1
|
Riley JS, McClain LE, Stratigis JD, Coons BE, Bose SK, Dave A, White BM, Li H, Loukogeorgakis SP, Fachin CG, Dias AIBS, Flake AW, Peranteau WH. Fetal allotransplant recipients are resistant to graft-versus-host disease. Exp Hematol 2023; 118:31-39.e3. [PMID: 36535408 PMCID: PMC9898145 DOI: 10.1016/j.exphem.2022.12.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Accepted: 12/10/2022] [Indexed: 12/23/2022]
Abstract
In utero hematopoietic cell transplantation (IUHCT) is an experimental treatment for congenital hemoglobinopathies, including Sickle cell disease and thalassemias. One of the principal advantages of IUHCT is the predisposition of the developing fetus toward immunologic tolerance. This allows for engraftment across immune barriers without immunosuppression and, potentially, decreased susceptibility to graft-versus-host disease (GVHD). We demonstrate fetal resistance to GVHD following T cell-replete allogeneic hematopoietic cell transplantation compared with the neonate. We show that this resistance is associated with elevated fetal serum interleukin-10 conducive to the induction of regulatory T cells (Tregs). Finally, we demonstrate that the adoptive transfer of Tregs from IUHCT recipients to neonates uniformly prevents GVHD, recapitulating the predisposition to tolerance observed after fetal allotransplantation. These findings demonstrate fetal resistance to GVHD following hematopoietic cell transplantation and elucidate Tregs as important contributors.
Collapse
Affiliation(s)
- John S Riley
- Center for Fetal Research, The Children's Hospital of Philadelphia, Philadelphia, PA
| | - Lauren E McClain
- Center for Fetal Research, The Children's Hospital of Philadelphia, Philadelphia, PA
| | - John D Stratigis
- Center for Fetal Research, The Children's Hospital of Philadelphia, Philadelphia, PA
| | - Barbara E Coons
- Center for Fetal Research, The Children's Hospital of Philadelphia, Philadelphia, PA
| | - Sourav K Bose
- Center for Fetal Research, The Children's Hospital of Philadelphia, Philadelphia, PA
| | - Apeksha Dave
- Center for Fetal Research, The Children's Hospital of Philadelphia, Philadelphia, PA
| | - Brandon M White
- Center for Fetal Research, The Children's Hospital of Philadelphia, Philadelphia, PA
| | - Haiying Li
- Center for Fetal Research, The Children's Hospital of Philadelphia, Philadelphia, PA
| | | | - Camila G Fachin
- Center for Fetal Research, The Children's Hospital of Philadelphia, Philadelphia, PA
| | - Andre I B S Dias
- Center for Fetal Research, The Children's Hospital of Philadelphia, Philadelphia, PA
| | - Alan W Flake
- Center for Fetal Research, The Children's Hospital of Philadelphia, Philadelphia, PA
| | - William H Peranteau
- Center for Fetal Research, The Children's Hospital of Philadelphia, Philadelphia, PA.
| |
Collapse
|
2
|
Berkowitz CL, Luks VL, Puc M, Peranteau WH. Molecular and Cellular In Utero Therapy. Clin Perinatol 2022; 49:811-820. [PMID: 36328600 DOI: 10.1016/j.clp.2022.06.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Significant advances in maternal-fetal medicine and gene sequencing technology have fostered a new frontier of in utero molecular and cellular therapeutics, including gene editing, enzyme replacement therapy, and stem cell transplantation to treat single-gene disorders with limited postnatal treatment strategies. In utero therapies take advantage of unique developmental properties of the fetus to allow for the correction of monogenic disorders before irreversible disease pathology develops. While early preclinical studies in animal models are encouraging, more studies are needed to further evaluate their safety and efficacy prior to widespread clinical use.
Collapse
Affiliation(s)
- Cara L Berkowitz
- Division of Pediatric General, Thoracic and Fetal Surgery, Children's Hospital of Philadelphia, 3401 Civic Center Boulevard, Philadelphia, PA 19104, USA
| | - Valerie L Luks
- Division of Pediatric General, Thoracic and Fetal Surgery, Children's Hospital of Philadelphia, 3401 Civic Center Boulevard, Philadelphia, PA 19104, USA
| | - Marcelina Puc
- Division of Pediatric General, Thoracic and Fetal Surgery, Children's Hospital of Philadelphia, 3401 Civic Center Boulevard, Philadelphia, PA 19104, USA
| | - William H Peranteau
- Division of Pediatric General, Thoracic and Fetal Surgery, Children's Hospital of Philadelphia, 3401 Civic Center Boulevard, Philadelphia, PA 19104, USA.
| |
Collapse
|
3
|
Shi C, Pan L, Hu Z. Experimental and clinical progress of in utero hematopoietic cell transplantation therapy for congenital disorders. Front Pharmacol 2022; 13:851375. [PMID: 36120324 PMCID: PMC9478511 DOI: 10.3389/fphar.2022.851375] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2022] [Accepted: 08/08/2022] [Indexed: 11/13/2022] Open
Abstract
In utero hematopoietic cell transplantation (IUHCT) is considered a potentially efficient therapeutic approach with relatively few side effects, compared to adult hematopoietic cell transplantation, for various hematological genetic disorders. The principle of IUHCT has been extensively studied in rodent models and in some large animals with close evolutionary similarities to human beings. However, IUHCT has only been used to rebuild human T cell immunity in certain patients with inherent immunodeficiencies. This review will first summarize the animal models utilized for IUHCT investigations and describe the associated outcomes. Recent advances and potential barriers for successful IUHCT are discussed, followed by possible strategies to overcome these barriers experimentally. Lastly, we will outline the progress made towards utilizing IUHCT to treat inherent disorders for patients, list out associated limitations and propose feasible means to promote the efficacy of IUHCT clinically.
Collapse
Affiliation(s)
- Chunyu Shi
- National-Local Joint Engineering Laboratory of Animal Models for Human Diseases, The First Hospital of Jilin University, Changchun, China
- Department of Gastrointestinal Colorectal and Anal Surgery, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Lu Pan
- Department of Pediatric Immunology, Allergy and Rheumatology, The First Hospital of Jilin University, Changchun, China
| | - Zheng Hu
- National-Local Joint Engineering Laboratory of Animal Models for Human Diseases, The First Hospital of Jilin University, Changchun, China
- *Correspondence: Zheng Hu,
| |
Collapse
|
4
|
Kandasamy K, Tan LG, B Johana N, Tan YW, Foo W, Yeo JSL, Ravikumar V, Ginhoux F, Choolani M, Chan JKY, Mattar CNZ. Maternal microchimerism and cell-mediated immune-modulation enhance engraftment following semi-allogenic intrauterine transplantation. FASEB J 2021; 35:e21413. [PMID: 33570785 DOI: 10.1096/fj.202002185rr] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 01/04/2021] [Accepted: 01/20/2021] [Indexed: 11/11/2022]
Abstract
Successful intrauterine hematopoietic cell transplantation (IUT) for congenital hemoglobinopathies is hampered by maternal alloresponsiveness. We investigate these interactions in semi-allogenic murine IUT. E14 fetuses (B6 females × BALB/c males) were each treated with 5E+6 maternal (B6) or paternal (BALB/c) bone marrow cells and serially monitored for chimerism (>1% engraftment), trafficked maternal immune cells, and immune responsiveness to donor cells. A total of 41.0% of maternal IUT recipients (mIUT) were chimeras (mean donor chimerism 3.0 ± 1.3%) versus 75.0% of paternal IUT recipients (pIUT, 3.6 ± 1.1%). Chimeras showed higher maternal microchimerism of CD4, CD8, and CD19 than non-chimeras. These maternal cells showed minimal responsiveness to B6 or BALB/c stimulation. To interrogate tolerance, mIUT were injected postnatally with 5E+6 B6 cells/pup; pIUT received BALB/c cells. IUT-treated pups showed no changes in trafficked maternal or fetal immune cell levels compared to controls. Donor-specific IgM and IgG were expressed by 1%-3% of recipients. mIUT splenocytes showed greater proliferation of regulatory T cells (Treg) upon BALB/c stimulation, while B6 stimulation upregulated the pro-inflammatory cytokines more than BALB/c. pIUT splenocytes produced identical Treg and cytokine responses to BALB/c and B6 cells, with higher Treg activity and lower pro-inflammatory cytokine expression upon exposure to BALB/c. In contrast, naïve fetal splenocytes demonstrated greater alloresponsiveness to BALB/c compared to B6 cells. Thus pIUT, associated with increased maternal cell trafficking, modulates fetal Treg, and cytokine responsiveness to donor cells more efficiently than mIUT, resulting in improved engraftment. Paternal donor cells may be considered alternatively to maternal donor cells for intrauterine and postnatal transplantation to induce tolerance and maintain engraftment.
Collapse
Affiliation(s)
- Karthikeyan Kandasamy
- Experimental Fetal Medicine Group, Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Lay Geok Tan
- Experimental Fetal Medicine Group, Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,Department of Obstetrics and Gynaecology, National University Hospital, National University Health System, Singapore, Singapore
| | - Nuryanti B Johana
- Reproductive Medicine, KK Women's and Children's Hospital, Singapore, Singapore
| | - Yi Wan Tan
- Reproductive Medicine, KK Women's and Children's Hospital, Singapore, Singapore
| | - Wanling Foo
- Experimental Fetal Medicine Group, Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Julie S L Yeo
- Reproductive Medicine, KK Women's and Children's Hospital, Singapore, Singapore
| | - Vikashini Ravikumar
- Experimental Fetal Medicine Group, Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Florent Ginhoux
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Mahesh Choolani
- Experimental Fetal Medicine Group, Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,Department of Obstetrics and Gynaecology, National University Hospital, National University Health System, Singapore, Singapore
| | - Jerry K Y Chan
- Reproductive Medicine, KK Women's and Children's Hospital, Singapore, Singapore.,Cancer and Stem Cell Biology Program, Duke-NUS Graduate Medical School, Singapore, Singapore
| | - Citra N Z Mattar
- Experimental Fetal Medicine Group, Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,Department of Obstetrics and Gynaecology, National University Hospital, National University Health System, Singapore, Singapore
| |
Collapse
|
5
|
Regulatory T cells promote alloengraftment in a model of late-gestation in utero hematopoietic cell transplantation. Blood Adv 2021; 4:1102-1114. [PMID: 32203584 DOI: 10.1182/bloodadvances.2019001208] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Accepted: 02/22/2020] [Indexed: 12/15/2022] Open
Abstract
In utero hematopoietic cell transplantation (IUHCT) has the potential to cure congenital hematologic disorders including sickle cell disease. However, the window of opportunity for IUHCT closes with the acquisition of T-cell immunity, beginning at approximately 14 weeks gestation, posing significant technical challenges and excluding from treatment fetuses evaluated after the first trimester. Here we report that regulatory T cells can promote alloengraftment and preserve allograft tolerance after the acquisition of T-cell immunity in a mouse model of late-gestation IUHCT. We show that allografts enriched with regulatory T cells harvested from either IUHCT-tolerant or naive mice engraft at 20 days post coitum (DPC) with equal frequency to unenriched allografts transplanted at 14 DPC. Long-term, multilineage donor cell chimerism was achieved in the absence of graft-versus-host disease or mortality. Decreased alloreactivity among recipient T cells was observed consistent with donor-specific tolerance. These findings suggest that donor graft enrichment with regulatory T cells could be used to successfully perform IUHCT later in gestation.
Collapse
|
6
|
Tai-MacArthur S, Lombardi G, Shangaris P. The Theoretical Basis of In Utero Hematopoietic Stem Cell Transplantation and Its Use in the Treatment of Blood Disorders. Stem Cells Dev 2021; 30:49-58. [PMID: 33280478 DOI: 10.1089/scd.2020.0181] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Since its conception, prenatal therapy has been successful in correction of mainly anatomical defects, although the range of application has been limited. Research into minimally invasive fetal surgery techniques and prenatal molecular diagnostics has facilitated the development of in utero stem cell transplantation (IUT)-a method of delivering healthy stem cells to the early gestation fetus with the hope of engraftment, proliferation, and migration to the appropriate hematopoietic compartment. An area of application that shows promise is the treatment of hematopoietic disorders like hemoglobinopathies. The therapeutic rationale of IUT with hematopoietic stem cells (HSCs) is based on the proposed advantages the fetal environment offers based on its unique physiology. These advantages include the immature immune system facilitating the development of donor-specific tolerance, the natural migration of endogenous hematopoietic cells providing space for homing and engraftment of donor cells, and the fetal environment providing HSCs with the same opportunity to survive and proliferate regardless of their origin (donor or host). Maternal immune tolerance to the fetus and placenta also implies that the maternal environment could be accepting of donor cells. In theory, the fetus is a perfect recipient for stem cell transplant. Clinically, however, IUT is yet to see widespread success calling into question these assumptions of fetal physiology. This review aims to discuss and evaluate research surrounding these key assumptions and the clinical success of IUT in the treatment of thalassemia.
Collapse
Affiliation(s)
- Sarah Tai-MacArthur
- School of Basic and Medical Biosciences, Faculty of Life Sciences and Medicine, King's College London, United Kingdom
| | - Giovanna Lombardi
- Immunoregulation Laboratory, School of Immunology, Microbial Sciences, and Faculty of Life Sciences and Medicine, King's College London, United Kingdom
| | - Panicos Shangaris
- Immunoregulation Laboratory, School of Immunology, Microbial Sciences, and Faculty of Life Sciences and Medicine, King's College London, United Kingdom.,School of Life Course Sciences, Faculty of Life Sciences and Medicine, King's College London, United Kingdom
| |
Collapse
|
7
|
Donor cell engineering with GSK3 inhibitor-loaded nanoparticles enhances engraftment after in utero transplantation. Blood 2020; 134:1983-1995. [PMID: 31570489 DOI: 10.1182/blood.2019001037] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Accepted: 08/23/2019] [Indexed: 01/04/2023] Open
Abstract
Host cell competition is a major barrier to engraftment after in utero hematopoietic cell transplantation (IUHCT). Here we describe a cell-engineering strategy using glycogen synthase kinase-3 (GSK3) inhibitor-loaded nanoparticles conjugated to the surface of donor hematopoietic cells to enhance their proliferation kinetics and ability to compete against their fetal host equivalents. With this approach, we achieved remarkable levels of stable, long-term hematopoietic engraftment for up to 24 weeks post-IUHCT. We also show that the salutary effects of the nanoparticle-released GSK3 inhibitor are specific to donor progenitor/stem cells and achieved by a pseudoautocrine mechanism. These results establish that IUHCT of hematopoietic cells decorated with GSK3 inhibitor-loaded nanoparticles can produce therapeutic levels of long-term engraftment and could therefore allow single-step prenatal treatment of congenital hematological disorders.
Collapse
|
8
|
Vrecenak JD, Partridge EA, Pearson EG, Flake AW. Simple Approach to Increase Donor Hematopoietic Stem Cell Dose and Improve Engraftment in the Murine Model of Allogeneic In Utero Hematopoietic Cell Transplantation. Biol Blood Marrow Transplant 2019; 26:e21-e24. [PMID: 31493540 DOI: 10.1016/j.bbmt.2019.08.024] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Revised: 08/07/2019] [Accepted: 08/22/2019] [Indexed: 11/19/2022]
Abstract
The rationale for in utero hematopoietic cell transplantation (IUHCT) rests on exploitation of normal events during hematopoietic and immunologic ontogeny to allow allogeneic hematopoietic engraftment without myeloablative conditioning. Host hematopoietic competition is among the primary barriers to engraftment in IUHCT. In the murine model this can be partially overcome by delivery of larger donor cell doses, but volume is limiting. Enrichment of donor hematopoietic stem cells (HSCs) would seem to offer a more efficient approach, but such enriched populations have engrafted poorly in existing models of IUHCT. To increase HSC dose while maintaining the presence of accessory cells, we used a less stringent enrichment protocol of single-step lineage depleted cells alone (lin-) or in combination with whole donor bone marrow mononuclear cells. Our results confirm that increasing doses of HSCs in combination with bone marrow accessory cells can dramatically improve engraftment after IUHCT. This represents a practical and clinically applicable strategy to maximize the engraftment potential of the donor graft without risk of treatment-associated toxicity.
Collapse
Affiliation(s)
- Jesse D Vrecenak
- Division of Pediatric Surgery, Washington University, St. Louis, Missouri
| | - Emily A Partridge
- Center for Fetal Research, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Erik G Pearson
- Center for Fetal Research, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Alan W Flake
- Center for Fetal Research, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania.
| |
Collapse
|
9
|
Loukogeorgakis SP, Shangaris P, Bertin E, Franzin C, Piccoli M, Pozzobon M, Subramaniam S, Tedeschi A, Kim AG, Li H, Fachin CG, Dias AIBS, Stratigis JD, Ahn NJ, Thrasher AJ, Bonfanti P, Peranteau WH, David AL, Flake AW, De Coppi P. In Utero Transplantation of Expanded Autologous Amniotic Fluid Stem Cells Results in Long-Term Hematopoietic Engraftment. Stem Cells 2019; 37:1176-1188. [PMID: 31116895 PMCID: PMC6773206 DOI: 10.1002/stem.3039] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2018] [Revised: 12/06/2018] [Accepted: 12/23/2018] [Indexed: 12/20/2022]
Abstract
In utero transplantation (IUT) of hematopoietic stem cells (HSCs) has been proposed as a strategy for the prenatal treatment of congenital hematological diseases. However, levels of long‐term hematopoietic engraftment achieved in experimental IUT to date are subtherapeutic, likely due to host fetal HSCs outcompeting their bone marrow (BM)‐derived donor equivalents for space in the hematopoietic compartment. In the present study, we demonstrate that amniotic fluid stem cells (AFSCs; c‐Kit+/Lin−) have hematopoietic characteristics and, thanks to their fetal origin, favorable proliferation kinetics in vitro and in vivo, which are maintained when the cells are expanded. IUT of autologous/congenic freshly isolated or cultured AFSCs resulted in stable multilineage hematopoietic engraftment, far higher to that achieved with BM‐HSCs. Intravascular IUT of allogenic AFSCs was not successful as recently reported after intraperitoneal IUT. Herein, we demonstrated that this likely due to a failure of timely homing of donor cells to the host fetal thymus resulted in lack of tolerance induction and rejection. This study reveals that intravascular IUT leads to a remarkable hematopoietic engraftment of AFSCs in the setting of autologous/congenic IUT, and confirms the requirement for induction of central tolerance for allogenic IUT to be successful. Autologous, gene‐engineered, and in vitro expanded AFSCs could be used as a stem cell/gene therapy platform for the in utero treatment of inherited disorders of hematopoiesis. stem cells2019;37:1176–1188
Collapse
Affiliation(s)
- Stavros P Loukogeorgakis
- Stem Cells and Regenerative Medicine, Great Ormond Street Institute of Child Health, University College London, London, United Kingdom.,Center for Fetal Research, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Panicos Shangaris
- Stem Cells and Regenerative Medicine, Great Ormond Street Institute of Child Health, University College London, London, United Kingdom.,Research Department of Maternal and Fetal Medicine, Institute for Women's Health, University College London, London, United Kingdom
| | - Enrica Bertin
- Stem Cell and Regenerative Medicine Laboratory, Fondazione Instituto di Ricerca Pediatrica Città della Speranza, University of Padova, Padova, Italy
| | - Chiara Franzin
- Stem Cell and Regenerative Medicine Laboratory, Fondazione Instituto di Ricerca Pediatrica Città della Speranza, University of Padova, Padova, Italy
| | - Martina Piccoli
- Stem Cell and Regenerative Medicine Laboratory, Fondazione Instituto di Ricerca Pediatrica Città della Speranza, University of Padova, Padova, Italy.,Department of Women's and Children's Health, University of Padova, Padova, Italy
| | - Michela Pozzobon
- Stem Cell and Regenerative Medicine Laboratory, Fondazione Instituto di Ricerca Pediatrica Città della Speranza, University of Padova, Padova, Italy
| | - Sindhu Subramaniam
- Stem Cells and Regenerative Medicine, Great Ormond Street Institute of Child Health, University College London, London, United Kingdom
| | - Alfonso Tedeschi
- Stem Cells and Regenerative Medicine, Great Ormond Street Institute of Child Health, University College London, London, United Kingdom
| | - Aimee G Kim
- Center for Fetal Research, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Haiying Li
- Center for Fetal Research, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Camila G Fachin
- Center for Fetal Research, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA.,Federal University of São Paulo, São Paulo, Brazil.,Federal University of Paraná, Curitiba, Brazil
| | - Andre I B S Dias
- Stem Cells and Regenerative Medicine, Great Ormond Street Institute of Child Health, University College London, London, United Kingdom.,Federal University of São Paulo, São Paulo, Brazil.,Federal University of Paraná, Curitiba, Brazil
| | - John D Stratigis
- Center for Fetal Research, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Nicholas J Ahn
- Center for Fetal Research, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Adrian J Thrasher
- Molecular and Cellular Immunology Section, Great Ormond Street Institute of Child Health, University College London, London, United Kingdom
| | - Paola Bonfanti
- Stem Cells and Regenerative Medicine, Great Ormond Street Institute of Child Health, University College London, London, United Kingdom.,The Francis Crick Institute, London, United Kingdom
| | - William H Peranteau
- Center for Fetal Research, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Anna L David
- Research Department of Maternal and Fetal Medicine, Institute for Women's Health, University College London, London, United Kingdom
| | - Alan W Flake
- Center for Fetal Research, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Paolo De Coppi
- Stem Cells and Regenerative Medicine, Great Ormond Street Institute of Child Health, University College London, London, United Kingdom
| |
Collapse
|
10
|
Ghafarzadeh M, Namdari P, Tarhani M, Tarhani F. A review of application of stem cell therapy in the management of congenital heart disease. J Matern Fetal Neonatal Med 2018; 33:1607-1615. [PMID: 30185081 DOI: 10.1080/14767058.2018.1520829] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Research on stem cells has been rapidly growing with impressive breakthroughs. Although merely a few of the laboratory researches have successfully transited to the clinical trial phase, the application of stem cells as a therapeutic option for some currently incapacitating diseases hold fascinating potentials. This review emphasis the various opportunities for the application of stem cell in the treatment of fetal diseases. First, we provide a brief commentary on the common stem cell strategy used in the treatment of congenital anomalies, thereafter we discuss how stem cell is being used in the management of some fetal disorders.
Collapse
Affiliation(s)
- Masoumeh Ghafarzadeh
- Faculty of Medicine, Department of Obstetrics and Genecology, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Parsa Namdari
- University of Debrecen Medical School, Debrecen, Hungary
| | - Mehrnoosh Tarhani
- Research Committee Student, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Fariba Tarhani
- Faculty of Medicine, Department of Paediatrics, Lorestan University of Medical Sciences, Khorramabad, Iran
| |
Collapse
|
11
|
Vrecenak JD, Pearson EG, Todorow CA, Li H, Johnson MP, Flake AW. Preclinical Canine Model of Graft-versus-Host Disease after In Utero Hematopoietic Cell Transplantation. Biol Blood Marrow Transplant 2018; 24:1795-1801. [PMID: 29802901 DOI: 10.1016/j.bbmt.2018.05.020] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Accepted: 05/15/2018] [Indexed: 12/12/2022]
Abstract
In utero hematopoietic cell transplantation (IUHCT) offers the potential to achieve allogeneic engraftment and associated donor-specific tolerance without the need for toxic conditioning, as we have previously demonstrated in the murine and canine models. This strategy holds great promise in the treatment of many hematopoietic disorders, including the hemoglobinopathies. Graft-versus-host disease (GVHD) represents the greatest theoretical risk of IUHCT and has never been characterized in the context of IUHCT. We recently described a preclinical canine model of IUHCT, allowing further study of the technique and its complications. We aimed to establish a threshold T cell dose for IUHCT-induced GVHD in the haploidentical canine model and to define the GVHD phenotype. Using a range of T cell concentrations within the donor inoculum, we were able to characterize the phenotype of IUHCT-induced GVHD and establish a clear threshold for its induction between 3% and 5% graft CD3+ cell content. Given the complete absence of GVHD at CD3 doses of 1% to 3% and the excellent engraftment with the lowest dose, there is a safe therapeutic index for a clinical trial of IUHCT.
Collapse
Affiliation(s)
- Jesse D Vrecenak
- Children's Center for Fetal Research, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania; Division of Pediatric Surgery, Washington University in St. Louis, St. Louis, Missouri
| | - Erik G Pearson
- Children's Center for Fetal Research, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Carlyn A Todorow
- Children's Center for Fetal Research, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Haiying Li
- Children's Center for Fetal Research, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Mark P Johnson
- Children's Center for Fetal Research, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Alan W Flake
- Children's Center for Fetal Research, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania.
| |
Collapse
|
12
|
Alhajjat A, Shaaban A. Maternal and Fetal Immune Response to in Utero Stem Cell Transplantation. CURRENT STEM CELL REPORTS 2018; 4:182-187. [PMID: 30873337 DOI: 10.1007/s40778-018-0129-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Purpose of Review In Utero Hematopoietic Cellular Transplantation (IUHCT) is a promising intervention for the non-toxic treatment of congenital disease that hinges on the assumption of fetal immunologic immaturity and an inability to reject a hematopoietic allograft. However, clinical IUCHT has failed except in cases where the fetus is severely immunocompromised. The current review examines recent studies of engraftment barriers stemming from either the fetal or maternal immune system. Recent Findings New reports have illuminated roles for maternal humoral and cellular immunity and fetal innate cellular immunity in the resistance to allogeneic IUHCT. These experimental findings have inspired new approaches to overcome these barriers. Despite these advances, postulates regarding a maternal immune barrier to IUHCT provide an inadequate explanation for the well-documented clinical success only in the treatment of fetal immunodeficiency with normal maternal immunity. Summary Characterization of the maternal and fetal immune response to allogeneic IUHCT provides new insight into the complexity of prenatal tolerance. Future work in this area should aim to provide a unifying explanation for the observed patterns of success and failure with clinical IUHCT.
Collapse
Affiliation(s)
- Amir Alhajjat
- Division of Pediatric Surgery, Phoenix Children's Hospital, 1919 E Thomas Rd, Phoenix, Arizona
| | - Aimen Shaaban
- The Chicago Institute for Fetal Health, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, Illinois
- Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| |
Collapse
|
13
|
Abstract
BACKGROUND In utero transplantation (IUT) of hematopoietic stem cells has the potential to treat a large number of hematologic and metabolic diseases amenable to partial replacement of the hematopoietic system. METHODS A review of the literature was conducted that focused on the clinical and experimental experience with IUT and, in this context, the development of the hematopoietic and immune systems. RESULTS Successful application of IUT has been limited to the treatment of various types of immunodeficiencies that affect lymphocyte development and function. Other congenital defects such as the thalassemias have not resulted in clinically significant engraftment. Recent efforts at understanding and overcoming the barriers to engraftment in the fetus have focused on providing a selective advantage to donor stem cells and fostering immune tolerance toward the donor cells. The critical cellular components of the graft that promote engraftment and tolerance induction are being evaluated in animal models. Improvements in engraftment have resulted from the inclusion of T cells and/or dendritic cells in the graft, as well as a strategy of combined prenatal and postnatal transplantation. CONCLUSIONS The advantages, necessity, and benefits of early treatment will continue to encourage development of IUT as a means to treat hematopoietic and other types of birth defects.
Collapse
Affiliation(s)
- Marcus O Muench
- Department of Laboratory Medicine, University of California, San Francisco, CA 94143, USA.
| | | |
Collapse
|
14
|
In Utero Stem Cell Transplantation: Potential Therapeutic Application for Muscle Diseases. Stem Cells Int 2017; 2017:3027520. [PMID: 28596791 PMCID: PMC5450178 DOI: 10.1155/2017/3027520] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Accepted: 04/26/2017] [Indexed: 01/07/2023] Open
Abstract
Muscular dystrophies, myopathies, and traumatic muscle injury and loss encompass a large group of conditions that currently have no cure. Myoblast transplantations have been investigated as potential cures for these conditions for decades. However, current techniques lack the ability to generate cell numbers required to produce any therapeutic benefit. In utero stem cell transplantation into embryos has been studied for many years mainly in the context of hematopoietic cells and has shown to have experimental advantages and therapeutic applications. Moreover, patient-derived cells can be used for experimental transplantation into nonhuman animal embryos via in utero injection as the immune response is absent at such early stages of development. We therefore propose in utero transplantation as a potential method to generate patient-derived humanized skeletal muscle as well as muscle stem cells in animals for therapeutic purposes as well as patient-specific drug screening.
Collapse
|
15
|
Almeida-Porada G, Atala A, Porada CD. In utero stem cell transplantation and gene therapy: rationale, history, and recent advances toward clinical application. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2016; 5:16020. [PMID: 27069953 PMCID: PMC4813605 DOI: 10.1038/mtm.2016.20] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/17/2015] [Revised: 01/29/2016] [Accepted: 01/29/2016] [Indexed: 12/11/2022]
Abstract
Recent advances in high-throughput molecular testing have made it possible to diagnose most genetic disorders relatively early in gestation with minimal risk to the fetus. These advances should soon allow widespread prenatal screening for the majority of human genetic diseases, opening the door to the possibility of treatment/correction prior to birth. In addition to the obvious psychological and financial benefits of curing a disease in utero, and thereby enabling the birth of a healthy infant, there are multiple biological advantages unique to fetal development, which provide compelling rationale for performing potentially curative treatments, such as stem cell transplantation or gene therapy, prior to birth. Herein, we briefly review the fields of in utero transplantation (IUTx) and in utero gene therapy and discuss the biological hurdles that have thus far restricted success of IUTx to patients with immunodeficiencies. We then highlight several recent experimental breakthroughs in immunology, hematopoietic/marrow ontogeny, and in utero cell delivery, which have collectively provided means of overcoming these barriers, thus setting the stage for clinical application of these highly promising therapies in the near future.
Collapse
Affiliation(s)
- Graça Almeida-Porada
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine , Winston Salem, North Carolina, USA
| | - Anthony Atala
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine , Winston Salem, North Carolina, USA
| | - Christopher D Porada
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine , Winston Salem, North Carolina, USA
| |
Collapse
|
16
|
Kinder JM, Jiang TT, Ertelt JM, Xin L, Strong BS, Shaaban AF, Way SS. Tolerance to noninherited maternal antigens, reproductive microchimerism and regulatory T cell memory: 60 years after 'Evidence for actively acquired tolerance to Rh antigens'. CHIMERISM 2015; 6:8-20. [PMID: 26517600 PMCID: PMC5063085 DOI: 10.1080/19381956.2015.1107253] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Compulsory exposure to genetically foreign maternal tissue imprints in offspring sustained tolerance to noninherited maternal antigens (NIMA). Immunological tolerance to NIMA was first described by Dr. Ray D. Owen for women genetically negative for erythrocyte rhesus (Rh) antigen with reduced sensitization from developmental Rh exposure by their mothers. Extending this analysis to HLA haplotypes has uncovered the exciting potential for therapeutically exploiting NIMA-specific tolerance naturally engrained in mammalian reproduction for improved clinical outcomes after allogeneic transplantation. Herein, we summarize emerging scientific concepts stemming from tolerance to NIMA that includes postnatal maintenance of microchimeric maternal origin cells in offspring, expanded accumulation of immune suppressive regulatory T cells with NIMA-specificity, along with teleological benefits and immunological consequences of NIMA-specific tolerance conserved across mammalian species.
Collapse
Affiliation(s)
- Jeremy M Kinder
- a Division of Infectious Diseases and Perinatal Institute, Cincinnati Children's Hospital, Cincinnati , OH , USA
| | - Tony T Jiang
- a Division of Infectious Diseases and Perinatal Institute, Cincinnati Children's Hospital, Cincinnati , OH , USA
| | - James M Ertelt
- a Division of Infectious Diseases and Perinatal Institute, Cincinnati Children's Hospital, Cincinnati , OH , USA
| | - Lijun Xin
- a Division of Infectious Diseases and Perinatal Institute, Cincinnati Children's Hospital, Cincinnati , OH , USA
| | - Beverly S Strong
- b Center for Fetal Cellular and Molecular Therapy, Cincinnati Children's Hospital , Cincinnati , OH , USA
| | - Aimen F Shaaban
- b Center for Fetal Cellular and Molecular Therapy, Cincinnati Children's Hospital , Cincinnati , OH , USA
| | - Sing Sing Way
- a Division of Infectious Diseases and Perinatal Institute, Cincinnati Children's Hospital, Cincinnati , OH , USA
| |
Collapse
|
17
|
Strong BSI, Ryken KO, Lee AE, Turner LE, Wadhwani RK, Newkold TJ, Alhajjat AM, Heusel JW, Shaaban AF. Prenatal Allogeneic Tolerance in Mice Remains Stable Despite Potent Viral Immune Activation. THE JOURNAL OF IMMUNOLOGY 2015; 195:4001-9. [PMID: 26363051 DOI: 10.4049/jimmunol.1500844] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/13/2015] [Accepted: 08/11/2015] [Indexed: 12/11/2022]
Abstract
Transplanting stem cells before birth offers an unparalleled opportunity to initiate corrective treatment for numerous childhood diseases with minimal or no host conditioning. Although long-term engraftment has been demonstrated following in utero hematopoietic cellular transplantation during immune quiescence, it is unclear if prenatal tolerance becomes unstable with immune activation such as during a viral syndrome. Using a murine model of in utero hematopoietic cellular transplantation, the impact of an infection with lymphocytic choriomeningitis virus on prenatal allospecific tolerance was examined. The findings in this report illustrate that established mechanisms of donor-specific tolerance are strained during potent immune activation. Specifically, a transient reversal in the anergy of alloreactive lymphocytes is seen in parallel with the global immune response toward the virus. However, these changes return to baseline following resolution of the infection. Importantly, prenatal engraftment remains stable during and after immune activation. Collectively, these findings illustrate the robust nature of allospecific tolerance in prenatal mixed chimerism compared with models of postnatal chimerism and provides additional support for the prenatal approach to the treatment of congenital benign cellular disease.
Collapse
Affiliation(s)
- Beverly S I Strong
- Department of Surgery, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229; University of Cincinnati College of Medicine, Cincinnati, OH 45229
| | - Katherine O Ryken
- Department of Surgery, University of Iowa Carver College of Medicine, Iowa City, IA 52242
| | - Amanda E Lee
- Department of Surgery, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229; University of Cincinnati College of Medicine, Cincinnati, OH 45229
| | - Lucas E Turner
- Department of Surgery, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229; University of Cincinnati College of Medicine, Cincinnati, OH 45229
| | - Ram K Wadhwani
- Department of Surgery, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229; University of Cincinnati College of Medicine, Cincinnati, OH 45229
| | - Tess J Newkold
- Department of Surgery, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229; University of Cincinnati College of Medicine, Cincinnati, OH 45229
| | - Amir M Alhajjat
- Department of Surgery, University of Iowa Carver College of Medicine, Iowa City, IA 52242
| | - Jonathan W Heusel
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110; and Department of Genetics, Washington University School of Medicine, St. Louis, MO 63110
| | - Aimen F Shaaban
- Department of Surgery, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229; University of Cincinnati College of Medicine, Cincinnati, OH 45229;
| |
Collapse
|
18
|
McClain LE, Flake AW. In utero stem cell transplantation and gene therapy: Recent progress and the potential for clinical application. Best Pract Res Clin Obstet Gynaecol 2015; 31:88-98. [PMID: 26483174 DOI: 10.1016/j.bpobgyn.2015.08.006] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2015] [Accepted: 08/31/2015] [Indexed: 12/20/2022]
Abstract
Advances in prenatal diagnosis have led to the prenatal management and treatment of a variety of congenital diseases. Although surgical treatment has been successfully applied to specific anatomic defects that place the fetus at a risk of death or life-long disability, the indications for fetal surgical intervention have remained relatively limited. By contrast, prenatal stem cell and gene therapy await clinical application, but they have tremendous potential to treat a broad range of genetic disorders. If there are biological advantages unique to fetal development that favor fetal stem cell or gene therapy over postnatal treatment, prenatal therapy may become the preferred approach to the treatment of any disease that can be prenatally diagnosed and cured by stem cell or gene therapy. Here, we review the field including recent progress toward clinical application and imminent clinical trials for cellular and gene therapy.
Collapse
Affiliation(s)
- Lauren E McClain
- Children's Center for Fetal Research, Children's Hospital of Philadelphia, University of Pennsylvania School of Medicine, Philadelphia, PA, USA
| | - Alan W Flake
- Children's Center for Fetal Research, Children's Hospital of Philadelphia, University of Pennsylvania School of Medicine, Philadelphia, PA, USA.
| |
Collapse
|
19
|
Correction of murine hemoglobinopathies by prenatal tolerance induction and postnatal nonmyeloablative allogeneic BM transplants. Blood 2015; 126:1245-54. [PMID: 26124498 DOI: 10.1182/blood-2015-03-636803] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2015] [Accepted: 06/19/2015] [Indexed: 12/16/2022] Open
Abstract
Sickle cell disease (SCD) and thalassemias (Thal) are common congenital disorders, which can be diagnosed early in gestation and result in significant morbidity and mortality. Hematopoietic stem cell transplantation, the only curative therapy for SCD and Thal, is limited by the absence of matched donors and treatment-related toxicities. In utero hematopoietic stem cell transplantation (IUHCT) is a novel nonmyeloablative transplant approach that takes advantage of the immunologic immaturity and normal developmental properties of the fetus to achieve mixed allogeneic chimerism and donor-specific tolerance (DST). We hypothesized that a combined strategy of IUHCT to induce DST, followed by postnatal nonmyeloablative same donor "booster" bone marrow (BM) transplants in murine models of SCD and Thal would result in high levels of allogeneic engraftment and donor hemoglobin (Hb) expression with subsequent phenotypic correction of SCD and Thal. Our results show that: (1) IUHCT is associated with DST and low levels of allogeneic engraftment in the murine SCD and Thal models; (2) low-level chimerism following IUHCT can be enhanced to high-level chimerism and near complete Hb replacement with normal donor Hb with this postnatal "boosting" strategy; and (3) high-level chimerism following IUHCT and postnatal "boosting" results in phenotypic correction in the murine Thal and SCD models. This study supports the potential of IUHCT, combined with a postnatal nonmyelablative "boosting" strategy, to cure Thal and SCD without the toxic conditioning currently required for postnatal transplant regimens while expanding the eligible transplant patient population due to the lack of a restricted donor pool.
Collapse
|
20
|
Alhajjat AM, Lee AE, Strong BS, Shaaban AF. NK cell tolerance as the final endorsement of prenatal tolerance after in utero hematopoietic cellular transplantation. Front Pharmacol 2015; 6:51. [PMID: 25852555 PMCID: PMC4364176 DOI: 10.3389/fphar.2015.00051] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2014] [Accepted: 03/02/2015] [Indexed: 01/19/2023] Open
Abstract
The primary benefits of in utero hematopoietic cellular transplantation (IUHCT) arise from transplanting curative cells prior to the immunologic maturation of the fetus. However, this approach has been routinely successful only in the treatment of congenital immunodeficiency diseases that include an inherent NK cell deficiency despite the existence of normal maternal immunity in either setting. These observations raise the possibility that fetal NK cells function as an early barrier to allogeneic IUHCT. Herein, we summarize the findings of previous studies of prenatal NK cell allospecific tolerance in mice and in humans. Cumulatively, this new information reveals the complexity of the fetal immune response in the setting of rejection or tolerance and illustrates the role for fetal NK cells in the final endorsement of allospecific prenatal tolerance.
Collapse
Affiliation(s)
- Amir M Alhajjat
- Department of Surgery, University of Iowa, Iowa City, IA USA
| | - Amanda E Lee
- Center for Fetal Cellular and Molecular Therapy and The Department of Surgery, Cincinnati Children's Hospital Medical Center, Cincinnati, OH USA
| | - Beverly S Strong
- Center for Fetal Cellular and Molecular Therapy and The Department of Surgery, Cincinnati Children's Hospital Medical Center, Cincinnati, OH USA
| | - Aimen F Shaaban
- Center for Fetal Cellular and Molecular Therapy and The Department of Surgery, Cincinnati Children's Hospital Medical Center, Cincinnati, OH USA
| |
Collapse
|
21
|
Loewendorf AI, Csete M, Flake A. Immunological considerations in in utero hematopoetic stem cell transplantation (IUHCT). Front Pharmacol 2015; 5:282. [PMID: 25610396 PMCID: PMC4285014 DOI: 10.3389/fphar.2014.00282] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2014] [Accepted: 12/02/2014] [Indexed: 01/19/2023] Open
Abstract
In utero hematopoietic stem cell transplantation (IUHCT) is an attractive approach and a potentially curative surgery for several congenital hematopoietic diseases. In practice, this application has succeeded only in the context of Severe Combined Immunodeficiency Disorders. Here, we review potential immunological hurdles for the long-term establishment of chimerism and discuss relevant models and findings from both postnatal hematopoietic stem cell transplantation and IUHCT.
Collapse
Affiliation(s)
- Andrea I Loewendorf
- Department of Obstetrics and Gynecology, David Geffen School of Medicine, University of California, Los Angeles Los Angeles, CA, USA
| | - Marie Csete
- Chief Scientific Officer, The Huntington Medical Research Institutes Pasadena, CA, USA
| | - Alan Flake
- The Children's Hospital of Philadelphia, Children's Institute of Surgical Science Philadelphia, PA, USA
| |
Collapse
|
22
|
Peranteau WH. In utero hematopoietic cell transplantation: induction of donor specific immune tolerance and postnatal transplants. Front Pharmacol 2014; 5:251. [PMID: 25429269 PMCID: PMC4228834 DOI: 10.3389/fphar.2014.00251] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2014] [Accepted: 10/28/2014] [Indexed: 11/20/2022] Open
Abstract
In utero hematopoietic cell transplantation (IUHCT) is a non-myeloablative non-immunosuppressive transplant approach that allows for donor cell engraftment across immunologic barriers. Successful engraftment is associated with donor-specific tolerance. IUHCT has the potential to treat a large number of congenital hematologic, immunologic, and genetic diseases either by achieving high enough engraftment levels following a single IUHCT or by inducing donor specific tolerance to allow for non-toxic same-donor postnatal transplants. This review evaluates donor specific tolerance induction achieved by IUHCT. Specifically it addresses the need to achieve threshold levels of donor cell engraftment following IUHCT to consistently obtain immunologic tolerance. The mechanisms of tolerance induction including partial deletion of donor reactive host T cells by direct and indirect antigen presentation and the role of regulatory T cells in maintaining tolerance are reviewed. Finally, this review highlights the promising clinical potential of in utero tolerance induction to provide a platform on which postnatal cellular and organ transplants can be performed without myeloablative or immunosuppressive conditioning.
Collapse
Affiliation(s)
- William H Peranteau
- Department of Surgery, Center for Fetal Research, The Children's Hospital of Philadelphia Philadelphia, PA, USA
| |
Collapse
|
23
|
In utero depletion of fetal hematopoietic stem cells improves engraftment after neonatal transplantation in mice. Blood 2014; 124:973-80. [PMID: 24879814 DOI: 10.1182/blood-2014-02-550327] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Although in utero hematopoietic cell transplantation is a promising strategy to treat congenital hematopoietic disorders, levels of engraftment have not been therapeutic for diseases in which donor cells have no survival advantage. We used an antibody against the murine c-Kit receptor (ACK2) to deplete fetal host hematopoietic stem cells (HSCs) and increase space within the hematopoietic niche for donor cell engraftment. Fetal mice were injected with ACK2 on embryonic days 13.5 to 14.5 and surviving pups were transplanted with congenic hematopoietic cells on day of life 1. Low-dose ACK2 treatment effectively depleted HSCs within the bone marrow with minimal toxicity and the antibody was cleared from the serum before the neonatal transplantation. Chimerism levels were significantly higher in treated pups than in controls; both myeloid and lymphoid cell chimerism increased because of higher engraftment of HSCs in the bone marrow. To test the strategy of repeated HSC depletion and transplantation, some mice were treated with ACK2 postnatally, but the increase in engraftment was lower than that seen with prenatal treatment. We demonstrate a successful fetal conditioning strategy associated with minimal toxicity. Such strategies could be used to achieve clinically relevant levels of engraftment to treat congenital stem cell disorders.
Collapse
|
24
|
Pixley JS, Zanjani ED. In utero transplantation: Disparate ramifications. World J Stem Cells 2013; 5:43-52. [PMID: 23671718 PMCID: PMC3648645 DOI: 10.4252/wjsc.v5.i2.43] [Citation(s) in RCA: 5] [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: 12/15/2012] [Revised: 01/07/2013] [Accepted: 01/24/2013] [Indexed: 02/06/2023] Open
Abstract
In utero stem cell transplantation, which promises treatment for a host of genetic disorders early in gestation before disease effect stems from Ray Owen’s seminal observation that self-tolerance, is acquired during gestation. To date, in utero transplantation (IUT) has proved useful in characterizing the hematopoietic stem cell. Recent observations support its use as an in vivo method to further understanding of self-tolerance. Preclinical development continues for its application as a treatment for childhood hematolymphoid diseases. In addition, IUT may offer therapeutic options in the treatment of diabetes among other diseases. Thus IUT serves as a technique or system important in both a basic and applied format. This review summarizes these findings.
Collapse
|
25
|
Direct and indirect antigen presentation lead to deletion of donor-specific T cells after in utero hematopoietic cell transplantation in mice. Blood 2013; 121:4595-602. [PMID: 23610372 DOI: 10.1182/blood-2012-10-463174] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
In utero hematopoietic cell transplantation (IUHCTx) is a promising method to induce donor-specific tolerance but the mechanisms of antigen presentation that educate host T cells and the relative importance of deletion vs regulation in this setting are unknown. We studied the roles of direct and indirect antigen presentation (mediated by donor- and host-derived antigen-presenting cells [APCs], respectively) in a mouse model of IUHCTx. We found that IUHCTx leads to precocious maturation of neonatal host dendritic cells (DCs) and that there is early differentiation of donor-derived DCs, even after transplantation of a stem cell source without mature APCs. We next performed allogeneic IUHCTx into donor-specific T-cell receptor transgenic mice and confirmed that both direct and indirect antigen presentation lead to clonal deletion of effector T cells in chimeras. Deletion did not persist when chimerism was lost. Importantly, although the percentage of regulatory T cells (Tregs) after IUHCTx increased, there was no expansion in Treg numbers. In wild-type mice, there was a similar deletion of effector cells without expansion of donor-specific Tregs. Thus, tolerance induction after IUHCTx depends on both direct and indirect antigen presentation and is secondary to thymic deletion, without de novo Treg induction.
Collapse
|
26
|
Vrecenak JD, Flake AW. In utero hematopoietic cell transplantation--recent progress and the potential for clinical application. Cytotherapy 2013; 15:525-35. [PMID: 23415921 DOI: 10.1016/j.jcyt.2013.01.003] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2012] [Accepted: 01/08/2013] [Indexed: 11/27/2022]
Abstract
In utero hematopoietic stem cell transplantation (IUHCT) is a potential therapeutic alternative to postnatal hematopoietic stem cell transplantation (HSCT) for congenital hematologic disorders that can be diagnosed early in gestation and can be cured by HSCT. The rationale is to take advantage of normal events during hematopoietic and immunologic ontogeny to facilitate allogeneic hematopoietic engraftment. Although the rationale remains compelling, IUHCT has not yet achieved its clinical potential. This review will discuss recent experimental progress toward overcoming the barriers to allogeneic engraftment and new therapeutic strategies that may hasten clinical application.
Collapse
Affiliation(s)
- Jesse D Vrecenak
- Children's Center for Fetal Research, Children's Hospital of Philadelphia, and Department of Surgery, University of Pennsylvania, School of Medicine, Philadelphia, PA, USA
| | | |
Collapse
|
27
|
Donor chimera model for tolerance induction in transplantation. Hum Immunol 2013; 74:550-6. [PMID: 23354322 DOI: 10.1016/j.humimm.2013.01.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2012] [Revised: 12/09/2012] [Accepted: 01/14/2013] [Indexed: 12/14/2022]
Abstract
Tolerance induction is the basis of a successful transplantation with the goal being the re-establishment of homeostasis after transplantation. Non-autograft transplantation disrupts this maintenance drastically which would be avoided by administration of a novel procedure. At present, the blood group antigens and the genotypes of the donor and recipient are cross-matched before transplantation combined with a drug regimen that confers general immunosuppression. But the 'specific' unresponsiveness of the recipient to the donor organ, implied by 'tolerance', is not achieved in this process. This article introduces the 'donor chimera model' via the concept of the 'closed transplantation loop' approach for tolerance induction which seeks to limit the use of immunosuppressive therapy after transplantation.
Collapse
|
28
|
Nijagal A, Flake AW, MacKenzie TC. In utero hematopoietic cell transplantation for the treatment of congenital anomalies. Clin Perinatol 2012; 39:301-10. [PMID: 22682381 DOI: 10.1016/j.clp.2012.04.004] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
In utero hematopoietic cell transplantation (IUHCTx) is a promising strategy for the treatment of common hematopoietic disorders and for inducing immune tolerance in the fetus. Although the efficacy of IUHCTx has been demonstrated in multiple small and large animal models, the clinical application of this technique in humans has had limited success. Recent studies in mice have demonstrated that the maternal immune system plays a critical role in limiting engraftment in the fetus. This article reviews the therapeutic rationale of IUHCTx, potential barriers to its applications, and recent experimental strategies to improve its clinical success.
Collapse
Affiliation(s)
- Amar Nijagal
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, Department of Surgery, 513 Parnassus Avenue, San Francisco, CA 94143-0570, USA
| | | | | |
Collapse
|
29
|
Migration of cells from the yolk sac to hematopoietic tissues after in utero transplantation of early and mid gestation canine fetuses. Transplantation 2011; 91:723-30. [PMID: 21325997 DOI: 10.1097/tp.0b013e31820c85bc] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
BACKGROUND In utero hematopoietic cell transplantation offers a means of early intervention for the treatment of diseases before birth. Delivery of cells to the yolk sac is a minimally invasive approach that results in low levels of chimerism. However, there is little information on the optimal doses, timing of delivery, and migration of transplanted cells from the yolk sac into the fetus. METHODS Varying cell doses of mesenchymal stromal cells or bone marrow mononuclear cells labeled with fluorescent supraparamagnetic iron oxide nanoparticles and a fluorescent intracellular dye, 5- and 6-([(4-chloromethyl)benzoyl]-amino) tetramethylrhodamine, were transplanted under ultrasound guidance to the yolk sacs of day 25 or day 35 canine fetuses. Ex vivo whole body fluorescence imaging and microscopy of tissue sections were correlated with the presence of iron oxide in injected and control fetuses. RESULTS Day 25 and day 35 recipients showed similar survival rates after injection of cells into yolk sacs, although increased fetal morality was associated with cell doses greater than 10 cells/kg to day 25 fetuses. The fluorescence and iron oxide signals were predominantly localized to the abdominal regions, with no fluorescence visible in yolk sacs. Microscopy of tissues revealed colocalization of fluorophore with iron oxide in donor cells detected in the fetal livers and bone marrow of recipients 7 and 17 days after receiving mesenchymal stromal cells or bone marrow mononuclear cells. CONCLUSIONS These studies demonstrated that cells injected into the yolk sacs of early gestation canine fetuses migrate to recipient hematopoietic tissues. Thus, yolk sac injection offers a safe and effective approach for engraftment of cells to fetal hematopoietic tissues.
Collapse
|
30
|
Nijagal A, Wegorzewska M, Jarvis E, Le T, Tang Q, MacKenzie TC. Maternal T cells limit engraftment after in utero hematopoietic cell transplantation in mice. J Clin Invest 2011; 121:582-92. [PMID: 21245575 DOI: 10.1172/jci44907] [Citation(s) in RCA: 114] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2010] [Accepted: 11/23/2010] [Indexed: 12/31/2022] Open
Abstract
Transplantation of allogeneic stem cells into the early gestational fetus, a treatment termed in utero hematopoietic cell transplantation (IUHCTx), could potentially overcome the limitations of bone marrow transplants, including graft rejection and the chronic immunosuppression required to prevent rejection. However, clinical use of IUHCTx has been hampered by poor engraftment, possibly due to a host immune response against the graft. Since the fetal immune system is relatively immature, we hypothesized that maternal cells trafficking into the fetus may pose the true barrier to effective IUHCTx. Here, we have demonstrated that there is macrochimerism of maternal leukocytes in the blood of unmanipulated mouse fetuses, with substantial increases in T cell trafficking after IUHCTx. To determine the contribution of these maternal lymphocytes to rejection after IUHCTx, we bred T and/or B cell-deficient mothers to wild-type fathers and performed allogeneic IUHCTx into the immunocompetent fetuses. There was a marked improvement in engraftment if the mother lacked T cells but not B cells, indicating that maternal T cells are the main barrier to engraftment. Furthermore, when the graft was matched to the mother, there was no difference in engraftment between syngeneic and allogeneic fetal recipients. Our study suggests that the clinical success of IUHCTx may be improved by transplanting cells matched to the mother.
Collapse
Affiliation(s)
- Amar Nijagal
- Eli and Edythe Broad Center of Regeneration Medicine, San Francisco, California, USA
| | | | | | | | | | | |
Collapse
|
31
|
Merianos DJ, Tiblad E, Santore MT, Todorow CA, Laje P, Endo M, Zoltick PW, Flake AW. Maternal alloantibodies induce a postnatal immune response that limits engraftment following in utero hematopoietic cell transplantation in mice. J Clin Invest 2009; 119:2590-600. [PMID: 19652363 DOI: 10.1172/jci38979] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2009] [Accepted: 06/03/2009] [Indexed: 12/14/2022] Open
Abstract
The lack of fetal immune responses to foreign antigens, i.e., fetal immunologic tolerance, is the most compelling rationale for prenatal stem cell and gene therapy. However, the frequency of engraftment following in utero hematopoietic cell transplantation (IUHCT) in the murine model is reduced in allogeneic, compared with congenic, recipients. This observation supports the existence of an immune barrier to fetal transplantation and challenges the classic assumptions of fetal tolerance. Here, we present evidence that supports the presence of an adaptive immune response in murine recipients of IUHCT that failed to maintain engraftment. However, when IUHCT recipients were fostered by surrogate mothers, they all maintained long-term chimerism. Furthermore, we have demonstrated that the cells responsible for rejection of the graft were recipient in origin. Our observations suggest a mechanism by which IUHCT-dependent sensitization of the maternal immune system and the subsequent transmission of maternal alloantibodies to pups through breast milk induces a postnatal adaptive immune response in the recipient, which, in turn, results in the ablation of engraftment after IUHCT. Finally, we showed that non-fostered pups that maintained their chimerism had higher levels of Tregs as well as a more suppressive Treg phenotype than their non-chimeric, non-fostered siblings. This study resolves the apparent contradiction of induction of an adaptive immune response in the pre-immune fetus and confirms the potential of actively acquired tolerance to facilitate prenatal therapeutic applications.
Collapse
Affiliation(s)
- Demetri J Merianos
- Children's Center for Fetal Research, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | | | | | | | | | | | | | | |
Collapse
|
32
|
Peranteau WH, Heaton TE, Gu YC, Volk SW, Bauer TR, Alcorn K, Tuschong LM, Johnson MP, Hickstein DD, Flake AW. Haploidentical in utero hematopoietic cell transplantation improves phenotype and can induce tolerance for postnatal same-donor transplants in the canine leukocyte adhesion deficiency model. Biol Blood Marrow Transplant 2009; 15:293-305. [PMID: 19203720 DOI: 10.1016/j.bbmt.2008.11.034] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2008] [Accepted: 11/25/2008] [Indexed: 11/19/2022]
Abstract
In the murine model, in utero hematopoietic cell transplantation (IUHCT) has been shown to achieve low levels of allogeneic chimerism and associated donor-specific tolerance permitting minimal conditioning postnatal hematopoietic stem cell transplantation (HSCT). In this pilot study, we investigated IUHCT in the canine leukocyte adhesion deficiency (CLAD) model. Haploidentical IUHCT resulted in stable low-level donor cell chimerism in all dogs that could be analyzed by sensitive detection methodology (4 of 10) through 18 months of follow-up. In the 2 CLAD recipients, low-level chimerism resulted in amelioration and complete reversal of the CLAD phenotype, respectively. Six recipients of IUHCT (5 carriers and 1 CLAD) subsequently received postnatal HSCT from the same haploidentical prenatal donor after minimal conditioning with busulfan 10 mg/kg. Chimerism in 2 of 5 CLAD carriers that underwent HSCT increased from < 1% pre-HSCT to sustained levels of 35% to 45%. Control animals undergoing postnatal haploidentical HSCT without IUHCT had no detectable donor chimerism. These results demonstrate that haploidentical IUHCT in the CLAD model can result in low-level donor chimerism that can prevent the lethal phenotype in CLAD dogs, and can result in donor-specific tolerance that can facilitate postnatal minimal conditioning HSCT.
Collapse
Affiliation(s)
- William H Peranteau
- Center for Fetal Research, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Abstract
At the present time, the most likely and eminent application of stem cell therapy to the fetus is in utero hematopoietic stem cell transplantation (IUHCT), and this stem cell type will be discussed as a paradigm for all prenatal stem cell therapy. The authors feel that the most likely initial application of IUHCT will use adult HSC derived from bone marrow (BM) or peripheral blood (PB), and will focus this article on this specific approach. The article also reviews the experimental data that support the capacity of IUHCT to induce donor-specific tolerance.
Collapse
|
34
|
Persisting NK cell alloreactivity in the presence of long-term stable hematopoietic chimerism. Exp Hematol 2009; 37:739-43. [DOI: 10.1016/j.exphem.2009.02.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2008] [Revised: 02/16/2009] [Accepted: 02/17/2009] [Indexed: 11/18/2022]
|
35
|
Skopal-Chase JL, Pixley JS, Torabi A, Cenariu MC, Bhat A, Thain DS, Frederick NM, Groza DM, Zanjani ED. Immune ontogeny and engraftment receptivity in the sheep fetus. Fetal Diagn Ther 2009; 25:102-10. [PMID: 19246928 DOI: 10.1159/000203399] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2007] [Accepted: 05/30/2008] [Indexed: 12/17/2022]
Abstract
OBJECTIVE The biologic explanation for fetal receptivity to donor engraftment and subsequent long-term tolerance following transplantation early in gestation is not known. We investigated the role fetal immune ontogeny might play in fetal transplantation tolerance in sheep. METHODS Engraftment of allogeneic and xenogeneic HSC was determined 60 days following transplantation at different time points in sheep fetal gestation. Parallel analysis of surface differentiation antigen expression on cells from lymphoid organs of timed gestational age fetal sheep was determined by flow cytometry using available reagents. RESULTS An engraftment window was identified after day 52 gestation lasting until day 71 (term gestation: 145 days). This period was associated with the expression of the leukocyte common antigen CD45 on all cells in the thymus. Double-positive and single-positive CD4 and CD8 cells began appearing in the thymus just prior (day 45 gestation) to the beginning of the engraftment window, while single-positive CD4 or CD8 cells do not begin appearing in peripheral organs until late in the engraftment period, suggesting deletional mechanisms may be operative. In concert, surface IgM-positive cells express CD45 in the thymus at day 45, with a comparable delay in the appearance of IgM/CD45 cells in the periphery until late in the engraftment window. CONCLUSIONS These findings support a central role for the thymus in multilineage immune cell maturation during the period of fetal transplantation receptivity. Further, they suggest that fetal engraftment receptivity is due to gestational age-dependent deletional tolerance.
Collapse
|
36
|
Chen JC, Chang ML, Huang SF, Chang PY, Muench MO, Fu RH, Ou LS, Kuo ML. Prenatal tolerance induction: relationship between cell dose, marrow T-cells, chimerism, and tolerance. Cell Transplant 2009; 17:495-506. [PMID: 18714669 DOI: 10.3727/096368908785095971] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
It was reported that the dose of self-antigens can determine the consequence of deletional tolerance and donor T cells are critical for tolerance induction in mixed chimeras. This study aimed at assessing the effect of cell doses and marrow T cells on engraftment and tolerance induction after prenatal bone marrow transplantation. Intraperitoneal cell transplantation was performed in FVB/N (H-2K(q)) mice at gestational day 14 with escalating doses of adult C57BL/6 (H-2K(b)) marrows. Peripheral chimerism was examined postnatally by flow cytometry and tolerance was tested by skin transplantation. Transplantation of light-density marrow cells showed a dose response. High-level chimerism emerged with a threshold dose of 5.0 x 10(6) and host leukocytes could be nearly replaced at a dose of 7.5-10.0 x 10(6). High-dose transplants conferred a steady long-lasting donor-specific tolerance but were accompanied by >50% incidence of graft-versus-host disease. Depletion of marrow T cells lessened graft-versus-host disease to the detriment of engraftment. With low-level chimerism, tolerance was a graded phenomenon dependent upon the level of chimerism. Durable chimerism within 6 months required a threshold of > or = 2% chimerism at 1 month of age and predicted a 50% chance of long-term tolerance, whereas transient chimerism (<2%) only caused hyporesponsiveness to the donor. Tolerance induction did not succeed without peripheral chimerism even if a large amount of injected donor cells persisted in the peritoneum. Neither did an increase in cell doses or donor T-cell contents benefit skin graft survivals unless it had substantially improved peripheral chimerism. Thus, peripheral chimerism level can be a simple and straightforward test to predict the degree of prenatal immune tolerance.
Collapse
Affiliation(s)
- Jeng-Chang Chen
- Department of Surgery, Chang Gung Children's Hospital, Chang Gung University College of Medicine, Taoyuan, Taiwan.
| | | | | | | | | | | | | | | |
Collapse
|
37
|
Early chimerism threshold predicts sustained engraftment and NK-cell tolerance in prenatal allogeneic chimeras. Blood 2008; 112:5245-53. [PMID: 18796629 DOI: 10.1182/blood-2007-12-128116] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The failure of engraftment in human cases of in utero hematopoietic cell transplantation (IUHCT) in which no immunodeficiency exists suggests the presence of an unrecognized fetal immune barrier. A similar barrier in murine IUHCT appears to be dependent on the chimerism level and is poorly explained by a lack of T-cell tolerance induction. Therefore, we studied the effect of the chimerism level on engraftment and host natural killer (NK)-cell education in a murine model of IUHCT. The dose of transplanted cells was found to exhibit a strong correlation with both the engraftment rate and chimerism level. More specifically, a threshold level of initial chimerism (> 1.8%) was identified that predicted durable engraftment for allogeneic IUHCT, whereas low initial chimerism (< 1.8%) predicted a loss of engraftment. NK cells taken from chimeras above the "chimerism threshold" displayed durable calibration of alloresponsive Ly49A receptors and tolerance to donor antigens. Depletion of recipient NK cells stabilized engraftment in low-level chimeras (< 1.8%). These studies illustrate the importance of the early chimerism threshold in predicting long-term engraftment and host NK-cell tolerance after in utero transplantation.
Collapse
|
38
|
In Utero Hematopoietic Stem Cell Transplantation: Progress toward Clinical Application. Biol Blood Marrow Transplant 2008; 14:729-40. [DOI: 10.1016/j.bbmt.2008.02.012] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2008] [Accepted: 02/15/2008] [Indexed: 11/22/2022]
|
39
|
Durkin ET, Jones KA, Elnaggar D, Shaaban AF. Donor major histocompatibility complex class I expression determines the outcome of prenatal transplantation. J Pediatr Surg 2008; 43:1142-7. [PMID: 18558197 PMCID: PMC2587093 DOI: 10.1016/j.jpedsurg.2008.02.046] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2008] [Accepted: 02/09/2008] [Indexed: 10/21/2022]
Abstract
PURPOSE The failure of in utero transplantation in immune-competent recipients suggests the existence of a fetal immune barrier. The importance of donor major histocompatibility complex (MHC) class I expression in the induction of prenatal tolerance remains undefined. We hypothesized that donor cell MHC class I expression facilitates engraftment in prenatal allogeneic recipients rather than promoting immune rejection. METHODS B6.Ly5.2 (class I(+)) or B6.TAP(-/-) (class I(-)) murine fetal liver cells were transplanted into age-matched allogeneic fetal recipients. Survival to weaning and subsequent growth was assessed. Engraftment rates and peripheral blood chimerism levels were measured serially. RESULTS The presence or absence of class I expression did not affect survival or growth of recipients and no graft-vs-host disease developed. Allogeneic recipients of B6.Ly5.2 cells exhibited significantly higher levels of donor hematopoietic chimerism when compared to recipients of B6.TAP(-/-) cells (27% + 10% vs 11% + 8%; P = .004) that deteriorated further over time. CONCLUSIONS Donor class I MHC antigen expression is essential for stable long-term engraftment and maintenance of donor-specific tolerance. Further studies are needed to better characterize the role of the fetal innate immune system in prenatal allotransplantation.
Collapse
|
40
|
Lin H, Mao Q, Wang YM, Jiang L. Proliferation of L02 human hepatocytes in tolerized genetically immunocompetent rats. World J Gastroenterol 2008; 14:2329-37. [PMID: 18416458 PMCID: PMC2705086 DOI: 10.3748/wjg.14.2329] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate whether human hepatocytes could proliferate after transplantation to normal immunocompetent rats treated with 2-acetaminofluorene or Retrorsine and partial hepatectomy.
METHODS: L02 hepatocyte-tolerant Sprague-Dawley rats were injected with Retrorsine, 2-acetaminofluorene or normal saline. L02 hepatocytes were then transplanted via the spleen. Human albumin and its mRNA, specific proliferating cell nuclear antigen (PCNA), L02 hepatocyte dynamic distribution, number density and area density of PCNA-positive cells in the liver were determined.
RESULTS: All the examined indicators were not significantly different between the rats treated with 2-acetaminofluorene and normal saline, which was not the case with rats treated with Retrorsine. A dynamic distribution of L02 hepatocytes in the rat liver was detected from wk 1 to mo 6 after transplantation in the Retrorsine group and from wk 1 to 10 in the 2-acetaminofluorene group. Human albumin and its mRNA were detected from wk 2 to mo 6 in the Retrorsine group and from wk 1 to 8 in the 2-acetaminofluorene group. Specific human PCNA was detected in the rat liver from wk 2 to mo 6 in the Retrorsine group and from wk 2 to 6 in the 2-acetaminofluorene group. Human albumin and its mRNA contents as well as the number of PCNA positive cells reached a peak at wk 4.
CONCLUSION: L02 human hepatocytes could not proliferate significiantly after transplantation to the normal, immunocompetent rats treated with 2-acetaminofluorene. L02 human hepatocytes can survive for 10 wk after transplantation and express human albumin for 8 wk. L02 human hepatocytes can proliferate and express human albumin for 6 mo after transplantation to the rats treated with Retrorsine. The chimeric L02 human hepatocytes, which then underwent transplantation into tolerant rats, were normal in morphogenesis, biochemistry and function.
Collapse
|
41
|
Shaaban AF, Kim HB, Gaur L, Liechty KW, Flake AW. Prenatal transplantation of cytokine-stimulated marrow improves early chimerism in a resistant strain combination but results in poor long-term engraftment. Exp Hematol 2006; 34:1278-87. [PMID: 16939821 PMCID: PMC3096442 DOI: 10.1016/j.exphem.2006.05.007] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2006] [Revised: 05/08/2006] [Accepted: 05/08/2006] [Indexed: 01/17/2023]
Abstract
OBJECTIVE In the absence of immunodeficiency, only microchimerism (<0.1%) has been achieved in human fetal recipients or nonhuman primates following in utero hematopoietic cell transplantation (IUHCT). We hypothesized that enhanced long-term engraftment might be more reliably achieved in microchimeric systems if higher levels of chimerism existed during development of adaptive immunity. To evaluate this hypothesis, we stimulated the donor cells with vascular endothelial growth factor (VEGF) and stem cell factor (SCF) prior to IUHCT in a chimerism-resistant murine strain combination. METHODS Donor Balb/c marrow was cultured in media with or without VEGF and SCF supplementation for 12 hours prior to IUHCT into B6 fetuses at 14 days postcoitum (dpc). Donor cell phenotype, homing, and chimerism were assessed at short and long-term time points and transplanted animals received skin allografts at 8 weeks. RESULTS In pretreated allogeneic recipients, early chimerism rates were more than double that of controls (71% vs 33%, p = 0.01). These differences were associated with higher numbers of pretransplant donor cell colony-forming cells without change in donor cell homing. Despite prolonged skin allograft survival for pretreated recipients compared with controls (mean survival = 20.8 vs 8.2 days, p < 0.001), long-term engraftment was unchanged. CONCLUSIONS These findings demonstrate that higher levels of early chimerism in recipients of cytokine-stimulated marrow result in improved short-term chimerism and tolerance. Future studies are needed to confirm the existence of a "threshold" level of chimerism necessary to sustain long-term engraftment.
Collapse
Affiliation(s)
- Aimen F Shaaban
- Laboratory for Fetal Cellular Therapy, Department of Surgery, University of Wisconsin Medical School, Madison, WI 53792-7375, USA.
| | | | | | | | | |
Collapse
|
42
|
Peranteau WH, Endo M, Adibe OO, Flake AW. Evidence for an immune barrier after in utero hematopoietic-cell transplantation. Blood 2006; 109:1331-3. [PMID: 17023584 PMCID: PMC1785153 DOI: 10.1182/blood-2006-04-018606] [Citation(s) in RCA: 78] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The competence of the immune system of the developing fetus to act as a barrier to in utero hematopoietic-cell transplantation (IUHCT) has been a source of debate. Until now, comparisons of allogeneic and congenic engraftment have been inconclusive due to methodologic limitations resulting in minimal and inefficient engraftment. In this study, E14 fetal mice received transplants of either allogeneic or congenic bone marrow using a new intravascular technique that allows definitive administration of much higher doses of donor cells. Our results demonstrate that 100% of surviving recipients demonstrate engraftment at 1 week of age, but that 70% of allogeneic recipients lose engraftment by 1 month of age, and 80% ultimately fail to sustain long-term chimerism. In contrast, all congenic recipients maintain stable, long-term, multilineage chimerism. These results strongly support an immune barrier to allogeneic engraftment after IUHCT.
Collapse
Affiliation(s)
- William H Peranteau
- Center for Fetal Research, Children's Hospital of Philadelphia, PA 19104, USA
| | | | | | | |
Collapse
|
43
|
Peranteau WH, Endo M, Adibe OO, Merchant A, Zoltick PW, Flake AW. CD26 inhibition enhances allogeneic donor-cell homing and engraftment after in utero hematopoietic-cell transplantation. Blood 2006; 108:4268-74. [PMID: 16954501 PMCID: PMC1895454 DOI: 10.1182/blood-2006-04-018986] [Citation(s) in RCA: 75] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
In utero hematopoietic-cell transplantation (IUHCT) can induce donor-specific tolerance to facilitate postnatal transplantation. Induction of tolerance requires a threshold level of mixed hematopoietic chimerism. CD26 is a peptidase whose inhibition increases homing and engraftment of hematopoietic cells in postnatal transplantation. We hypothesized that CD26 inhibition would increase donor-cell homing to the fetal liver (FL) and improve allogeneic engraftment following IUHCT. To evaluate this hypothesis, B6GFP bone marrow (BM) or enriched hematopoietic stem cells (HSCs) were transplanted into allogeneic fetal mice with or without CD26 inhibition. Recipients were analyzed for FL homing and peripheral-blood chimerism from 4 to 28 weeks of life. We found that CD26 inhibition of donor cells results in (1) increased homing of allogeneic BM and HSCs to the FL, (2) an increased number of injected animals with evidence of postnatal engraftment, (3) increased donor chimerism levels following IUHCT, and (4) a competitive engraftment advantage over noninhibited congenic donor cells. This study supports CD26 inhibition as a potential method to increase the level of FL homing and engraftment following IUHCT. The resulting increased donor chimerism suggests that CD26 inhibition may in the future be used as a method of increasing donor-specific tolerance following IUHCT.
Collapse
Affiliation(s)
- William H Peranteau
- The Center for Fetal Research, Children's Hospital of Philadelphia, Abramson Research Bldg, Rm 1116B, 3615 Civic Center Blvd, Philadelphia, PA 19104-4318, USA
| | | | | | | | | | | |
Collapse
|
44
|
Ashizuka S, Peranteau WH, Hayashi S, Flake AW. Busulfan-conditioned bone marrow transplantation results in high-level allogeneic chimerism in mice made tolerant by in utero hematopoietic cell transplantation. Exp Hematol 2006; 34:359-68. [PMID: 16543070 PMCID: PMC1934419 DOI: 10.1016/j.exphem.2005.11.011] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2005] [Revised: 11/16/2005] [Accepted: 11/17/2005] [Indexed: 12/18/2022]
Abstract
OBJECTIVE In utero hematopoietic cell transplantation (IUHCT) is a non-ablative approach that achieves mixed allogeneic chimerism and donor-specific tolerance. However, clinical application of IUHCT has been limited by minimal engraftment. We have previously demonstrated in the murine model that low-level allogeneic chimerism achieved by IUHCT can be enhanced to near-complete donor chimerism by postnatal minimally myeloablative total body irradiation (TBI) followed by same-donor bone marrow transplantation. Because of concerns of toxicity related to even low-dose TBI in early life, we wondered if a potentially less toxic strategy utilizing a single myelosuppressive agent, Busulfan (BU), would provide similar enhancement of engraftment. METHODS In this study, mixed chimerism was created by IUHCT in a fully allogeneic strain combination. After birth, chimeric mice were conditioned with BU followed by transplantation of bone marrow cells congenic to the prenatal donor. RESULTS We demonstrate that: 1) low-level chimerism after IUHCT can be converted to high-level chimerism by this protocol; 2) enhancement of chimerism is BU dose-dependent; and 3) BU reduces the proliferative potential of hematopoietic progenitor cells thus conferring a competitive advantage to the non-BU-treated postnatal donor cells. CONCLUSION This study confirms the potential of IUHCT for facilitation of minimally toxic postnatal regimens to achieve therapeutic levels of allogeneic engraftment.
Collapse
Affiliation(s)
- Shuichi Ashizuka
- The Children's Institute for Surgical Science, The Children's Hospital of Philadelphia, Philadelphia, PA 19104-4318, USA
| | | | | | | |
Collapse
|
45
|
Abstract
In utero haematopoietic cell transplantation (IUHCT) is a promising approach for the treatment of a variety of genetic disorders. The rationale is to take advantage of normal events during haematopoietic and immunological ontogeny to facilitate allogeneic haematopoietic engraftment. Strategies that will be discussed include the direct achievement of therapeutic levels of donor cell engraftment by IUHCT, the achievement of adequate levels of engraftment to donor-specific tolerance by IUHCT, followed by postnatal non-myeloablative regimens to enhance levels of donor cell engraftment into the therapeutic range. Although in utero haematopoietic cell transplantation has been clinically successful in severe combined immunodeficiency disease (SCID), it has been unsuccessful in target disorders where there is not a selective advantage for donor cells. This chapter presents the recognized barriers to engraftment in the fetus and discusses promising experimental strategies to overcome these barriers.
Collapse
Affiliation(s)
- Alan W Flake
- Surgery and Obstetrics and Gynecology, University of Pennsylvania, USA.
| |
Collapse
|
46
|
Bouchard S, MacKenzie TC, Radu AP, Hayashi S, Peranteau WH, Chirmule N, Flake AW. Long-term transgene expression in cardiac and skeletal muscle following fetal administration of adenoviral or adeno-associated viral vectors in mice. J Gene Med 2004; 5:941-50. [PMID: 14601131 DOI: 10.1002/jgm.421] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND In utero gene transfer may provide advantages for the correction of congenital genetic disorders. In the present study we compare the ability of adenovirus (AdCMVLacZ), and two serotypes of adeno-associated virus (AAVCMVLacZ serotypes 2 and 2/5), to target cardiac and skeletal muscle after prenatal systemic or intramuscular injection in mice and assess the immune response to the vectors. METHODS Day 14 gestation fetal mice underwent direct intraperitoneal or intramuscular injection of AdCMVLacZ, and AAVCMVLacZ serotypes 2 and 2/5 vectors. Tissues were processed for beta-galactosidase expression in frozen or high-resolution thin plastic sections at early and late time points. Neutralizing antibodies to Ad and AAV were analyzed in separate fetal experimental and neonatal or adult control groups after administration and re-administration of the vectors. RESULTS A single injection of each vector in utero resulted in sustained expression of beta-galactosidase transgene in skeletal and cardiac muscle. Transgene expression was detected for the length of the study, i.e. 86, 58, and 31 weeks after birth for AdCMVLacZ, and AAVCMVLacZ serotypes 2 and 2/5, respectively. High-level expression in the myocardium was observed independent of the vector or route of administration. Neutralizing antibody responses to AAV and Ad antigens were reduced and long-term expression in muscle was not ablated on postnatal re-administration of vector. CONCLUSIONS Sustained, high-level cardiac and skeletal muscle transgene expression can be obtained after prenatal gene transfer with each of these vectors. The potential for immune response to viral antigens is altered, but not entirely ablated after in utero exposure.
Collapse
Affiliation(s)
- Sarah Bouchard
- Division of Pediatric General, Thoracic, and Fetal Surgery, The Children's Institute for Surgical Science, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | | | | | | | | | | | | |
Collapse
|
47
|
Peranteau WH, Hayashi S, Kim HB, Shaaban AF, Flake AW. In utero hematopoietic cell transplantation: what are the important questions? Fetal Diagn Ther 2004; 19:9-12. [PMID: 14646411 DOI: 10.1159/000074253] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The treatment of congenital hematologic disorders before birth by in utero hematopoietic stem cell transplantation remains a challenging goal. Although success has been achieved in X-linked severe combined immunodeficiency, the approach has failed in all other disorders attempted thus far. In this review, we examine relevant experimental data from the perspective of an analysis of why failure has occurred. We will also attempt to pose the important questions that will need to be answered prior to further clinical attempts to treat most target disorders by this approach.
Collapse
Affiliation(s)
- William H Peranteau
- Children's Institute of Surgical Science, Children's Hospital of Philadelphia, Philadelphia, Pa. 19104-4318, USA
| | | | | | | | | |
Collapse
|
48
|
Hayashi S, Hsieh M, Peranteau WH, Ashizuka S, Flake AW. Complete allogeneic hematopoietic chimerism achieved by in utero hematopoietic cell transplantation and cotransplantation of LLME-treated, MHC-sensitized donor lymphocytes. Exp Hematol 2004; 32:290-9. [PMID: 15003315 DOI: 10.1016/j.exphem.2003.12.008] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2003] [Revised: 12/22/2003] [Accepted: 12/24/2003] [Indexed: 10/26/2022]
Abstract
OBJECTIVE In utero hematopoietic cell transplantation (IUHCT) typically achieves low-level mixed hematopoietic chimerism. However, the goal of IUHCT is to achieve therapeutic levels of chimerism. We hypothesized that prenatal adoptive immunotherapy might achieve high-level donor chimerism after IUHCT. MATERIALS AND METHODS BALB/CE15 fetal mice were transplanted with a mixture of C57BL/6 (B6) T-cell-depleted bone marrow (TCD BM) cells and splenocytes from B6 mice presensitized to BALB/C alloantigen. The splenocytes were preincubated in L-leucyl-L-leucine methyl ester (LLME), to minimize graft vs host disease (GVHD). Recipients were followed after birth for donor cell chimerism and GVHD. RESULTS Full donor hematopoietic chimerism following a single prenatal transplant was achieved in seven transplanted animals. Fully chimeric animals were healthy, without evidence of GVHD, and maintained their engraftment for the duration of the study (48 weeks). However, the addition of presensitized LLME-treated cells decreased survival until weaning relative to TCD BM alone, suggesting that some animals were lost to acute GVHD. Surviving chimeric animals demonstrated increased frequencies of T-regulatory cell populations in their spleen and BM, suggesting that they had successfully suppressed GVHD, allowing survival. CONCLUSIONS This study represents "proof in principle" that prenatal immunotherapeutic strategies may achieve complete hematopoietic engraftment across full MHC barriers when combined with IUHCT. However, strategies with greater hematopoietic specificity must be developed prior to consideration of clinical application.
Collapse
Affiliation(s)
- Satoshi Hayashi
- The Children's Institute for Surgical Science, The Children's Hospital of Philadelphia, Philadelphia, Pa. 19104-4318, USA
| | | | | | | | | |
Collapse
|
49
|
Sefrioui H, Donahue J, Gilpin EA, Srivastava AS, Carrier E. Tolerance and immunity following in utero transplantation of allogeneic fetal liver cells: the cytokine shift. Cell Transplant 2003; 12:75-82. [PMID: 12693667 DOI: 10.3727/000000003783985188] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Although in utero transplantation (IUT) has resulted in donor-specific tolerance to posnatal solid organ transplantation, the mechanisms of this tolerance remain poorly understood. Our recent findings demonstrate that under specific conditions prenatal injection of allogeneic cells may lead to allosensitization instead of tolerance. These laboratory observations were supported by clinical findings as well, and therefore suggested that, depending on the conditions of prenatal transplantation, tolerance or immunity may develop. The present study explored the role of CD4 cells, cytokines, and I-E superantigen in developing tolerance vs. immunity after in utero transplantation. Sixteen animals survived IUT (40-60% survival rate) and were free from any signs of graft-versus-host disease (GVHD). Mice were considered tolerant when their antidonor and antihost CTL responses were similar, sensitized when antidonor responses were significantly higher than antihost and anti-third-party responses, and nontolerant when antidonor responses in transplanted and control mice were similar. The TH1 --> TH2 shift was associated with tolerance and TH2 --> TH1 shift with allosensitization. Our results showed that tolerant BALB/c (H-2d, I-E+) --> CS7BL/6 (H-2b, I-E-) (2/7) mice showed higher IL-4 (p < 0.05) in antidonor MLR, and partial deletion of recipient I-E-reactive T cells (CD3Vbeta11) (p < 0.045). On the other hand, nontolerant animals (5/7) demonstrated high production of IFN-gamma (p < 0.05) without deletion of CD3Vbeta11 T cells. In C57CBL/6 (H-2b, I-E-) --> C3H (H-2k, I-E+) mice CD3Vbeta11 T cells do not play any role in tolerance induction because they are deleted in the C3H background. Tolerant mice (4/9) showed an overproduction of IL-4 (p < 0.05) in antidonor MLR whereas allosensitized animals (5/9) demonstrated high level of IFN-gamma (p < 0.05). Suppressor cells seem to play no role in tolerant C57BL/6 --> C3H as demonstrated by suppressor assay. Hence, a shift from TH1 --> TH2 or TH2 --> TH1 cytokines may determine whether tolerance or immunity develops.
Collapse
Affiliation(s)
- H Sefrioui
- Department of Medicine, Pediatrics, and Family and Preventive Medicine, University of California San Diego School of Medicine, USA
| | | | | | | | | |
Collapse
|
50
|
Abstract
In the near future, prenatal therapy may include stem cell-based cellular therapy or gene therapy. There is considerable overlap in the rationale and potential applications for these 2 approaches. The purpose of this manuscript is to consider current progress in both areas relevant to prenatal treatment. Although clinical application is currently limited to a few highly selected disorders that are amenable to cellular therapy, there is reason to believe that a dramatic increase in application will occur in the near future.
Collapse
Affiliation(s)
- Alan W Flake
- The Center for Fetal Diagnosis and Treatment, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| |
Collapse
|