B, Phuphuakrat A, Pasomsub E, Bhukhai K, Wongkummool W, Priengprom T, Khamaikawin W, Chaisavaneeyakorn S, Anurathapan U, Apiwattanakul N, Hongeng S. HIV-1 proviral DNA in purified peripheral blood CD34 + stem and progenitor cells in individuals with long-term HAART; paving the way to HIV gene therapy. Heliyon 2024; 10:e26613. [PMID: 38434025 PMCID: PMC10906414 DOI: 10.1016/j.heliyon.2024.e26613] [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: 08/04/2023] [Revised: 02/03/2024] [Accepted: 02/16/2024] [Indexed: 03/05/2024] Open
Abstract
Human immunodeficiency virus (HIV)-1 infection is an important public health problem worldwide. After primary HIV-1 infection, transcribed HIV-1 DNA is integrated into the host genome, serving as a reservoir of the virus and hindering a definite cure. Although highly active antiretroviral therapy suppresses active viral replication, resulting in undetectable levels of HIV RNA in the blood, a viral rebound can be detected after a few weeks of treatment interruption. This supports the concept that there is a stable HIV-1 reservoir in people living with HIV-1. Recently, a few individuals with HIV infection were reported to be probably cured by hematopoietic stem transplantation (HSCT). The underlying mechanism for this success involved transfusion of uninfected hematopoietic stem and progenitor cells (HSPCs) from CCR5-mutated donors who were naturally resistant to HIV infection. Thus, gene editing technology to provide HIV-resistant HSPC has promise in the treatment of HIV infections by HSCT. In this study, we aimed to find HIV-infected individuals likely to achieve a definite cure via gene editing HSCT. We screened for total HIV proviral DNA by Alu PCR in peripheral blood mononuclear cells (PBMCs) of 20 HIV-infected individuals with prolonged viral suppression. We assessed the amount of intact proviral DNA via a modified intact proviral DNA assay (IPDA) in purified peripheral CD34+ HSPCs. PBMCs from all 20 individuals were positive for the gag gene in Alu PCR, and peripheral CD34+ HSPCs were IPDA-negative for six individuals. Our results suggested that these six HIV-infected individuals could be candidates for further studies into the ability of gene editing HSCT to lead to a definite HIV cure.
Collapse
Affiliation(s)
- Boonrat Tassaneetrithep
- Center of Research Excellence in Immunoregulation, Faculty of Medicine Siriraj Hospital, Mahidol University, Thailand
| | - Angsana Phuphuakrat
- Department of Medicine, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Thailand
| | - Ekawat Pasomsub
- Department of Pathology, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Thailand
| | - Kanit Bhukhai
- Department of Physiology, Faculty of Science, Mahidol University, Thailand
| | | | - Thongkoon Priengprom
- Center of Research Excellence in Immunoregulation, Faculty of Medicine Siriraj Hospital, Mahidol University, Thailand
| | - Wannisa Khamaikawin
- Faculty of Medicine, King Mongkut's Institute of Technology Ladkrabang, Thailand
| | | | - Usanarat Anurathapan
- Department of Pediatrics, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Thailand
| | - Nopporn Apiwattanakul
- Department of Pediatrics, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Thailand
| | - Suradej Hongeng
- Department of Pediatrics, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Thailand
| |
Collapse
|
2
|
Ash S, Askenasy N. Immunotherapy for neuroblastoma by hematopoietic cell transplantation and post-transplant immunomodulation. Crit Rev Oncol Hematol 2023; 185:103956. [PMID: 36893946 DOI: 10.1016/j.critrevonc.2023.103956] [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: 06/04/2021] [Revised: 12/14/2022] [Accepted: 03/04/2023] [Indexed: 03/09/2023] Open
Abstract
Neuroblastoma represents a relatively common childhood tumor that imposes therapeutic difficulties. High risk neuroblastoma patients have poor prognosis, display limited response to radiochemotherapy and may be treated by hematopoietic cell transplantation. Allogeneic and haploidentical transplants have the distinct advantage of reinstitution of immune surveillance, reinforced by antigenic barriers. The key factors favorable to ignition of potent anti-tumor reactions are transition to adaptive immunity, recovery from lymphopenia and removal of inhibitory signals that inactivate immune cells at the local and systemic levels. Post-transplant immunomodulation may further foster anti-tumor reactivity, with positive but transient impact of infusions of lymphocytes and natural killer cells both from the donor, the recipient or third party. The most promising approaches include introduction of antigen-presenting cells in early post-transplant stages and neutralization of inhibitory signals. Further studies will likely shed light on the nature and actions of suppressor factors within tumor stroma and at the systemic level.
Collapse
Affiliation(s)
- Shifra Ash
- Department of Pediatric Hematology-Oncology, Rambam Medical Center, Haifa, Israel; Frankel Laboratory of Bone Marrow Transplantation, Schneider Children's Medical Center of Israel, Petach Tikva, Israel.
| | - Nadir Askenasy
- Frankel Laboratory of Bone Marrow Transplantation, Schneider Children's Medical Center of Israel, Petach Tikva, Israel
| |
Collapse
|
3
|
Systemic Treatment of Ewing Sarcoma: Current Options and Future Perspectives. FORUM OF CLINICAL ONCOLOGY 2022. [DOI: 10.2478/fco-2021-0005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Abstract
Ewing sarcoma (ES) is an uncommon malignant neoplasm, mostly affecting young adults and adolescents. Surgical excision, irradiation, and combinations of multiple chemotherapeutic agents are currently used as a multimodal strategy for the treatment of local and oligometastatic disease. Although ES usually responds to the primary treatment, relapsed and primarily refractory disease remains a difficult therapeutic challenge. The growing understanding of cancer biology and the subsequent development of new therapeutic strategies have been put at the service of research in recurrent and refractory ES, generating a great number of ongoing studies with compounds that could find superior clinical outcomes in the years to come. This review gathers the current available information on the treatment and clinical investigation of ES and aims to be a point of support for future research.
Collapse
|
4
|
Multiple Reactivations of Viral Infections Followed by Cerebral Toxoplasmosis After Allogeneic Hematopoietic Stem Cell Transplantation in an Adolescent With Ph(+) Acute Lymphoblastic Leukemia: A Case Report. Transplant Proc 2021; 53:1355-1359. [PMID: 33785195 DOI: 10.1016/j.transproceed.2021.02.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 02/05/2021] [Indexed: 11/22/2022]
Abstract
After allogeneic hematopoietic stem cell transplantation (allo-HSCT), especially from an unrelated donor, infectious complications are frequent and severe, sometimes with fatal outcomes. Despite using highly sensitive molecular techniques for close monitoring in the early post-transplant period for early diagnosis, not every viral infection or reactivation can be detected adequately early, even with highly sensitive methods. Particularly after toxic and deeply immunosuppressive treatment, multiple infections or reactivations, uncommon infections, or infections in unusual locations can occur. Here, we present a case of multiple viral infections or reactivations and cerebral toxoplasmosis in a 17-year-old youth with Philadelphia chromosome-positive acute lymphoblastic leukemia (Ph+ ALL) treated with allo-HSCT who suffered multiple viral infections followed by cerebral toxoplasmosis.
Collapse
|
5
|
Şahin U, Demirer T. Graft-versus-cancereffect and innovative approaches in thetreatment of refractory solid tumors. Turk J Med Sci 2020; 50:1697-1706. [PMID: 32178508 PMCID: PMC7672351 DOI: 10.3906/sag-1911-112] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Accepted: 03/14/2020] [Indexed: 12/23/2022] Open
Abstract
Background/aim Allogeneic hematopoietic stem cell transplantation (allo-HSCT) has been used for the treatment of various refractory solid tumors during the last two decades. After the demonstration of graft-versus-leukemia (GvL) effect in a leukemic murine model following allo-HSCT from other strains of mice, graft-versus-tumor (GvT) effect in a solid tumor after allo-HSCT has also been reported in a murine model in 1984. Several trials have reported the presence of a GvT effect in patients with various refractory solid tumors, including renal, ovarian and colon cancers, as well as soft tissue sarcomas [1]. The growing data on haploidentical transplants also indicate GvT effect in some pediatric refractory solid tumors. Novel immunotherapy-based treatment modalities aim at inducing an allo-reactivity against the metastatic solid tumor via a GvT effect. Recipient derived immune effector cells (RDICs) in the antitumor reactivity following allo-HSCT have also been considered as an emerging therapy for advanced refractory solid tumors. Conclusion This review summarizes the background, rationale, and clinical results of immune-based strategies using GvT effect for the treatment of various metastatic and refractory solid tumors, as well as innovative approaches such as haploidentical HSCT, CAR-T cell therapies and tumor infiltrating lymphocytes (TIL).
Collapse
Affiliation(s)
- Uğur Şahin
- Hematology Unit, Yenimahalle Education and Research Hospital, Yıldırım Beyazıt University, Ankara, Turkey
| | - Taner Demirer
- Department of Hematology, School of Medicine, Ankara University, Ankara, Turkey
| |
Collapse
|
6
|
Mondino A, Manzo T. To Remember or to Forget: The Role of Good and Bad Memories in Adoptive T Cell Therapy for Tumors. Front Immunol 2020; 11:1915. [PMID: 32973794 PMCID: PMC7481451 DOI: 10.3389/fimmu.2020.01915] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Accepted: 07/16/2020] [Indexed: 12/17/2022] Open
Abstract
The generation of immunological memory is a hallmark of adaptive immunity by which the immune system "remembers" a previous encounter with an antigen expressed by pathogens, tumors, or normal tissues; and, upon secondary encounters, mounts faster and more effective recall responses. The establishment of T cell memory is influenced by both cell-intrinsic and cell-extrinsic factors, including genetic, epigenetic and environmental triggers. Our current knowledge of the mechanisms involved in memory T cell differentiation has instructed new opportunities to engineer T cells with enhanced anti-tumor activity. The development of adoptive T cell therapy has emerged as a powerful approach to cure a subset of patients with advanced cancers. Efficacy of this approach often requires long-term persistence of transferred T cell products, which can vary according to their origin and manufacturing conditions. Host preconditioning and post-transfer supporting strategies have shown to promote their engraftment and survival by limiting the competition with a hostile tumor microenvironment and between pre-existing immune cell subsets. Although in the general view pre-existing memory can confer a selective advantage to adoptive T cell therapy, here we propose that also "bad memories"-in the form of antigen-experienced T cell subsets-co-evolve with consequences on newly transferred lymphocytes. In this review, we will first provide an overview of selected features of memory T cell subsets and, then, discuss their putative implications for adoptive T cell therapy.
Collapse
Affiliation(s)
- Anna Mondino
- Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Teresa Manzo
- Department of Experimental Oncology, IRCCS European Institute of Oncology, Milan, Italy
| |
Collapse
|
7
|
Yu D, Wang L, Wu T, Zhang Y, Tian Y, Wang Y, Cui C, Li H, Zhang J, Zhou L, Yan S, Zheng S. Graft-Versus-Tumor Effect in Major Histocompatibility Complex-Mismatched Mouse Liver Transplantation. Liver Transpl 2019; 25:1251-1264. [PMID: 31152624 PMCID: PMC6771797 DOI: 10.1002/lt.25574] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Accepted: 05/16/2019] [Indexed: 12/21/2022]
Abstract
Liver transplantation (LT) is currently considered an important method in treating hepatocellular carcinoma (HCC) and an alternative treatment for other liver malignancies. Here, we demonstrated that the graft-versus-tumor (GVT) effect exists in allogeneic liver transplantation (allo LT). Recipient-derived T cells played a critical role in the GVT process of allo LT, as demonstrated by extensive infiltration and significant activation of recipient T cells in the tumor after surgery. Moreover, this process was related to donor-derived T/B cells by improving the immune microenvironment in the tumor, as demonstrated by elevated levels of interferon-γ (IFN-γ), tumor necrosis factor-α (TNF-α), interleukin-2 (IL-2), IL-6, IL-16, chemokine (C-X-C motif) ligand 10 (CXCL10), and CXCL11 and decreased levels of IL-10 and IL-4 at tumor sites. Additionally, tacrolimus (FK506) treatment inhibited the GVT effect on allo LT. Donor liver-derived T/B cells infiltrate extrahepatic tumors to trigger a strong T-cell-mediated immune response and thus improve the tumor immune microenvironment.
Collapse
Affiliation(s)
- Dongdong Yu
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated HospitalZhejiang UniversityHangzhouChina,National Health and Family Planning Commission of China Key Laboratory of Combined Multi-Organ TransplantationHangzhouChina,Key Laboratory of the Diagnosis and Treatment of Organ TransplantationCAMSHangzhouChina,Key Laboratory of Organ TransplantationHangzhouChina,Collaborative Innovation Center for Diagnosis Treatment of Infectious DiseaseHangzhouChina
| | - Lidong Wang
- Department of Hepatobiliary and Pancreatic SurgeryShulan (Hangzhou) HospitalHangzhouChina
| | - Tianchun Wu
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated HospitalZhejiang UniversityHangzhouChina,National Health and Family Planning Commission of China Key Laboratory of Combined Multi-Organ TransplantationHangzhouChina,Key Laboratory of the Diagnosis and Treatment of Organ TransplantationCAMSHangzhouChina,Key Laboratory of Organ TransplantationHangzhouChina,Collaborative Innovation Center for Diagnosis Treatment of Infectious DiseaseHangzhouChina
| | - Yaohui Zhang
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated HospitalZhejiang UniversityHangzhouChina,National Health and Family Planning Commission of China Key Laboratory of Combined Multi-Organ TransplantationHangzhouChina,Key Laboratory of the Diagnosis and Treatment of Organ TransplantationCAMSHangzhouChina,Key Laboratory of Organ TransplantationHangzhouChina,Collaborative Innovation Center for Diagnosis Treatment of Infectious DiseaseHangzhouChina
| | - Yang Tian
- National Health and Family Planning Commission of China Key Laboratory of Combined Multi-Organ TransplantationHangzhouChina,Key Laboratory of the Diagnosis and Treatment of Organ TransplantationCAMSHangzhouChina,Key Laboratory of Organ TransplantationHangzhouChina,Collaborative Innovation Center for Diagnosis Treatment of Infectious DiseaseHangzhouChina,Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, Second Affiliated Hospital, School of MedicineZhejiang UniversityHangzhouChina
| | - Yan Wang
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated HospitalZhejiang UniversityHangzhouChina,National Health and Family Planning Commission of China Key Laboratory of Combined Multi-Organ TransplantationHangzhouChina,Key Laboratory of the Diagnosis and Treatment of Organ TransplantationCAMSHangzhouChina,Key Laboratory of Organ TransplantationHangzhouChina,Collaborative Innovation Center for Diagnosis Treatment of Infectious DiseaseHangzhouChina
| | - Chenwei Cui
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated HospitalZhejiang UniversityHangzhouChina,Key Laboratory of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Tumor of Zhejiang ProvinceHangzhouChina
| | - Hui Li
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated HospitalZhejiang UniversityHangzhouChina,National Health and Family Planning Commission of China Key Laboratory of Combined Multi-Organ TransplantationHangzhouChina,Key Laboratory of the Diagnosis and Treatment of Organ TransplantationCAMSHangzhouChina,Key Laboratory of Organ TransplantationHangzhouChina,Collaborative Innovation Center for Diagnosis Treatment of Infectious DiseaseHangzhouChina
| | - Jinhua Zhang
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated HospitalZhejiang UniversityHangzhouChina,National Health and Family Planning Commission of China Key Laboratory of Combined Multi-Organ TransplantationHangzhouChina,Key Laboratory of the Diagnosis and Treatment of Organ TransplantationCAMSHangzhouChina,Key Laboratory of Organ TransplantationHangzhouChina,Collaborative Innovation Center for Diagnosis Treatment of Infectious DiseaseHangzhouChina
| | - Lin Zhou
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated HospitalZhejiang UniversityHangzhouChina,National Health and Family Planning Commission of China Key Laboratory of Combined Multi-Organ TransplantationHangzhouChina,Key Laboratory of the Diagnosis and Treatment of Organ TransplantationCAMSHangzhouChina,Key Laboratory of Organ TransplantationHangzhouChina,Collaborative Innovation Center for Diagnosis Treatment of Infectious DiseaseHangzhouChina
| | - Sheng Yan
- National Health and Family Planning Commission of China Key Laboratory of Combined Multi-Organ TransplantationHangzhouChina,Key Laboratory of the Diagnosis and Treatment of Organ TransplantationCAMSHangzhouChina,Key Laboratory of Organ TransplantationHangzhouChina,Collaborative Innovation Center for Diagnosis Treatment of Infectious DiseaseHangzhouChina,Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, Second Affiliated Hospital, School of MedicineZhejiang UniversityHangzhouChina
| | - Shusen Zheng
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated HospitalZhejiang UniversityHangzhouChina,National Health and Family Planning Commission of China Key Laboratory of Combined Multi-Organ TransplantationHangzhouChina,Key Laboratory of the Diagnosis and Treatment of Organ TransplantationCAMSHangzhouChina,Key Laboratory of Organ TransplantationHangzhouChina,Collaborative Innovation Center for Diagnosis Treatment of Infectious DiseaseHangzhouChina
| |
Collapse
|
8
|
Duarte RF, Labopin M, Bader P, Basak GW, Bonini C, Chabannon C, Corbacioglu S, Dreger P, Dufour C, Gennery AR, Kuball J, Lankester AC, Lanza F, Montoto S, Nagler A, Peffault de Latour R, Snowden JA, Styczynski J, Yakoub-Agha I, Kröger N, Mohty M. Indications for haematopoietic stem cell transplantation for haematological diseases, solid tumours and immune disorders: current practice in Europe, 2019. Bone Marrow Transplant 2019; 54:1525-1552. [PMID: 30953028 DOI: 10.1038/s41409-019-0516-2] [Citation(s) in RCA: 197] [Impact Index Per Article: 39.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Revised: 03/05/2019] [Accepted: 03/07/2019] [Indexed: 12/20/2022]
Abstract
This is the seventh special EBMT report on the indications for haematopoietic stem cell transplantation for haematological diseases, solid tumours and immune disorders. Our aim is to provide general guidance on transplant indications according to prevailing clinical practice in EBMT countries and centres. In order to inform patient decisions, these recommendations must be considered together with the risk of the disease, the risk of the transplant procedure and the results of non-transplant strategies. In over two decades since the first report, the EBMT indications manuscripts have incorporated changes in transplant practice coming from scientific and technical developments in the field. In this same period, the establishment of JACIE accreditation has promoted high quality and led to improved outcomes of patient and donor care and laboratory performance in transplantation and cellular therapy. An updated report with operating definitions, revised indications and an additional set of data with overall survival at 1 year and non-relapse mortality at day 100 after transplant in the commonest standard-of-care indications is presented. Additional efforts are currently underway to enable EBMT member centres to benchmark their risk-adapted outcomes as part of the Registry upgrade Project 2020 against national and/or international outcome data.
Collapse
Affiliation(s)
- Rafael F Duarte
- Hospital Universitario Puerta de Hierro Majadahonda - Universidad Autónoma de Madrid, Madrid, Spain.
| | - Myriam Labopin
- EBMT Paris Study Office, Hopital Saint Antoine, Paris, France
| | - Peter Bader
- Goethe University Hospital, Frankfurt/Main, Germany
| | | | - Chiara Bonini
- Vita-Salute San Raffaele University & Ospedale San Raffaele Scientific Institute, Milan, Italy
| | - Christian Chabannon
- Institut Paoli Calmettes & Centre d'Investigations Cliniques en Biothérapies, Marseille, France
| | | | - Peter Dreger
- Medizinische Klinik V, Universität Heidelberg, Heidelberg, Germany
| | - Carlo Dufour
- Giannina Gaslini Children's Hospital, Genoa, Italy
| | | | - Jürgen Kuball
- University Medical Center Utrecht, Utrecht, The Netherlands
| | - Arjan C Lankester
- Willem-Alexander Children's Hospital, Leiden University Medical Center, Leiden, The Netherlands
| | | | | | - Arnon Nagler
- Chaim Sheva Medical Center, Tel-Hashomer, Israel
| | | | - John A Snowden
- Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, United Kingdom
| | - Jan Styczynski
- Collegium Medicum, Nicolaus Copernicus University, Bydgoszcz, Poland
| | | | | | - Mohamad Mohty
- Hopital Saint Antoine, Sorbonne Université, Paris, France
| | | |
Collapse
|
9
|
The mechanistic study behind suppression of GVHD while retaining GVL activities by myeloid-derived suppressor cells. Leukemia 2019; 33:2078-2089. [PMID: 30737483 PMCID: PMC6687551 DOI: 10.1038/s41375-019-0394-z] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2018] [Revised: 01/09/2019] [Accepted: 01/18/2019] [Indexed: 12/25/2022]
Abstract
Graft-versus-host disease (GVHD) is a major barrier to the widespread use of allogeneic hematopoietic stem cell transplantation (allo-HSCT) for treating hematologic malignancies. Myeloid-derived suppressor cells (MDSCs) have been recognized as crucial immunosuppressive cells in various pathologic settings. Here, we investigated whether the unique functional properties of MDSCs could be harnessed to control allo-HSCT-associated GVHD. Using multiple murine GVHD/GVL models including both MHC-mismatched and miHA-mismatched, we demonstrated that treatment with CD115+ MDSCs efficiently suppressed GVHD but did not significantly impair graft-versus-leukemia (GVL) activity, leading to 80% and 67% protection in treated mice in GVHD and GVL models, respectively. The mechanism for this dissociation of GVHD from GVL, specifically the emergence of donor-derived NKG2D+ CD8 T cells with a memory phenotype in MDSC-treated recipient mice, was identified. NKG2D expression on donor T cells was required for eradication of allogeneic lymphoma cells. Furthermore, long-term surviving MDSC recipients that exhibited cytolytic activities against allogeneic leukemia cells had a significantly increased percentage of T regulatory cells and, more importantly, NKG2D+ CD8 T cells. These findings indicate that MDSCs can be used as a novel cell-based therapy to suppress GVHD while maintaining GVL activities through selective induction of NKG2D+ CD8 memory T cells.
Collapse
|
10
|
Pre-immunization of donor lymphocytes with GITR agonistic antibody enhances antitumor immunity in autologous hematopoietic stem cell transplantation. Biochem Biophys Res Commun 2019; 509:96-101. [PMID: 30579597 DOI: 10.1016/j.bbrc.2018.12.069] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Accepted: 12/11/2018] [Indexed: 01/24/2023]
Abstract
The lymphopenic condition following autologous hematopoietic stem cell transplantation (HSCT) enhances the proliferation of T cells by engaging tumor-associated antigens, leading to the alteration of the T-cell repertoire towards antitumor immunity. However, cure by autologous HSCT alone have rarely occurred in the clinical setting. Since tumor-reactive lymphocytes preferentially proliferate during reconstitution of the immune system, we examined whether the priming of donor lymphocytes can strengthen the antitumor effect by HSCT in a CT26 murine colon cancer model. The systemic administration of an anti-glucocorticoid-induced TNF receptor (GITR) agonistic antibody (Ab) significantly increased the number of CT26-responsive T cells but not that of auto-reactive lymphocytes in donor mice. The infusion of non-primed and GITR Ab-primed donor lymphocytes suppressed the CT26 tumor growth, and only the primed lymphocytes eliminated tumors in all the treated mice. The frequency of CT26-responsive T cells was elevated in recipient mice infused with both primed and non-primed lymphocytes until 4 weeks after transplantation, while the frequency in recipients with primed lymphocytes was markedly elevated compared with that in mice harboring non-primed lymphocytes at 2 weeks. The frequencies of regulatory T cells and myeloid-derived suppressor cells were elevated in recipient mice infused with primed and non-primed lymphocytes 2 weeks after transplantation, and returned to normal levels by week 4. The combination of autologous HSCT with pre-immunization of donor lymphocytes is a promising strategy to induce strong antitumor immunity.
Collapse
|
11
|
Dierckx de Casterlé I, Billiau AD, Sprangers B. Recipient and donor cells in the graft-versus-solid tumor effect: It takes two to tango. Blood Rev 2018; 32:449-456. [PMID: 29678553 DOI: 10.1016/j.blre.2018.04.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2017] [Revised: 03/05/2018] [Accepted: 04/06/2018] [Indexed: 12/16/2022]
Abstract
Allogeneic hematopoietic stem cell transplantation (alloHSCT) produces -similar to the long-established graft-versus-leukemia effect- graft-versus-solid-tumor effects. Clinical trials reported response rates of up to 53%, occurring mostly but not invariably in association with full donor chimerism and/or graft-versus-host disease. Although donor-derived T cells are considered the principal effectors of anti-tumor immunity after alloHSCT or donor leukocyte infusion (DLI), growing evidence indicate that recipient-derived immune cells may also contribute. Whereas the role of recipient-derived antigen-presenting cells in eliciting graft-versus-host reactions and priming donor T cells following DLI is well known, resulting inflammatory responses may also break tolerance of recipient effector cells towards the tumor. Additionally, mouse studies indicated that post-transplant recipient leukocyte infusion produces anti-leukemia and anti-solid-tumor effects that were exclusively mediated by recipient-type effector cells, without graft-versus-host disease. Here, we review current preclinical and clinical evidence on graft-versus-solid-tumor effects and growing evidence on the effector role of recipient-derived immune cells in the anti-tumor effect of alloHSCT.
Collapse
Affiliation(s)
- Isabelle Dierckx de Casterlé
- Department of Microbiology and Immunology, Laboratory of Experimental Transplantation, KU Leuven, Herestraat 49, 3000 Leuven, Belgium.
| | - An D Billiau
- Department of Microbiology and Immunology, Laboratory of Experimental Transplantation, KU Leuven, Herestraat 49, 3000 Leuven, Belgium.
| | - Ben Sprangers
- Department of Microbiology and Immunology, Laboratory of Experimental Transplantation, KU Leuven, Herestraat 49, 3000 Leuven, Belgium; Department of Nephrology, University Hospitals Leuven, Herestraat 49, 3000 Leuven, Belgium.
| |
Collapse
|
12
|
Dierckx de Casterlé I, Fevery S, Rutgeerts O, Poosti F, Struyf S, Lenaerts C, Waer M, Billiau AD, Sprangers B. Reduction of myeloid-derived suppressor cells reinforces the anti-solid tumor effect of recipient leukocyte infusion in murine neuroblastoma-bearing allogeneic bone marrow chimeras. Cancer Immunol Immunother 2018; 67:589-603. [PMID: 29299660 PMCID: PMC11028213 DOI: 10.1007/s00262-017-2114-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Accepted: 12/28/2017] [Indexed: 12/23/2022]
Abstract
Allogeneic hematopoietic stem cell transplantation is an emerging treatment option for solid tumors because of its capacity to elicit immune graft-versus-tumor effects. However, these are often limited and associated with GvHD. Adoptive recipient leukocyte infusion (RLI) was shown to enhance anti-tumor responses of allogeneic bone marrow transplantation in murine neuroblastoma (Neuro2A)-bearing chimeras. In contrast to the clinically used donor leukocyte infusion, the RLI anti-tumor effect-elicited by host-versus-graft lymphohematopoietic reactivity-does not cause GvHD; however, the tumor growth-inhibitory effect is incomplete, because overall survival is not prolonged. Here, we studied the anti-solid tumor mechanisms of RLI with the objective to improve its efficacy. Host-versus-graft reactivity following RLI was associated with a systemic cytokine storm, lymph node DC activation, and systemic expansion of host-derived IFN-γ-expressing CD4+ T cells and IFN-γ-and granzyme B-expressing CD8+ T cells, which acquired killing activity against Neuro2A and third-party tumor cells. The tumor showed up-regulation of MHC class I and a transient accumulation of IFN-γ-and granzyme B-expressing CD8+ T cells: the intra-tumor decline in cytotoxic CD8+ T cells coincided with a systemic-and to a lesser extent intra-tumoral-expansion of MDSC. In vivo MDSC depletion with 5-FU significantly improved the local tumor growth-inhibitory effect of RLI as well as overall survival. In conclusion, the RLI-induced alloreactivity gives rise to a host-derived cytotoxic T-cell anti-neuroblastoma response, but also drives an expansion of host-type MDSC that counteracts the anti-tumor effect. This finding identifies MDSC as a novel target to increase the effectiveness of RLI, and possibly other cancer immunotherapies.
Collapse
Affiliation(s)
- Isabelle Dierckx de Casterlé
- Laboratory of Experimental Transplantation, Department of Microbiology and Immunology, Katholieke Universiteit (KU) Leuven, Herestraat 49, Box 811, 3000, Leuven, Belgium
| | - Sabine Fevery
- Laboratory of Experimental Transplantation, Department of Microbiology and Immunology, Katholieke Universiteit (KU) Leuven, Herestraat 49, Box 811, 3000, Leuven, Belgium
| | - Omer Rutgeerts
- Laboratory of Experimental Transplantation, Department of Microbiology and Immunology, Katholieke Universiteit (KU) Leuven, Herestraat 49, Box 811, 3000, Leuven, Belgium
| | - Fariba Poosti
- Laboratory of Molecular Immunology, Department of Microbiology and Immunology, KU Leuven, Herestraat 49, 3000, Leuven, Belgium
| | - Sofie Struyf
- Laboratory of Molecular Immunology, Department of Microbiology and Immunology, KU Leuven, Herestraat 49, 3000, Leuven, Belgium
| | - Caroline Lenaerts
- Laboratory of Experimental Transplantation, Department of Microbiology and Immunology, Katholieke Universiteit (KU) Leuven, Herestraat 49, Box 811, 3000, Leuven, Belgium
| | - Mark Waer
- Laboratory of Experimental Transplantation, Department of Microbiology and Immunology, Katholieke Universiteit (KU) Leuven, Herestraat 49, Box 811, 3000, Leuven, Belgium
| | - An D Billiau
- Laboratory of Experimental Transplantation, Department of Microbiology and Immunology, Katholieke Universiteit (KU) Leuven, Herestraat 49, Box 811, 3000, Leuven, Belgium
| | - Ben Sprangers
- Laboratory of Experimental Transplantation, Department of Microbiology and Immunology, Katholieke Universiteit (KU) Leuven, Herestraat 49, Box 811, 3000, Leuven, Belgium.
- Department of Nephrology, University Hospitals Leuven, Herestraat 49, 3000, Leuven, Belgium.
| |
Collapse
|
13
|
Nguyen J, Alexander T, Jiang H, Hill N, Abdullaev Z, Pack SD, Hsu AP, Holland SM, Hickstein DD, Engels EA, Brownell I. Melanoma in patients with GATA2 deficiency. Pigment Cell Melanoma Res 2017; 31:337-340. [PMID: 29156497 DOI: 10.1111/pcmr.12671] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Accepted: 11/13/2017] [Indexed: 01/11/2023]
Abstract
GATA2 deficiency is a recently described genetic disorder affecting hematopoietic stem cells and is associated with immunodeficiency, hematologic malignancy, and various cutaneous pathologies including cutaneous tumors. To explore the incidence and clinical course of melanoma in patients with germline GATA2 deficiencies, we conducted a retrospective chart review of 71 such patients and identified two with invasive melanoma. One melanoma was diagnosed early because it was associated with pruritus due to a graft-versus-tumor effect following bone marrow transplantation. The other one, a lentigo maligna melanoma, was locally excised but progressed to widespread metastasis and death several years later. Our observations and published studies of melanoma biology suggest an association between decreased GATA2 expression and melanoma progression. These findings suggest that GATA2 deficient patients may have an increased risk of melanoma and should be observed closely for new or changing skin lesions.
Collapse
Affiliation(s)
- Jannett Nguyen
- Dermatology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Tiffany Alexander
- Dermatology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Hong Jiang
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Natasha Hill
- Dermatology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Zied Abdullaev
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Svetlana D Pack
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Amy P Hsu
- Laboratory of Clinical Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Steven M Holland
- Laboratory of Clinical Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Dennis D Hickstein
- Experimental Transplantation and Immunology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Eric A Engels
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Rockville, MD, USA
| | - Isaac Brownell
- Dermatology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
14
|
Llosa NJ, Cooke KR, Chen AR, Gamper CJ, Klein OR, Zambidis ET, Luber B, Rosner G, Siegel N, Holuba MJ, Robey N, Hayashi M, Jones RJ, Fuchs E, Holdhoff M, Loeb DM, Symons HJ. Reduced-Intensity Haploidentical Bone Marrow Transplantation with Post-Transplant Cyclophosphamide for Solid Tumors in Pediatric and Young Adult Patients. Biol Blood Marrow Transplant 2017; 23:2127-2136. [PMID: 28807769 PMCID: PMC5986177 DOI: 10.1016/j.bbmt.2017.08.012] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2017] [Accepted: 08/09/2017] [Indexed: 11/21/2022]
Abstract
High-risk, recurrent, or refractory solid tumors in pediatric, adolescent, and young adult (AYA) patients have an extremely poor prognosis despite current intensive treatment regimens. We piloted an allogeneic bone marrow transplant platform using reduced-intensity conditioning (RIC) and partially HLA-mismatched (haploidentical) related donors for this population of pediatric and AYA solid tumor patients. Sixteen patients received fludarabine, cyclophosphamide, melphalan, and low-dose total body irradiation RIC haploidentical BMT (haploBMT) followed by post-transplantation cyclophosphamide (PTCy), mycophenolate mofetil, and sirolimus. All assessable patients were full donor chimeras on day 30 with a median neutrophil recovery of 19 days and platelet recovery of 21 days. One patient (7%) exhibited secondary graft failure associated with concomitant infection. The median follow-up time was 15 months. Overall survival was 88%, 56%, and 21% at 6, 12, and 24 months, respectively. Median survival from transplant date was 14 months with a median progression-free survival 7 months. We observed limited graft-versus-host disease in 3 patients and nonrelapse mortality in 1 patient. We demonstrated that RIC haploBMT with PTCy is feasible and has acceptable toxicities in patients with incurable pediatric and AYA solid tumors; thus, this approach serves as a platform for post-transplant strategies to prevent relapse and optimize progression-free survival.
Collapse
Affiliation(s)
- Nicolas J Llosa
- Blood and Marrow Transplantation Program, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins Hospital, Baltimore, Maryland.
| | - Kenneth R Cooke
- Blood and Marrow Transplantation Program, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins Hospital, Baltimore, Maryland
| | - Allen R Chen
- Blood and Marrow Transplantation Program, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins Hospital, Baltimore, Maryland
| | - Christopher J Gamper
- Blood and Marrow Transplantation Program, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins Hospital, Baltimore, Maryland
| | - Orly R Klein
- Blood and Marrow Transplantation Program, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins Hospital, Baltimore, Maryland
| | - Elias T Zambidis
- Blood and Marrow Transplantation Program, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins Hospital, Baltimore, Maryland
| | - Brandon Luber
- Blood and Marrow Transplantation Program, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins Hospital, Baltimore, Maryland
| | - Gary Rosner
- Blood and Marrow Transplantation Program, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins Hospital, Baltimore, Maryland
| | - Nicholas Siegel
- Blood and Marrow Transplantation Program, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins Hospital, Baltimore, Maryland
| | - Mary Jo Holuba
- Blood and Marrow Transplantation Program, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins Hospital, Baltimore, Maryland
| | - Nancy Robey
- Blood and Marrow Transplantation Program, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins Hospital, Baltimore, Maryland
| | - Masanori Hayashi
- Blood and Marrow Transplantation Program, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins Hospital, Baltimore, Maryland
| | - Richard J Jones
- Blood and Marrow Transplantation Program, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins Hospital, Baltimore, Maryland
| | - Ephraim Fuchs
- Blood and Marrow Transplantation Program, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins Hospital, Baltimore, Maryland
| | - Matthias Holdhoff
- Blood and Marrow Transplantation Program, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins Hospital, Baltimore, Maryland
| | - David M Loeb
- Blood and Marrow Transplantation Program, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins Hospital, Baltimore, Maryland
| | - Heather J Symons
- Blood and Marrow Transplantation Program, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins Hospital, Baltimore, Maryland
| |
Collapse
|
15
|
Impact of HLA-G polymorphism on the outcome of allogeneic hematopoietic stem cell transplantation for metastatic renal cell carcinoma. Bone Marrow Transplant 2017; 53:213-218. [DOI: 10.1038/bmt.2017.243] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2017] [Revised: 08/29/2017] [Accepted: 09/08/2017] [Indexed: 01/28/2023]
|
16
|
Gallo S, Sangiolo D, Carnevale Schianca F, Aglietta M, Montemurro F. Treating breast cancer with cell-based approaches: an overview. Expert Opin Biol Ther 2017; 17:1255-1264. [PMID: 28728493 DOI: 10.1080/14712598.2017.1356816] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
INTRODUCTION Breast cancer is the most common malignancy in women. Despite there being considerable progress in the treatment of this disease, metastatic dissemination is still considered an incurable condition at the present time, causing 500,000 deaths worldwide every year. Although most of the research efforts have been focused on pharmacological approaches, over the last three decades, the use of bone marrow and peripheral blood-derived cell therapy approaches have been attempted and developed. Areas covered: This review will briefly address cell therapy for breast cancer, including autologous stem cell transplantations for overcoming the myelosuppressive effects of high-dose chemotherapy, allogeneic stem cell transplants and adoptive immunotherapy using bone-marrow derived T-cells. Expert opinion: The treatment of breast cancer using bone marrow or peripheral-blood derived cells has evolved from a supportive care approach to allow dose escalation of conventional chemotherapy to a therapeutic strategy aimed at eliciting immune cell mediated anticancer immunity. This latter principle has led to the development of adoptive immunotherapies, either with 'natural' or genetically engineered effectors, which are being intensively investigated for their great potential against several solid tumors, including breast cancer.
Collapse
Affiliation(s)
- Susanna Gallo
- a Medical Oncology , Candiolo Cancer Institute-FPO (IRCCS) , Candiolo , Italy
| | - Dario Sangiolo
- a Medical Oncology , Candiolo Cancer Institute-FPO (IRCCS) , Candiolo , Italy.,b Department of Oncology , University of Turin , Turin , Italy
| | | | - Massimo Aglietta
- a Medical Oncology , Candiolo Cancer Institute-FPO (IRCCS) , Candiolo , Italy.,b Department of Oncology , University of Turin , Turin , Italy
| | - Filippo Montemurro
- c Investigative Clinical Oncology , Candiolo Cancer Institute-FPO (IRCCS) , Candiolo , Italy
| |
Collapse
|
17
|
Lee YK, Kang M, Choi EY. TLR/MyD88-mediated Innate Immunity in Intestinal Graft-versus-Host Disease. Immune Netw 2017; 17:144-151. [PMID: 28680375 PMCID: PMC5484644 DOI: 10.4110/in.2017.17.3.144] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2017] [Revised: 04/07/2017] [Accepted: 04/13/2017] [Indexed: 12/13/2022] Open
Abstract
Graft-versus-host disease (GHVD) is a severe complication after allogeneic hematopoietic stem cell transplantation. The degree of inflammation in the gastrointestinal tract, a major GVHD target organ, correlates with the disease severity. Intestinal inflammation is initiated by epithelial damage caused by pre-conditioning irradiation. In combination with damages caused by donor-derived T cells, such damage disrupts the epithelial barrier and exposes innate immune cells to pathogenic and commensal intestinal bacteria, which release ligands for Toll-like receptors (TLRs). Dysbiosis of intestinal microbiota and signaling through the TLR/myeloid differentiation primary response gene 88 (MyD88) pathways contribute to the development of intestinal GVHD. Understanding the changes in the microbial flora and the roles of TLR signaling in intestinal GVHD will facilitate the development of preventative and therapeutic strategies.
Collapse
Affiliation(s)
- Young-Kwan Lee
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Myungsoo Kang
- BioMembrane Plasticity Research Center (MPRC), Seoul National University College of Medicine, Seoul 03080, Korea
| | - Eun Young Choi
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Korea.,BioMembrane Plasticity Research Center (MPRC), Seoul National University College of Medicine, Seoul 03080, Korea
| |
Collapse
|
18
|
Atilla E, Ataca Atilla P, Demirer T. A Review of Myeloablative vs Reduced Intensity/Non-Myeloablative Regimens in Allogeneic Hematopoietic Stem Cell Transplantations. Balkan Med J 2017; 34:1-9. [PMID: 28251017 PMCID: PMC5322516 DOI: 10.4274/balkanmedj.2017.0055] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2017] [Accepted: 01/19/2017] [Indexed: 02/07/2023] Open
Abstract
Allogeneic hematopoietic stem cell transplantation (Allo-HSCT) is a curative treatment option for both malignant and some benign hematological diseases. During the last decade, many of the newer high-dose regimens in different intensity have been developed specifically for patients with hematologic malignancies and solid tumors. Today there are three main approaches used prior to allogeneic transplantation: Myeloablative (MA), Reduced Intensity Conditioning (RIC) and Non-MA (NMA) regimens. MA regimens cause irreversible cytopenia and there is a requirement for stem cell support. Patients who receive NMA regimen have minimal cytopenia and this type of regimen can be given without stem cell support. RIC regimens do not fit the criteria of MA and NMA: the cytopenia is reversible and the stem cell support is necessary. NMA/RIC for Allo-HSCT has opened a new era for treating elderly patients and those with comorbidities. The RIC conditioning was used for 40% of all Allo-HSCT and this trend continue to increase. In this paper, we will review these regimens in the setting of especially allogeneic HSCT and our aim is to describe the history, features and impact of these conditioning regimens on specific diseases.
Collapse
Affiliation(s)
- Erden Atilla
- Department of Hematology, Ankara University School of Medicine, Ankara, Turkey
| | - Pınar Ataca Atilla
- Department of Hematology, Ankara University School of Medicine, Ankara, Turkey
| | - Taner Demirer
- Department of Hematology, Ankara University School of Medicine, Ankara, Turkey
| |
Collapse
|
19
|
Karadurmus N, Sahin U, Basgoz BB, Arpaci F, Demirer T. Review of allogeneic hematopoietic stem cell transplantation with reduced intensity conditioning in solid tumors excluding breast cancer. World J Transplant 2016; 6:675-681. [PMID: 28058217 PMCID: PMC5175225 DOI: 10.5500/wjt.v6.i4.675] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2016] [Revised: 11/01/2016] [Accepted: 11/22/2016] [Indexed: 02/05/2023] Open
Abstract
Solid tumors in adults constitute a heterogeneous group of malignancy originating from various organ systems. Solid tumors are not completely curable by chemotherapy, even though some subgroups are very chemo-sensitive. Recently, oncologists have focused on the use of allogeneic hematopoietic stem cell transplantation (allo-HSCT) with reduced intensity conditioning (RIC) for the treatment of some refractory solid tumors. After the demonstration of allogeneic graft-versus-leukemia effect in patients with hematological malignancies who received allo-HSCT, investigators evaluated this effect in patients with refractory metastatic solid tumors. According to data from experimental animal models and preliminary clinical trials, a graft-versus-tumor (GvT) effect may also be observed in the treatment of some solid tumors (e.g., renal cell cancer, colorectal cancer, etc.) after allo-HSCT with RIC. The use of RIC regimens offers an opportunity of achieving full-donor engraftment with GvT effect, as well as, a reduced transplant-related mortality. Current literature suggests that allo-HSCT with RIC might become a choice for elderly and medically fragile patients with refractory metastatic solid tumors.
Collapse
|
20
|
An overview of hematopoietic stem cell transplantation related thrombotic complications. Crit Rev Oncol Hematol 2016; 107:149-155. [DOI: 10.1016/j.critrevonc.2016.09.004] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Revised: 06/19/2016] [Accepted: 09/21/2016] [Indexed: 02/07/2023] Open
|
21
|
Sahin U, Toprak SK, Atilla PA, Atilla E, Demirer T. An overview of infectious complications after allogeneic hematopoietic stem cell transplantation. J Infect Chemother 2016; 22:505-14. [PMID: 27344206 DOI: 10.1016/j.jiac.2016.05.006] [Citation(s) in RCA: 146] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2016] [Revised: 05/05/2016] [Accepted: 05/20/2016] [Indexed: 12/31/2022]
Abstract
Infections are the most common and significant cause of mortality and morbidity after allogeneic hematopoietic stem cell transplantation (allo-HSCT). The presence of neutropenia and mucosal damage are the leading risk factors in the early pre-engraftment phase. In the early post-engraftment phase, graft versus host disease (GvHD) induced infection risk is increased in addition to catheter related infections. In the late phase, in which reconstitution of cellular and humoral immunity continues, as well as the pathogens seen during the early post-engraftment phase, varicella-zoster virus and encapsulated bacterial infections due to impaired opsonization are observed. An appropriate vaccination schedule following the cessation of immunosuppressive treatment after transplantation, intravenous immunoglobulin administration, and antimicrobial prophylaxis with penicillin or macrolide antibiotics during immunosuppressive treatment for GvHD might decrease the risk of bacterial infections. Older age, severe mucositis due to toxicity of chemotherapy, gastrointestinal tract colonization, prolonged neutropenia, unrelated donor and cord blood originated transplantations, acute and chronic GvHD are among the most indicative clinical risk factors for invasive fungal infections. Mold-active anti-fungal prophylaxis is suggested regardless of the period of transplantation among high risk patients. The novel serological methods, including Aspergillus galactomannan antigen and beta-D-glucan detection and computed tomography are useful in surveillance. Infections due to adenovirus, influenza and respiratory syncytial virus are encountered in all phases after allo-HSCT, including pre-engraftment, early post-engraftment and late phases. Infections due to herpes simplex virus-1 and -2 are mostly seen during the pre-engraftment phase, whereas, infections due to cytomegalovirus and human herpes virus-6 are seen in the early post-engraftment phase and Epstein-Barr virus and varicella-zoster virus infections often after +100th day. In order to prevent mortality and morbidity of infections after allo-HSCT, the recipients should be carefully followed-up with appropriate prophylactic measures in the post-transplant period.
Collapse
Affiliation(s)
- Ugur Sahin
- Ankara University Medical School, Department of Hematology, Ankara, Turkey
| | | | - Pinar Ataca Atilla
- Ankara University Medical School, Department of Hematology, Ankara, Turkey
| | - Erden Atilla
- Ankara University Medical School, Department of Hematology, Ankara, Turkey
| | - Taner Demirer
- Ankara University Medical School, Department of Hematology, Ankara, Turkey.
| |
Collapse
|
22
|
Abstract
Our understanding of cancer biology has been radically transformed over recent years with a more realistic grasp of its multilayered cellular and genetic complexity. These advances are being translated into more selective and effective treatment of cancers and, although there are still considerable challenges, particularly with drug resistance and metastatic disease, many patients with otherwise lethal malignancies now enjoy protracted remissions or cure. One largely unheralded theme of this story is the extent to which new biological insights and novel clinical applications have their origins with leukaemia and related blood cell cancers, including lymphoma. In this Timeline article, I review the remarkable and ground-breaking role that studies in leukaemia have had at the forefront of this progress.
Collapse
Affiliation(s)
- Mel Greaves
- Centre for Evolution and Cancer, The Institute of Cancer Research, Brookes Lawley Building, 15 Cotswold Road, Sutton SM2 5NG, UK
| |
Collapse
|
23
|
Abstract
Tumors originate from a number of genetic events that deregulate homeostatic mechanisms controlling normal cell behavior. The immune system, devoted to patrol the organism against pathogenic events, can identify transformed cells, and in several cases cause their elimination. It is however clear that several mechanisms encompassing both central and peripheral tolerance limit antitumor immunity, often resulting into progressive diseases. Adoptive T-cell therapy with either allogeneic or autologous T cells can transfer therapeutic immunity. To date, genetic engineering of T cells appears to be a powerful tool for shaping tumor immunity. In this review, we discuss the most recent achievements in the areas of suicide gene therapy, and TCR-modified T cells and chimeric antigen receptor gene-modified T cells. We provide an overview of current strategies aimed at improving the safety and efficacy of these approaches, with an outlook on prospective developments.
Collapse
Affiliation(s)
- Chiara Bonini
- Experimental Hematology Unit, Division of Immunology, Transplantation and Infectious Diseases, San Raffaele Scientific Institute, Milan, Italy
| | - Anna Mondino
- Lymphocyte Activation Unit, Division of Immunology, Transplantation and Infectious Diseases, San Raffaele Scientific Institute, Milan, Italy
| |
Collapse
|
24
|
Sureda A, Bader P, Cesaro S, Dreger P, Duarte RF, Dufour C, Falkenburg JHF, Farge-Bancel D, Gennery A, Kröger N, Lanza F, Marsh JC, Nagler A, Peters C, Velardi A, Mohty M, Madrigal A. Indications for allo- and auto-SCT for haematological diseases, solid tumours and immune disorders: current practice in Europe, 2015. Bone Marrow Transplant 2015; 50:1037-56. [PMID: 25798672 DOI: 10.1038/bmt.2015.6] [Citation(s) in RCA: 216] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2014] [Accepted: 01/09/2015] [Indexed: 12/17/2022]
Abstract
This is the sixth special report that the European Society for Blood and Marrow Transplantation regularly publishes on the current practice and indications for haematopoietic SCT for haematological diseases, solid tumours and immune disorders in Europe. Major changes have occurred in the field of haematopoietic SCT over the last years. Cord blood units as well as haploidentical donors have been increasingly used as stem cell sources for allo-SCT, thus, augmenting the possibility of finding a suitable donor for a patient. Continuous refinement of conditioning strategies has also expanded not only the number of potential indications but also has permitted consideration of older patients or those with co-morbidity for a transplant. There is accumulating evidence of the role of haematopoietic SCT in non-haematological disorders such as autoimmune diseases. On the other hand, the advent of new drugs and very effective targeted therapy has challenged the role of SCT in some instances or at least, modified its position in the treatment armamentarium of a given patient. An updated report with revised tables and operating definitions is presented.
Collapse
Affiliation(s)
- A Sureda
- Department of Haematology, Institut Catala d'Oncologia, Hospital Duran I Reynals, Barcelona, Spain
| | - P Bader
- Universitätsklinikum Frankfurt, Goethe-Universität, Klinik für Kinder- und Jugendmedizin, Frankfurt, Germany
| | - S Cesaro
- Paediatric Haematology Oncology, Policlinico G.B. Rossi, Verona, Italy
| | - P Dreger
- Department of Internal Medicine V, University of Heidelberg, Heidelberg, Germany
| | - R F Duarte
- Department of Haematology, Institut Catala d'Oncologia, Hospital Duran I Reynals, Barcelona, Spain
| | - C Dufour
- Clinical And Experimental Hematology Unit. Institute G. Gaslini, Genoa, Italy
| | - J H F Falkenburg
- Department of Haematology, Leiden University Medical Center, Leiden, The Netherlands
| | - D Farge-Bancel
- Department of Haematology-BMT, Hopital St Louis, Paris, France
| | - A Gennery
- Children's BMT Unit, Great North Children's Hospital, Newcastle-Upon-Tyne, UK
| | - N Kröger
- Department of Stem Cell Transplantation, University hospital Eppendorf, Hamburg, Germany
| | - F Lanza
- Haematology and BMT Unit, Cremona, Italy
| | - J C Marsh
- Department of Haematological Medicine, King's College Hospital/King's College London, London, UK
| | - A Nagler
- Chaim Sheva Medical Center, Tel-Hashomer, Israel
| | - C Peters
- Stem Cell Transplantation Unit, St Anna Kinderspital, Vienna, Austria
| | - A Velardi
- Sezione di Ematologia, Dipartimento di Medicina Clinica e Sperimentale, Università di Perugia, Perugia, Italy
| | - M Mohty
- Department of Haematology, H. Saint Antoine, Paris, France
| | - A Madrigal
- Anthony Nolan Research Institute, Royal Free and University College, London, UK
| |
Collapse
|
25
|
Mesiano G, Leuci V, Giraudo L, Gammaitoni L, Carnevale Schianca F, Cangemi M, Rotolo R, Capellero S, Pignochino Y, Grignani G, Aglietta M, Sangiolo D. Adoptive immunotherapy against sarcomas. Expert Opin Biol Ther 2014; 15:517-28. [PMID: 25516119 DOI: 10.1517/14712598.2015.987121] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
INTRODUCTION Conventional treatments reached an unsatisfactory therapeutic plateau in the treatment of advanced unresectable bone and soft tissue sarcomas that remain an unsolved medical need. Several evidences support the concept that adoptive immunotherapy may effectively integrate within the complex and multidisciplinary treatment of sarcomas. AREAS COVERED In this work we reviewed adoptive immunotherapy strategies that have been explored in sarcoma settings, with specific focus on issues related to their clinic transferability. We schematically divided approaches based on T lymphocytes specific for MHC-restricted tumor-associated antigens or relying on MHC-independent immune effectors such as natural killer (NK), cytokine-induced killer (CIK) or γδ T cells. EXPERT OPINION Preclinical findings and initial clinical reports showed the potentialities and drawbacks of different adoptive immunotherapy strategies. The expansion of tumor infiltrating lymphocytes is difficult to be reproduced outside melanoma. Genetically redirected T cells appear to be a promising option and initial reports are encouraging against patients with sarcomas. Adoptive immunotherapy with MHC-unrestricted effectors such as NK, CIK or γδ T cells has recently shown great preclinical potential in sarcoma setting and biologic features that may favor clinical transferability. Combination of different immunotherapy approaches and integration with conventional treatments appear to be key issues for successful designing of next clinical trials.
Collapse
Affiliation(s)
- Giulia Mesiano
- Candiolo Cancer Institute-IRCCS, Laboratory of Medical Oncology, Experimental Cell Therapy , Candiolo, Turin , Italy
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Willems L, Waer M, Billiau AD. The graft-versus-neuroblastoma effect of allogeneic hematopoietic stem cell transplantation, a review of clinical and experimental evidence and a perspective on mechanisms. Pediatr Blood Cancer 2014; 61:2151-7. [PMID: 25156335 DOI: 10.1002/pbc.25169] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2014] [Accepted: 06/11/2014] [Indexed: 01/17/2023]
Abstract
Despite aggressive treatment, patients with high-risk neuroblastoma face high relapse rates and bleak prognoses. Increasing evidence that neuroblastoma cells are or can become immunogenic has stimulated research into novel therapies based on triggering or enhancing tumor immunity. Here we review clinical and experimental studies on this subject, the underlying immune mechanisms and perspectives for clinical application. Allogeneic hematopoietic stem cell transplantation has proven to be of substantial benefit in the treatment of certain leukemias through the generation of a graft-versus-leukemia-effect and has become of interest as a possible treatment for patients with solid tumors, including neuroblastoma.
Collapse
Affiliation(s)
- Leen Willems
- Laboratory of Experimental Transplantation, KULeuven, Leuven, Belgium
| | | | | |
Collapse
|
27
|
Schmitz-Winnenthal FH, Schmidt T, Lehmann M, Beckhove P, Kieser M, Ho AD, Dreger P, Büchler MW. Stem cell Transplantation for Eradication of Minimal PAncreatic Cancer persisting after surgical Excision (STEM PACE Trial, ISRCTN47877138): study protocol for a phase II study. BMC Cancer 2014; 14:168. [PMID: 24612467 PMCID: PMC4008264 DOI: 10.1186/1471-2407-14-168] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2013] [Accepted: 03/03/2014] [Indexed: 01/09/2023] Open
Abstract
Background Pancreatic cancer is the third most common cancer related cause of death. Even in the 15% of patients who are eligible for surgical resection the outlook is dismal with less than 10% of patients surviving after 5 years. Allogeneic hematopoietic (allo-HSCT) stem cell transplantation is an established treatment capable of to providing cure in a variety of hematopoietic malignancies. Best results are achieved when the underlying neoplasm has been turned into a stage of minimal disease by chemotherapy. Allo-HSCT in advanced solid tumors including pancreatic cancer have been of limited success, however studies of allo-HSCT in solid tumors in minimal disease situations have never been performed. The aim of this trial is to provide evidence for the clinical value of allo-HSCT in pancreatic cancer put into a minimal disease status by effective surgical resection and standard adjuvant chemotherapy. Methods/Design The STEM PACE trial is a single center, phase II study to evaluate adjuvant allogeneic hematopoietic stem cell transplantation in pancreatic cancer after surgical resection. The study will evaluate as primary endpoint 2 year progression free survival and will generate first time state-of-the-art scientific clinical evidence if allo-HSCT is feasible and if it can provide long term disease control in patients with effectively resected pancreatic cancer. Screened eligible patients after surgical resection and standard adjuvant chemotherapy with HLA matched related stem cell donor can participate. Patients without a matched donor will be used as a historical control. Study patients will undergo standard conditioning for allo-HSCT followed by transplantation of allogeneic unmanipulated peripheral blood stem cells. The follow up of the patients will continue for 2 years. Secondary endpoints will be evaluated on 7 postintervention visits. Discussion The principal question addressed in this trial is whether allo-HSCT can change the unfavourable natural course of this disease. The underlying hypothesis is that allo-HSCT has the capacity to provide long-term disease control to an extent otherwise not possible in pancreatic cancer, thereby substantially improving survival of affected patients. Trial registration This trial has been registered: ISRCTN47877138
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Markus W Büchler
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Im Neuenheimer Feld 110, 69120 Heidelberg, Germany.
| |
Collapse
|
28
|
Rashidi A. Tumors with a more complex genome have a higher frequency of HLA class I total loss: a unifying pan-cancer hypothesis. ACTA ACUST UNITED AC 2014; 83:286-9. [PMID: 24571087 DOI: 10.1111/tan.12315] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2014] [Revised: 01/29/2014] [Accepted: 01/30/2014] [Indexed: 12/13/2022]
Affiliation(s)
- A Rashidi
- Division of Oncology, Washington University School of Medicine, St. Louis, MO, USA
| |
Collapse
|
29
|
Suzuki K, Aida K, Miyakawa R, Narumi K, Udagawa T, Yoshida T, Ohshima Y, Aoki K. Preimmunization of donor lymphocytes enhances antitumor immunity of autologous hematopoietic stem cell transplantation. Cancer Med 2014; 2:636-45. [PMID: 24403229 PMCID: PMC3892795 DOI: 10.1002/cam4.117] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2013] [Revised: 08/07/2013] [Accepted: 08/08/2013] [Indexed: 01/10/2023] Open
Abstract
Lymphopenia-induced homeostatic proliferation (HP) of T cells following autologous hematopoietic stem cell transplantation (HSCT) skews the T-cell repertoire by engaging tumor-associated antigens (TAAs), leading to an induction of antitumor immunity. Here, as the tumor-reactive lymphocytes preferentially proliferate during the condition of HP, we examined whether the priming of a donor lymphocytes to TAAs could enhance HP-induced antitumor immunity in autologous HSCT recipients. First, to examine whether the tumor-bearing condition of donor influences the antitumor effect of HSCT, the lymphocytes isolated from CT26 tumor-bearing mice were infused into lethally irradiated mice. The growth of tumors was substantially suppressed in the mice that received HSCT from a tumor-bearing donor compared with a naïve donor, suggesting that a fraction of donor lymphocytes from tumor-bearing mice are primed in response to TAAs and remain responsive upon transplantation. We previously reported that type I interferon (IFN) maturates the dendritic cells and promotes the priming of T cells. We then investigated whether the further priming of donor cells by IFN-α can strengthen the antitumor effect of HSCT. The intratumoral IFN-α gene transfer significantly increased the number of IFN-γ-positive lymphocytes in response to CT26 cells but not the syngeneic lymphocytes in donor mice. The infusion of primed donor lymphocytes markedly suppressed the tumor growth in recipient mice, and cured 64% of the treated mice. Autologous HSCT with the infusion of primed donor lymphocytes is a promising strategy to induce an effective antitumor immunity for solid cancers.
Collapse
Affiliation(s)
- Koji Suzuki
- Division of Gene and Immune Medicine, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan; Department of Pediatrics, Fukui University School of Medicine, 23-3 Shimoaizuki, Matsuoka, Yoshida-gun, Fukui, 910-1193, Japan
| | | | | | | | | | | | | | | |
Collapse
|
30
|
Willems L, Fevery S, Sprangers B, Rutgeerts O, Lenaerts C, Ibrahimi A, Gijsbers R, Van Gool S, Waer M, Billiau AD. Recipient leukocyte infusion enhances the local and systemic graft-versus-neuroblastoma effect of allogeneic bone marrow transplantation in mice. Cancer Immunol Immunother 2013; 62:1733-44. [PMID: 24081484 PMCID: PMC11028935 DOI: 10.1007/s00262-013-1479-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2013] [Accepted: 09/23/2013] [Indexed: 01/05/2023]
Abstract
Allogeneic hematopoietic stem cell transplantation and donor leukocyte infusion (DLI) may hold potential as a novel form of immunotherapy for high-risk neuroblastoma. DLI, however, carries the risk of graft-versus-host disease (GvHD). Recipient leukocyte infusion (RLI) induces graft-versus-leukemia responses without GvHD in mice and is currently being explored clinically. Here, we demonstrate that both DLI and RLI, when given to mixed C57BL/6→A/J radiation chimeras carrying subcutaneous Neuro2A neuroblastoma implants, can slow the local growth of such tumors. DLI provoked full donor chimerism and GvHD; RLI produced graft rejection but left mice healthy. Flow cytometric studies showed that the chimerism of intratumoral leukocytes paralleled the systemic chimerism. This was associated with increased CD8/CD4 ratios, CD8+ T-cell IFN-γ expression and NK-cell Granzyme B expression within the tumor, following both DLI and RLI. The clinically safe anti-tumor effect of RLI was further enhanced by adoptively transferred naïve recipient-type NK cells. In models of intravenous Neuro2A tumor challenge, allogeneic chimeras showed superior overall survival over syngeneic chimeras. Bioluminescence imaging in allogeneic chimeras challenged with luciferase-transduced Neuro2A cells showed both DLI and RLI to prolong metastasis-free survival. This is the first experimental evidence that RLI can safely produce a local and systemic anti-tumor effect against a solid tumor. Our data indicate that RLI may provide combined T-cell and NK-cell reactivity effectively targeting Neuro2A neuroblastoma.
Collapse
Affiliation(s)
- Leen Willems
- Laboratory of Experimental Transplantation, KU Leuven, Herestraat 49, box 811, 3000, Leuven, Belgium,
| | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Udagawa T, Narumi K, Suzuki K, Aida K, Miyakawa R, Ikarashi Y, Makimoto A, Chikaraishi T, Yoshida T, Aoki K. Vascular endothelial growth factor-D-mediated blockade of regulatory T cells within tumors is induced by hematopoietic stem cell transplantation. THE JOURNAL OF IMMUNOLOGY 2013; 191:3440-52. [PMID: 23966628 DOI: 10.4049/jimmunol.1201454] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Lymphopenia-induced homeostatic proliferation of T cells after autologous hematopoietic stem cell transplantation (HSCT) skews the T cell repertoire by engaging tumor-associated Ags, leading to an induction of antitumor immunity. However, how HSCT alters the immunosuppressive microenvironment in the tumors is unknown. In this study, we first analyzed the kinetics of regulatory T cells (Tregs) in the tumors after syngeneic HSCT. Unexpectedly, the frequency of CD4⁺ cells expressing Foxp3 was increased in the spleens, whereas the frequency was clearly decreased in the tumors after HSCT. The origin of reconstituted CD4⁺ and Foxp3⁺ cells in the tumors was mainly from the expansion of transferred splenic T cells. Then, to examine the mechanism of Treg suppression after HSCT, we isolated CD11c⁺ cells from tumors. A large amount of Treg-inhibitory cytokine IL-6 was secreted from the CD11c⁺ cells in the tumors, but not in the spleens in the recipient mice. Furthermore, to understand what factor affects the activity of CD11c⁺ cells in the tumors after HSCT, we analyzed the expression of various cytokines/chemokines with mouse cytokine Ab arrays, and noticed that VEGF-D concentration was increased in the tumors in the early period after HSCT. The CD11c⁺ cells produced IL-6 in response to VEGF-D stimulation, and an administration of VEGF receptor-3 neutralizing Ab significantly suppressed the production of IL-6 from CD11c⁺ cells accompanied with the increase of Tregs in the tumors of HSCT recipients. Autologous HSCT creates an environment that strongly supports the enhancement of antitumor immunity in reconstituted lymphopenic recipients through the suppression of Tregs.
Collapse
Affiliation(s)
- Takeshi Udagawa
- Division of Gene and Immune Medicine, National Cancer Center Research Institute, Chuo-ku, Tokyo 104-0045, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Reduced-intensity conditioning followed by allogeneic transplantation in pediatric malignancies: a report from the Société Française des Cancers de l'Enfant and the Société Française de Greffe de Moelle et de Thérapie Cellulaire. Bone Marrow Transplant 2013; 48:1401-8. [PMID: 23749103 DOI: 10.1038/bmt.2013.82] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2012] [Revised: 03/18/2013] [Accepted: 04/23/2013] [Indexed: 01/15/2023]
Abstract
We report French prospective experience with reduced-intensity conditioning-allo-SCT in 46 patients (median age: 15.5 years, 4.8-20.2) presenting high-risk AL (n=11), Hodgkin's lymphoma (n=15) or solid tumors (n=20). Graft sources were BM (n=21), PBSC (n=20) and cord blood (CB; n=5) from related (n=20) or unrelated (n=26) donors. For CB grafts, only one patient out of five achieved sustained engraftment. For PBSC/BM grafts, engraftment rate was 95%, hematopoietic recovery times were not significantly different between BM, PBSC, sibling or unrelated grafts, day+100. Full donor chimerism was achieved in 94% of patients, and incidences of primary acute GVHD and chronic GVHD were 49% and 14%, respectively. Underlying disease was fatal in 39% of patients. TRM was 6.9%. Three-year OS was 49.15%. OS and EFS were not significantly different between patients transplanted with different grafts and with or without primary GVHD. Patients with solid tumor or measurable disease at transplant had poorer outcomes. Three-year EFS: 33.3% for ALL, 75.0% for AML, 51.8% for Hodgkin's lymphoma, 28.6% for neuroblastoma and 22.2% for sarcoma patients. This multicentre study concluded that Bu/fludarabine/anti-thymocyte globulin conditioning with PB or BM, related or unrelated grafts in patients with various malignancies at high-risk for transplantation toxicity results in high engraftment rates, low TRM and acceptable survival.
Collapse
|
33
|
Lin S, Fu S. The correlation of hematopoietic stem cells with cancer stem cells through the regulation of stromal cells in tumor microenvironment. Med Hypotheses 2013; 80:494-7. [PMID: 23317540 DOI: 10.1016/j.mehy.2012.12.016] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2012] [Revised: 12/09/2012] [Accepted: 12/13/2012] [Indexed: 01/15/2023]
Abstract
Cancer stem cells are a small population of tumor cells that have many malignant features such as chemotherapy resistance, radiotherapy resistance, tumorigenicity and are responsible for tumor progression, disease recurrence and metastasis. Therefore, insight into the regulation of the biology of cancer stem cells is important to eradicate cancer. Recently, studies suggested that hematopoietic stem cells could incorporate into tumor stroma and differentiated into stromal cells and the cells derived from hematopoietic stem cells play an important role on tumor progress. Moreover, cancer cells competed with hematopoietic stem cells for occupancy of the hematopoietic stem cell niches to regulate bone metastasis and most cancer cells in bone marrow metastasis were cancer stem cells. Therefore, we hypothesize that cancer stem cells could promote hematopoietic stem cells incorporating into tumor microenvironment and resulting into transformation of hematopoietic stem cells to stromal cells, which could impact the biological behavior of cancer stem cells.
Collapse
Affiliation(s)
- Shuchen Lin
- Department of Radiation Oncology, Sixth People's Hospital of Jiao Tong University, Shanghai, People's Republic of China
| | | |
Collapse
|
34
|
Successful allogeneic hematopoietic cell engraftment after a minimal conditioning regimen in children with relapsed or refractory solid tumors. Biol Blood Marrow Transplant 2012; 19:291-7. [PMID: 23063628 DOI: 10.1016/j.bbmt.2012.10.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2012] [Accepted: 10/03/2012] [Indexed: 01/10/2023]
Abstract
Children with relapsed or refractory solid tumors face dismal prognoses, and novel therapies are desperately needed. Allogeneic hematopoietic cell transplantation (HCT) offers potential for cell-based therapy, but the toxicity of myeloablation limits this approach in heavily pretreated patients. We sought to determine the feasibility of HCT in a cohort of 24 children with incurable solid tumors using human leukocyte antigen-matched sibling or unrelated donors and a minimal conditioning regimen. Before stem cell infusion, all patients received 3 daily doses of 30 mg/m(2) fludarabine followed by 2 Gy of total body irradiation. Hematopoietic cell recovery was rapid and reliable. Median time to neutrophil engraftment was 13.5 days for sibling donors and 12 days for unrelated donors. Donor lymphocyte infusions were used safely in 4 patients, all of whom had either improved chimerism or apparent tumor response. Graft-versus-host disease was comparable across donor sources and did not affect survival. Relapse remains a substantial barrier, although objective graft-versus-tumor effect was observed in several patients. Four patients with detectable disease before HCT achieved a complete response for at least 30 days after HCT, and two remain long-term survivors. Three patients were in complete response before HCT and remained in remission for 3, 6, and 74 months after HCT. Early disease response was associated with improved survival. Allogeneic HCT using this conditioning regimen offers a potential platform for novel immunotherapies.
Collapse
|
35
|
Morris GS, des Bordes JK, Holmes HM, Giralt S. Frailty and stem cell transplantation in the older patient with cancer. J Geriatr Oncol 2012. [DOI: 10.1016/j.jgo.2012.03.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
36
|
Song Y, Sun W, Weng X, Liang Z, Yu Q, Wang Z, Ouyang L, Chen J, Wu X, Shen G, Wu X. Tumor rejection effects of allorestricted tumor peptide-specific CD4(+) T cells on human cervical cancer cell xenograft in nude mice. Cell Transplant 2012; 21:1503-14. [PMID: 22525838 DOI: 10.3727/096368912x640510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Generation of tumor specific alloreactive CD4(+) T cells is important to circumvent tumor tolerance. Here, we generate allorestricted peptide-specific CD4(+) T cells by coculture of lymphocytes and autologous monocytes bearing allogeneic HLA-DR15 molecule associated with its restricted peptide. Binding of a dimeric HLA-DR15/IgG1-Fc fusion protein (the dimer) to HLA-DR15 negative (HLA-DR15-ve) monocytes made the monocytes coated with the allogeneic epitope. An increased proliferation of CD4(+) T cells and induction of Th1 cells appeared after coculturing of HLA-DR15-ve lymphocytes and the autologous monocytes loaded with the dimer. The cocultural bulks showed an increased frequency of the specific dimer-stained CD4(+) T cells and the expanded CD4(+) T cells exhibited an elevated IFN-γ production in response to specific TCR ligand. Tumor rejection effects of the allorestricted E7-specific CD4(+) T cells raised by the coculture were observed in nude mice challenged with human cervical cancer cell SiHa expressing both HLA-DR15 and E7 antigens, as the tumor avoidance and life span of the mice were improved after adoptive transfer of the CD4(+) T cells. This study may help to develop strategies to separate graft-versus-leukemia or graft-versus-tumor reaction from graft-versus-host disease, and add to the pool of human high-avidity TCRs specific for tumor or virus antigens.
Collapse
Affiliation(s)
- Yinhong Song
- Department of Immunology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Reduced-intensity allogeneic stem cell transplantation in children and young adults with ultrahigh-risk pediatric sarcomas. Biol Blood Marrow Transplant 2011; 18:698-707. [PMID: 21896345 DOI: 10.1016/j.bbmt.2011.08.020] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2011] [Accepted: 08/25/2011] [Indexed: 11/23/2022]
Abstract
Some subsets of pediatric sarcoma patients have very poor survival rates. We sought to determine the feasibility and efficacy of allogeneic hematopoietic stem cell transplantation (alloHSCT) in pediatric sarcoma populations with <25% predicted overall survival (OS). Patients with ultrahigh-risk Ewing's sarcoma family of tumors (ESFT), alveolar rhabdomyosarcoma, or desmoplastic small round cell tumors received EPOCH-fludarabine induction, a cyclophosphamide/fludarabine/melphalan preparative regimen, and HLA matched related peripheral blood stem cells. Thirty patients enrolled; 7 did not undergo alloHSCT because of progressive disease with diminishing performance status during induction. All 23 alloHSCT recipients experienced rapid full-donor engraftment, with no peritransplantation mortality. Five of 23 alloHSCT recipients (22%) remain alive (OS of 30% by Kaplan-Meier analysis at 3 years), including 3 of 7 (42%) transplanted without overt disease (median survival 14.5 versus 29.0 months from alloHSCT for patients transplanted with versus without overt disease, respectively). Among the 28 patients who progressed on the study, the median survival from date of progression was 1.9 months for the 7 who did not receive a transplant compared with 11.4 months for the 21 transplanted (P = .0003). We found prolonged survival after posttransplantation progression with several patients exhibiting indolent tumor growth. We also saw several patients with enhanced antitumor effects from posttransplantation chemotherapy (objective response to pretransplantation EPOCH-F was 24% versus 67% to posttransplantation EOCH); however, this was associated with increased toxicity. This largest reported series of alloHSCT in sarcomas demonstrates that alloHSCT is safe in this population, and that patients undergoing alloHSCT without overt disease show higher survival rates than reported using standard therapies. Enhanced chemo- and radiosensitivity of tumors and normal tissues was observed posttransplantation.
Collapse
|
38
|
Zinke-Cerwenka W, Valentin A, Posch U, Beham-Schmid C, Groselj-Strele A, Linkesch W, Wölfler A, Sill H. Reduced-intensity allografting in patients with therapy-related myeloid neoplasms and active primary malignancies. Bone Marrow Transplant 2011; 46:1540-4. [PMID: 21860429 DOI: 10.1038/bmt.2011.165] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Therapy-related myeloid neoplasms (t-MNs) are severe long-term consequences of cytotoxic treatments for a primary, often, malignant disorder. So far, the majority of patients eligible for transplantation have undergone myeloablative allo haematopoietic SCT (HSCT) as a potentially curative treatment, but it has been associated with high transplantation-related mortality (TRM) rates. In this retrospective study, we analysed the outcome of patients with t-MNs undergoing HSCT with reduced-intensity conditioning (RIC). Of 55 patients, seen at a single centre over a 10-year period, 17 underwent RIC HSCT with related or unrelated donors. The estimated overall survival was 53% at 1 year and 47% at 3 years, and disease-free survival was 47% at 1 year. At 1 year, the cumulative incidence of relapse and TRM were 24% and 30%, respectively. Of five patients with active primary neoplasms who underwent transplantation, two are alive beyond 1 year and show CR of both t-MNs and the primary malignancy. These data indicate that RIC HSCT is an encouraging approach for patients with t-MNs. The issue of primary malignancies not being in remission at the time of transplantation should be explored in further studies.
Collapse
Affiliation(s)
- W Zinke-Cerwenka
- Division of Haematology, Medical University of Graz, Graz, Austria
| | | | | | | | | | | | | | | |
Collapse
|
39
|
Porta C, Pedrazzoli P, Paglino C. Is immunotherapy re-entering the kidney cancer arena from the back door? Considerations from the Phase I/II study of siltuximab. Immunotherapy 2011; 3:487-90. [PMID: 21463189 DOI: 10.2217/imt.11.19] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Evaluation of: Rossi JF, Negrier S, James ND et al.: A Phase I/II study of siltuximab (CNTO 328), an anti-interleukin-6 monoclonal antibody, in metastatic renal cell cancer. Br. J. Cancer 103, 1154-1162 (2010). For years, immunotherapy was the mainstay of treatment for advanced kidney cancer. The recent development of molecularly targeted agents, and of antiangiogenic agents in particular, has completely changed this scenario, leaving immunotherapy with a minor role in treatment, if any. Recent insights in the mechanisms of resistance to these drugs suggest that the immune system could play a relevant role. In particular, IL-6 appears to cooperate with other growth factors in the recruitment of bone marrow-derived immune cells, ultimately leading to resistance to antiangiogenic agents. Furthermore, it is now clear that monoclonal antibodies actively interact with the immune system and show significant immune-modulating effects per se. Further translation research on this issue is much needed.
Collapse
Affiliation(s)
- Camillo Porta
- Medical Oncology, IRCCS San Matteo University Hospital Foundation, Pavia, Italy.
| | | | | |
Collapse
|
40
|
Abstract
Among the novel biological therapeutics that will increase our ability to cure human cancer in years to come, adoptive cellular therapy is one of the most promising approaches. Although this is a complex and challenging field, there have been major advances in basic and translational research resulting in clinical trial activity that is now beginning to confirm this promise. The results obtained with tumor-infiltrating lymphocytes therapy for melanoma, and virus-specific CTLs for EBV-associated malignancies are encouraging in terms of both ability to obtain clinical benefit and limited toxicity profile. In both settings, objective responses were obtained in at least 50% of treated patients. However, improvements to the clinical protocols, in terms of better patient selection and timing of administration, as well as cell product quality and availability, are clearly necessary to further ameliorate outcome, and logistical solutions are warranted to extend T-cell therapy beyond academic centers. In particular, there is a need to simplify cell production, in order to decrease costs and ease preparation. Promising implementations are underway, including harnessing the therapeutic potential of T cells transduced with TCRs directed against shared tumor antigens, and delineating strategies aimed at targeting immune evasion mechanisms exerted by tumor cells.
Collapse
|
41
|
Pedrazzoli P, Secondino S, Perfetti V, Comoli P, Montagna D. Immunotherapeutic Intervention against Sarcomas. J Cancer 2011; 2:350-6. [PMID: 21716856 PMCID: PMC3119402 DOI: 10.7150/jca.2.350] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2011] [Accepted: 06/03/2011] [Indexed: 12/16/2022] Open
Abstract
Advances in systemic therapy for sarcoma have produced, over the last two decades, relatively short-term benefits for the majority of patient. Among the novel biologic therapeutics that will likely increase our ability to cure human cancer in the years to come, immunotherapy is one of the most promising approaches. While past attempts to use immunotherapy have failed to dramatically shift the paradigm of care for the treatment of patients with sarcoma, major advances in basic and translational research have resulted, in more recent years, in clinical trial activity that is now beginning to generate promising results. However, to move from “proof of principle” to large scale clinical applicability, we need well-designed, multi-institutional clinical trials, along with continuous laboratory research to explore further the immunological characteristics of individual sarcoma subtypes and the consequent tailoring of therapy.
Collapse
Affiliation(s)
- Paolo Pedrazzoli
- 1. SC Oncologia, Fondazione IRCCS Policlinico S. Matteo, Pavia, Italy
| | | | | | | | | |
Collapse
|
42
|
Abstract
Better understanding of the antitumor effect of allogeneic transplant and the need to reduce the toxicity of the procedure, particularly in elderly patients have spurred the development of reduced-intensity conditioning regimens (RIC). These regimens allow fast engraftment with very low chemotherapy-induced toxicity. They are widely used in adults and there are numerous studies to demonstrate their feasibility and efficiency, but in pediatrics, the place of RIC remains to be determined. They can be proposed in two pediatric populations. First, solid tumors or hematological malignancies remaining unresponsive to the reference strategies according to best practices in pediatrics. Second, in children presenting malignancies for which allografting is the only recognized curative indication but is contraindicated with myeloablative conditioning regimens. More than 100 pediatrics cases have been reported in various pathologies, including blood diseases, acute leukemia, Hodgkin's lymphoma and solid tumors, and promising results published recently underline how RIC warrants further investigation in prospective comparative multicentric trials. The use of new post-graft treatment modalities is expected to pave the way to the development of RIC in pediatric patients.
Collapse
|
43
|
Anti-leukemia effect of ex vivo expanded DNT cells from AML patients: a potential novel autologous T-cell adoptive immunotherapy. Leukemia 2011; 25:1415-22. [PMID: 21566657 DOI: 10.1038/leu.2011.99] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
CD3(+)CD56(-), CD4 and CD8 double negative T (DNT) cells comprise 1-3% of peripheral blood (PB) mononuclear cells. Their role in tumor immunity remains largely unknown due to their limited numbers and lack of effective methods to expand them. Here we developed a novel protocol by which DNT cells can be expanded ex vivo to therapeutic levels in 2 weeks from 13 of 16 acute myeloid leukemia (AML) patients during chemotherapy-induced complete remission. The expanded DNT cells expressed similar or higher levels of interferon-γ and tumor necrosis factor-α and Granzyme B as that seen in bulk activated CD8T cells from the same patient but significantly higher levels of perforin. The expanded DNT cells could effectively kill both allogeneic and autologous primary CD34(+) leukemic blasts isolated from PB of AML patients in a perforin-dependant manner. These results demonstrate, for the first time, that DNT cells from AML patients can be expanded ex vivo even after intensive chemotherapy, and are effective at killing both allogeneic and autologous primary leukemic blasts. These findings warrant studies further exploring the potential of DNT cells as a novel adjuvant immunotherapy to decrease the risk of relapse in patients with AML and, perhaps, other cancers.
Collapse
|
44
|
Peinemann F, Smith LA, Kromp M, Bartel C, Kröger N, Kulig M. Autologous hematopoietic stem cell transplantation following high-dose chemotherapy for non-rhabdomyosarcoma soft tissue sarcomas. Cochrane Database Syst Rev 2011:CD008216. [PMID: 21328307 DOI: 10.1002/14651858.cd008216.pub3] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
BACKGROUND Soft tissue sarcomas (STS) are a highly heterogeneous group of rare malignant solid tumors. Non-rhabdomyosarcoma soft tissue sarcomas (NRSTS) comprise all STS except rhabdomyosarcoma. In patients with advanced local or metastatic disease, autologous hematopoietic stem cell transplantation (HSCT) applied after high-dose chemotherapy (HDCT) is a planned rescue therapy for HDCT-related severe hematologic toxicity. OBJECTIVES To assess the effectiveness and safety of HDCT followed by autologous HSCT for all stages of soft tissue sarcomas in children and adults. SEARCH STRATEGY We searched the electronic databases CENTRAL (The Cochrane Library 2010, Issue 2), MEDLINE and EMBASE (February 2010). Online trial registers, congress abstracts and reference lists of reviews were searched and expert panels and authors were contacted. SELECTION CRITERIA Terms representing STS and autologous HSCT were required in the title, abstract or keywords. In studies with aggregated data, participants with NRSTS and autologous HSCT had to constitute at least 80% of the data. Comparative non-randomized studies were included because randomized controlled trials (RCTs) were not expected. Case series and case reports were considered for an additional descriptive analysis. DATA COLLECTION AND ANALYSIS Study data were recorded by two review authors independently. For studies with no comparator group, we synthesised results for studies reporting aggregate data and conducted a pooled analysis of individual participant data using the Kaplan-Meyer method. The primary outcomes were overall survival (OS) and treatment-related mortality (TRM). MAIN RESULTS We included 54 studies, from 467 full texts articles screened (11.5%), reporting on 177 participants that received HSCT and 69 participants that received standard care. Only one study reported comparative data. In the one comparative study, OS at two years after HSCT was estimated as statistically significantly higher (62.3%) compared with participants that received standard care (23.2%). In a single-arm study, the OS two years after HSCT was reported as 20%. In a pooled analysis of the individual data of 54 participants, OS at two years was estimated as 49% (95% CI 34% to 64%). Data on TRM, secondary neoplasia and severe toxicity grade 3 to 4 after transplantation were sparse. All 54 studies had a high risk of bias. AUTHORS' CONCLUSIONS Due to a lack of comparative studies, it is unclear whether participants with NRSTS have improved survival from autologous HSCT following HDCT. Owing to this current gap in knowledge, at present HDCT and autologous HSCT for NRSTS should only be used within controlled trials.
Collapse
Affiliation(s)
- Frank Peinemann
- Department of Non-Drug Interventions, Institute for Quality and Efficiency in Health Care (IQWiG), Dillenburger Str. 27, Cologne, Germany, 51105
| | | | | | | | | | | |
Collapse
|
45
|
Abstract
Explorative knowledge of cellular and molecular mechanisms of immune function and regulation has provided optimism in developing cancer immunotherapy. However, three decades of experimental and clinical investigations to offer powerful immunotherapeutic strategies against solid tumors, with the possible exception of monoclonal antibody-targeted therapies, have not succeeded in significantly prolonging patient survival. Nonspecific immune approaches, including cytokine-based therapies and allogeneic hematopoietic stem cell transplantation, have so far produced consistent, although limited, results. In this review, we present the developments of cell transfer-based strategies that, in preclinical studies, have demonstrated potential efficacy, but have only established tumor regression in limited numbers of patients. The key to success demands creative combinations of tumor antigens, adjuvance, gene modification and various administration strategies in the development of cell-based therapies together with other cancer-treatment principles, often in a stepwise 'space-rocket-type' approach. Combined efforts of several scientific disciplines, such as tumor biology and immunology, as well as cell and gene research in transplantation, will open new venues. New regulation for clinical trials with advanced therapy medicine products to ensure patient safety will be highlighted.
Collapse
Affiliation(s)
- Lisbeth Barkholt
- Division of Clinical Immunology & Transfusion Medicine, Department of Laboratory Medicine, Karolinska University Hospital Huddinge F79, Stockholm, Sweden.
| | | |
Collapse
|
46
|
Jubert C, Wall DA, Grimley M, Champagne MA, Duval M. Engraftment of unrelated cord blood after reduced-intensity conditioning regimen in children with refractory neuroblastoma: a feasibility trial. Bone Marrow Transplant 2011; 46:232-7. [PMID: 20436519 DOI: 10.1038/bmt.2010.107] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2009] [Revised: 03/19/2010] [Accepted: 03/26/2010] [Indexed: 11/09/2022]
Abstract
Relapsed or refractory neuroblastoma is uniformly fatal. We hypothesized that allogeneic response could provide a platform for immunotherapy in neuroblastoma. We therefore undertook a pilot trial of unrelated cord blood transplantation after reduced intensity conditioning regimen (RIC) in children with relapsed neuroblastoma to assess engraftment and tolerability in this heavily pretreated population. The RIC included CY (50 mg/kg, day -6), fludarabine (40 mg/m(2), days -6 to -2), total body irradiation (200 cGy, day -1), and rabbit anti-thymocyte globulin (2.5 mg/kg, days -3 to -1). Six patients were enrolled: four were in partial responsive relapse, one with a mixed response and one in refractory relapse. All patients tolerated the regimen well and had donor engraftment with full neutrophil and plt recovery (median time 12 and 35 days, respectively). One patient never experienced neutropenia and another did not need plt transfusions. All patients progressed after transplant (median time 55 days, 26-180 days). Natural killer (NK) cell counts were normal within 2 months, whereas T-cell recovery was slower. In conclusion, unrelated cord blood engrafts after RIC in children with refractory neuroblastoma. Future research should be aimed at transplanting patients with minimal residual disease, using less intensive immunosuppression and adding NK-cell based post transplant immunotherapy.
Collapse
Affiliation(s)
- C Jubert
- Unité d'Hématologie-Oncologie Pédiatrique, Hôpital des Enfants, CHU Bordeaux, France
| | | | | | | | | |
Collapse
|
47
|
Ash S, Stein J, Askenasy N, Yaniv I. Immunomodulation with dendritic cells and donor lymphocyte infusion converge to induce graft vs neuroblastoma reactions without GVHD after allogeneic bone marrow transplantation. Br J Cancer 2010; 103:1597-605. [PMID: 20978501 PMCID: PMC2990575 DOI: 10.1038/sj.bjc.6605924] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Revised: 08/31/2010] [Accepted: 09/02/2010] [Indexed: 11/09/2022] Open
Abstract
BACKGROUND Mounting evidence points to the efficacy of donor lymphocyte infusion (DLI) and immunisation with tumour-pulsed dendritic cells (DC) in generating graft vs leukaemia reactions after allogeneic bone marrow transplantation (BMT). We assessed the efficacy of DLI and DC in generating potent graft vs neuroblastoma tumour (GVT) reactions following allogeneic BMT. METHODS Mice bearing congenic (H2K(a)) Neuro-2a tumours were grafted with allogeneic (H2K(b)) T-cell-depleted bone marrow cells. Tumour-pulsed donor DC (DC(Neuro2a)) were inoculated (on day +7) in conjunction with donor (H2K(b)) and haploidentical (H2K(a/b)) lymphocytes. RESULTS Murine Neuro-2a cells elicit immune reactions as efficient as B lymphoma in major histocompatibility complex antigen-disparate mice. Lymphopenia induced by conditioning facilitates GVT, and transition to adaptive immunity is enhanced by simultaneous infusion of and DC(Neuro2a) and lymphocytes devoid of graft vs host (GVH) activity (H2K(a/b)). In variance, the efficacy of DC-mediated immunomodulation was diminished by severe graft vs host disease (GVHD), showing mechanistic dissociation and antagonising potential to GVT. CONCLUSIONS The GVHD is not a prerequisite to induce GVT reactivity after allogeneic BMT, but is rather detrimental to induction of anti-tumour immunity by DC-mediated immunomodulation. Simultaneous inoculation of tumour-pulsed donor DC and DLI synergise in stimulation of potent GVT reactions to the extent of eradication of established NB tumours.
Collapse
Affiliation(s)
- S Ash
- Department of Pediatric Hematology-Oncology, Schneider Children's Medical Center of Israel, Petach Tikva 49202, Israel
- Zaizov Cancer Immunotherapy Laboratory, Schneider Children's Medical Center of Israel, Petach Tikva 49202, Israel
| | - J Stein
- Department of Pediatric Hematology-Oncology, Schneider Children's Medical Center of Israel, Petach Tikva 49202, Israel
- Pediatric Bone Marrow Transplant Unit, Schneider Children's Medical Center of Israel, Petach Tikva 49202, Israel
| | - N Askenasy
- Frankel Laboratory of Experimental Bone Marrow Transplantation, Schneider Children's Medical Center of Israel, 14 Kaplan Street, Petach Tikva 49202, Israel
| | - I Yaniv
- Department of Pediatric Hematology-Oncology, Schneider Children's Medical Center of Israel, Petach Tikva 49202, Israel
- Zaizov Cancer Immunotherapy Laboratory, Schneider Children's Medical Center of Israel, Petach Tikva 49202, Israel
| |
Collapse
|
48
|
Zhong XY, Zhang B, Asadollahi R, Low SH, Holzgreve W. Umbilical cord blood stem cells: what to expect. Ann N Y Acad Sci 2010; 1205:17-22. [DOI: 10.1111/j.1749-6632.2010.05659.x] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
49
|
Lupo-Stanghellini MT, Provasi E, Bondanza A, Ciceri F, Bordignon C, Bonini C. Clinical impact of suicide gene therapy in allogeneic hematopoietic stem cell transplantation. Hum Gene Ther 2010; 21:241-50. [PMID: 20121594 DOI: 10.1089/hum.2010.014] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Allogeneic hematopoietic stem cell transplantation (allo-SCT) from an HLA-matched related or unrelated donor is a curative option for patients with high-risk hematological diseases. In the absence of a matched donor, patients have been offered investigational transplantation strategies such as umbilical cord blood SCT or family haploidentical SCT. Besides the activity of the conditioning regimen, most of the antileukemic potential of allo-SCT relies on alloreactivity, promoted by donor lymphocytes reacting against patient-specific antigens, such as minor and major histocompatibility antigens, ultimately translating into cancer immunotherapy. Unfortunately, alloreactivity is also responsible for the most serious and frequent complication of allo-SCT: graft-versus-host-disease (GvHD). The risk of GvHD increases with the level of HLA disparity between host and donor, and leads to impaired quality of life and reduced survival expectancy, particularly among patients receiving transplants from HLA-mismatched donors. Gene transfer technologies are promising tools to manipulate donor T cell immunity to enforce the graft-versus-tumor effect, to promote functional immune reconstitution (graft vs. infection), and to prevent or control GvHD. To this purpose, several cell and gene transfer approaches have been investigated at the preclinical level, and are being implemented in clinical trials. Suicide gene therapy is to date the most extensive clinical application of T cell-based gene therapy. In several phase I-II clinical studies conducted worldwide this approach proved highly feasible, safe, and effective in promoting a dynamic and patient-specific modulation of alloreactivity. This review focuses on this approach.
Collapse
Affiliation(s)
- Maria Teresa Lupo-Stanghellini
- Hematology and Bone Marrow Transplantation Unit, Division of Regenerative Medicine, Gene Therapy, and Stem Cells, Program in Immunology, Gene Therapy, and Bioimmunotherapy of Cancer, San Raffaele Scientific Institute, 20132 Milan, Italy
| | | | | | | | | | | |
Collapse
|
50
|
Hess Michelini R, Freschi M, Manzo T, Jachetti E, Degl'Innocenti E, Grioni M, Basso V, Bonini C, Simpson E, Mondino A, Bellone M. Concomitant tumor and minor histocompatibility antigen-specific immunity initiate rejection and maintain remission from established spontaneous solid tumors. Cancer Res 2010; 70:3505-14. [PMID: 20388780 DOI: 10.1158/0008-5472.can-09-4253] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Nonmyeloablative hematopoietic cell transplantation can cure patients with hematologic malignancies but has reported limited success against solid tumors. This is possibly because of profound peripheral tolerance mechanisms and/or suboptimal tumor recognition by effector T lymphocytes. We report that in mice developing spontaneous prostate cancer, nonmyeloablative minor histocompatibility mismatched hematopoietic stem cell transplantation, and donor lymphocyte infusion of unmanipulated lymphocytes combined with posttransplant tumor-specific vaccination circumvents tumor-specific tolerance, allowing acute tumor rejection and the establishment of protective immunosurveillance. Although donor-derived tumor-specific T cells readily differentiated into effector cells and infiltrated the tumor soon after infusion, they were alone insufficient for tumor eradication, which instead required the concomitance of minor histocompatibiltiy antigen-specific CD8(+) T-cell responses. The establishment of protective immunosurveillance was best induced by posttransplant tumor-specific vaccination. Hence, these results provide the proof of principle that tumor-specific T-cell responses have to be harnessed together with minor histocompatibility responses and sustained by posttransplant tumor-specific vaccination to improve the efficacy of allotransplantion for the cure of solid tumors.
Collapse
Affiliation(s)
- Rodrigo Hess Michelini
- Lymphocyte Activation Unit, Cellular Immunology Unit, Pathology Unit, Experimental Hematology, San Raffaele Scientific Institute, and Universita Vita-Salute San Raffaele, Milan, Italy
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|