Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 239
1.
J Pediatric Infect Dis Soc ; 13(1): 75-83, 2024 Jan 29.
Article En | MEDLINE | ID: mdl-38019957

BACKGROUND: Respiratory viral infections are common among pediatric transplant patients, with human rhinovirus (HRV) being the most frequent. In pediatric patients undergoing hemopoietic cell transplant (HCT), infection with HRV has been associated with progression to lower respiratory tract infection (LRTI) and adverse outcomes. We describe the clinical presentation and outcomes of HRV infection in children undergoing HCT. METHODS: Single-center retrospective study. HCT recipients who were positive for HRV/EV (HRV+) or negative for any respiratory virus (VN) by BioFire® FilmArray® panel between October 2014 and December 2017, were included. Primary outcomes were progression to LRTI, ICU admission, all-cause mortality at 3 and 6 months, and respiratory event-related mortality at 6 months. RESULTS: 227 patients (160 allogeneic HCT) were included. Of all patients, 108/227 (47.6%) were HRV+. From all HRV+, 95/108 (88%) were symptomatic and 68/107 (63.6%) of the diagnosis were made pretransplant. The median age of HRV+ was significantly lower than VN patients (5 vs 10 years). Cough and rhinorrhea were more frequently observed in HRV+ (53.7 and 60% vs 19.8 and 22.8%, respectively). No differences were found between both groups pretransplant and overall in rates progression to LRTI, ICU admission, mechanical ventilation, all-cause within 3 and 6 months, and mortality related with respiratory failure. No significant association was found between the severity of respiratory disease and the type of conditioning, type of transplant, or absolute lymphocyte count. CONCLUSIONS: HRV infection is frequently detected in HCT recipients but is not associated with severity of respiratory disease, need for intensive care unit or mortality, including those diagnosed before transplant, suggesting that delaying HCT in this scenario may not be needed. Multicenter larger studies are required to confirm these findings.


Enterovirus Infections , Enterovirus , Picornaviridae Infections , Respiratory Tract Infections , Child , Humans , Cell Transplantation/adverse effects , Retrospective Studies , Rhinovirus , Child, Preschool , Infant
2.
J Clin Endocrinol Metab ; 108(6): 1273-1289, 2023 05 17.
Article En | MEDLINE | ID: mdl-36611246

Primary adrenal insufficiency (PAI) occurs in 1 in 5 to 7000 adults. Leading etiologies are autoimmune adrenalitis in adults and congenital adrenal hyperplasia (CAH) in children. Oral replacement of cortisol is lifesaving, but poor quality of life, repeated adrenal crises, and dosing uncertainty related to lack of a validated biomarker for glucocorticoid sufficiency persists. Adrenocortical cell therapy and gene therapy may obviate many of the shortcomings of adrenal hormone replacement. Physiological cortisol secretion regulated by pituitary adrenocorticotropin could be achieved through allogeneic adrenocortical cell transplantation, production of adrenal-like steroidogenic cells from either stem cells or lineage conversion of differentiated cells, or for CAH, gene therapy to replace or repair a defective gene. The adrenal cortex is a high-turnover organ and thus failure to incorporate progenitor cells within a transplant will ultimately result in graft exhaustion. Identification of adrenocortical progenitor cells is equally important in gene therapy, for which new genetic material must be specifically integrated into the genome of progenitors to ensure a durable effect. Delivery of gene-editing machinery and a donor template, allowing targeted correction of the 21-hydroxylase gene, has the potential to achieve this. This review describes advances in adrenal cell transplants and gene therapy that may allow physiological cortisol production for children and adults with PAI.


Adrenal Hyperplasia, Congenital , Adrenal Insufficiency , Child , Adult , Humans , Hydrocortisone , Quality of Life , Adrenal Insufficiency/genetics , Adrenal Insufficiency/therapy , Adrenal Insufficiency/complications , Adrenal Hyperplasia, Congenital/genetics , Genetic Therapy/adverse effects , Cell Transplantation/adverse effects
3.
Physiol Behav ; 251: 113807, 2022 07 01.
Article En | MEDLINE | ID: mdl-35427673

BACKGROUND: Cell therapy is a promising treatment method for relieving neuropathic pain caused by spinal cord injuries (SCI). Sertoli cells (SCs) are an attractive choice given their demonstrated secretion of growth factors and immunosuppressant effect. This study mechanistically characterizes the analgesic effect of SCs transplantation. METHODS: The clip compression SCI model was carried out on the T12-T13 level in male Wistar rats. One-week post-SCI, SCs were transplanted into the site of injury. Animals underwent Basso, Beattie, and Bresnahan locomotor scoring, mechanical allodynia, and thermal hyperalgesia on a weekly basis for a duration of six weeks. Histological examination of the spinal cord and molecular evaluation of Iba-1, P2Y4, TRPC6, and P-mTOR were performed. SCs survival, measured by anti-Müllerian hormone expression in the spinal cord. RESULTS: Animals that received SCs transplantation showed improvement in motor function recovery and pain relief. Furthermore, a cavity was significantly decreased in the transplanted animals (p = 0.0024), the expression level of TRPC6 and caspase3 and the number of activated microglia decreased compared to the SCI animals, and p-mTOR and P2Y4R expression remarkably increased compared to the SCI group. CONCLUSION: SCs transplantation produces an analgesic effect which may represent a promising treatment for SCI-induced chronic pain.


Neuralgia , Spinal Cord Injuries , Analgesics , Animals , Cell Transplantation/adverse effects , Hyperalgesia/etiology , Hyperalgesia/therapy , Male , Microglia/metabolism , Neuralgia/etiology , Neuralgia/metabolism , Neuralgia/therapy , Rats , Rats, Sprague-Dawley , Rats, Wistar , Sertoli Cells/metabolism , Sertoli Cells/pathology , Spinal Cord/pathology , Spinal Cord Injuries/complications , Spinal Cord Injuries/therapy , TOR Serine-Threonine Kinases/metabolism , TRPC Cation Channels , TRPC6 Cation Channel/metabolism
4.
Brasília; CONITEC; fev. 2022.
Non-conventional Pt | BRISA | ID: biblio-1370292

INTRODUÇÃO: O mieloma múltiplo é uma neoplasia de plasmócitos com incidência anual no Brasil de 1,24 casos a cada 100.000 habitantes. Apesar de avanços no tratamento da doença, o mieloma múltiplo é considerado incurável e o objetivo da terapia é induzir a remissão e prolongar a sobrevida do paciente preservando sua qualidade de vida. Um dos principais tratamentos é o TCTH. No entanto, uma parcela significativa dos pacientes não é elegível ao TCTH podendo ser apenas submetida a medicamentos. A lenalidomida é um imunomodulador que pode ser utilizado na terapia de indução e manutenção nesses pacientes. Desta forma, o objetivo deste Relatório é comparar eficácia, segurança, custo-efetividade e impacto orçamentário da lenalidomida em relação à talidomida, imunomodulador disponível no SUS, em pacientes com mieloma múltiplo inelegíveis ao TCTH. TECNOLOGIA: Lenalidomida. PERGUNTA: A lenalidomida é mais eficaz, segura e custo-efetiva em esquemas com dois ou três medicamentos comparada à talidomida em esquemas terapêuticos com dois ou três medicamentos para o t


Humans , Thalidomide/therapeutic use , Cell Transplantation/adverse effects , Lenalidomide/therapeutic use , Multiple Myeloma/drug therapy , Unified Health System , Brazil , Cost-Benefit Analysis/economics
5.
J Virol ; 96(3): e0139421, 2022 02 09.
Article En | MEDLINE | ID: mdl-34818071

Humanized mouse models are based on the engraftment of human cells in immunodeficient mouse strains, most notably the NSG strain. Most used models have a major limitation in common, the development of graft-versus-host disease (GVHD). GVHD not only introduces variabilities into the research data but also leads to animal welfare concerns. A new mouse strain, B6.129S-Rag2tm1Fwa CD47tm1Fpl Il2rgtm1Wjl/J, which lacks Rag1, IL2rg, and CD47 (triple knockout [TKO]), is resistant to GVHD development. We transplanted TKO mice with human peripheral blood mononuclear cells (PBMCs) to establish a new humanized PBMC (hu-PBMC) mouse model. A cohort of these mice was infected with HIV-1 and monitored for plasma HIV viremia and CD4+ T cell depletion. The onset and progression of GVHD were monitored by clinical signs. This study demonstrates that TKO mice transplanted with human PBMCs support engraftment of human immune cells in primary and secondary lymphoid tissues, rectum, and brain. Moreover, the TKO hu-PBMC model supports HIV-1 infection via the intraperitoneal, rectal, or vaginal route, as confirmed by robust plasma HIV viremia and CD4+ T cell depletion. Lastly, TKO mice showed a delayed onset of GVHD clinical signs (∼24 days) and exhibited significant decreases in plasma levels of tumor necrosis factor beta (TNF-ß). Based on these results, the TKO hu-PBMC mouse model not only supports humanization and HIV-1 infection but also has a delayed onset of GVHD development, making this model a valuable tool in HIV research. IMPORTANCE Currently, there is no cure or vaccine for HIV infection; thus, continued research is needed to end the HIV pandemic. While many animal models are used in HIV research, none is used more than the humanized mouse model. A major limitation with current humanized mouse models is the development of graft-versus-host disease (GVHD). Here, we describe a novel humanized-PBMC mouse model that has a delayed onset GVHD development and supports and models HIV infection comparably to well-established humanized mouse models.


Cell Transplantation , Disease Models, Animal , Graft vs Host Disease/etiology , HIV Infections/immunology , HIV Infections/virology , Leukocytes, Mononuclear/transplantation , Animals , Cell Transplantation/adverse effects , Cell Transplantation/methods , Disease Susceptibility , Female , HIV-1/immunology , Humans , Male , Mice , Mice, Knockout , Research , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Time Factors
6.
J Cell Mol Med ; 25(22): 10554-10564, 2021 11.
Article En | MEDLINE | ID: mdl-34632708

Acute respiratory distress syndrome (ARDS) is the most common cause of death in COVID-19 patients. The cytokine storm is the main driver of the severity and magnitude of ARDS. Placenta-derived decidua stromal cells (DSCs) have a stronger immunosuppressive effect than other sources of mesenchymal stromal cells. Safety and efficacy study included 10 patients with a median age of 50 (range 14-68) years with COVID-19-induced ARDS. DSCs were administered 1-2 times at a dose of 1 × 106 /kg. End points were safety and efficacy by survival, oxygenation and effects on levels of cytokines. Oxygenation levels increased from a median of 80.5% (range 69-88) to 95% (range 78-99) (p = 0.012), and pulmonary infiltrates disappeared in all patients. Levels of IL-6 decreased from a median of 69.3 (range 35.0-253.4) to 11 (range 4.0-38.3) pg/ml (p = 0.018), and CRP decreased from 69 (range 5-169) to 6 (range 2-31) mg/ml (p = 0.028). Two patients died, one of a myocardial infarction and the other of multiple organ failure, diagnosed before the DSC therapy. The other patients recovered and left the intensive care unit (ICU) within a median of 6 (range 3-12) days. DSC therapy is safe and capable of improving oxygenation, decreasing inflammatory cytokine level and clearing pulmonary infiltrates in patients with COVID-19.


COVID-19 Drug Treatment , Cell Transplantation/methods , Cytokine Release Syndrome/therapy , Respiratory Distress Syndrome/virology , Stromal Cells/transplantation , Adolescent , Adult , Aged , COVID-19/complications , COVID-19/therapy , Cell Transplantation/adverse effects , Cytokine Release Syndrome/etiology , Cytokines/blood , Female , Humans , Length of Stay , Male , Middle Aged , Placenta/cytology , Pregnancy , Respiratory Distress Syndrome/therapy , Stromal Cells/physiology , Treatment Outcome
7.
Front Immunol ; 12: 664244, 2021.
Article En | MEDLINE | ID: mdl-33841448

A number of immune regulatory cellular therapies, including regulatory T cells and mesenchymal stromal cells, have emerged as novel alternative therapies for the control of transplant alloresponses. Clinical studies have demonstrated their feasibility and safety, however developing our understanding of the impact of cellular therapeutics in vivo requires advanced immune monitoring strategies. To accurately monitor the immune response, a combination of complementary methods is required to measure the cellular and molecular phenotype as well as the function of cells involved. In this review we focus on the current immune monitoring strategies and discuss which methods may be utilized in the future.


Cell Transplantation , Cell- and Tissue-Based Therapy , Clinical Trials as Topic , Monitoring, Immunologic/methods , Animals , Cell Transplantation/adverse effects , Cell Transplantation/methods , Cell- and Tissue-Based Therapy/adverse effects , Cell- and Tissue-Based Therapy/methods , Cell- and Tissue-Based Therapy/standards , Clinical Decision-Making , Clinical Trials as Topic/standards , Disease Management , Humans , Monitoring, Immunologic/standards , Organ Specificity , Treatment Outcome
8.
Angiology ; 72(9): 861-866, 2021 10.
Article En | MEDLINE | ID: mdl-33783233

Autologous cell therapy (ACT) is a new treatment for patients with no-option critical limb ischemia (NO-CLI). We evaluated the factors involved in the nonresponse to ACT in patients with CLI and diabetic foot. Diabetic patients (n = 72) with NO-CLI treated using ACT in our foot clinic over a period of 8 years were divided into responders (n = 57) and nonresponders (n = 15). Nonresponder was defined as an insufficient increase in transcutaneous oxygen pressure by <5 mm Hg, 3 months after ACT. Patient demographics, diabetes duration and treatment, and comorbidities as well as a cellular response to ACT, limb-related factors, and the presence of inherited thrombotic disorders were compared between the 2 groups. The main independent predictors for an impaired response to ACT were heterozygote Leiden mutation (OR 10.5; 95% CI, 1.72-4) and homozygote methylenetetrahydrofolate reductase (MTHFR 677) mutation (OR 3.36; 95% CI, 1.0-14.3) in stepwise logistic regression. Univariate analysis showed that lower mean protein C levels (P = .041) were present in nonresponders compared with responders. In conclusion, the significant predictors of an impaired response to ACT in diabetic patients with NO-CLI were inherited thrombotic disorders.


Blood Coagulation Disorders, Inherited/complications , Cell Transplantation , Diabetic Foot/surgery , Ischemia/surgery , Activated Protein C Resistance/complications , Activated Protein C Resistance/genetics , Aged , Blood Coagulation Disorders, Inherited/diagnosis , Blood Coagulation Disorders, Inherited/genetics , Cell Transplantation/adverse effects , Critical Illness , Diabetic Foot/complications , Diabetic Foot/diagnosis , Factor V/genetics , Female , Heterozygote , Homozygote , Humans , Ischemia/complications , Ischemia/diagnosis , Male , Methylenetetrahydrofolate Reductase (NADPH2)/genetics , Middle Aged , Mutation , Risk Assessment , Risk Factors , Transplantation, Autologous , Treatment Failure
10.
Methods Mol Biol ; 2174: 219-244, 2021.
Article En | MEDLINE | ID: mdl-32813253

The use of immunotherapy as an alternative treatment for cancer patients has become of great interest in the scientific community as it is required to overcome many of the currently unsolved problems such as tumor escape, immunosuppression and unwanted unspecific toxicity. The use of chimeric antigen receptor T cells has been a very successful strategy in some hematologic malignancies. However, the application of CAR T cells has been limited to solid tumors, and this has aimed the development of new generation of CARs with enhanced effectivity and specificity. Here, we review the state of the art of CAR T cell therapy with special emphasis on the current challenges and opportunities.


Cell Transplantation/adverse effects , Cell Transplantation/methods , Neoplasms/therapy , Receptors, Chimeric Antigen , T-Lymphocytes/physiology , Genes, Transgenic, Suicide , Hematologic Neoplasms/therapy , Humans , Receptors, Chimeric Antigen/chemistry , Receptors, Chimeric Antigen/genetics , Receptors, Chimeric Antigen/metabolism , T-Lymphocytes/cytology
11.
Neurobiol Dis ; 148: 105175, 2021 01.
Article En | MEDLINE | ID: mdl-33188920

Prevalent in approximately 20% of the worldwide human population, the rs6265 (also called 'Val66Met') single nucleotide polymorphism (SNP) in the gene for brain-derived neurotrophic factor (BDNF) is a common genetic variant that can alter therapeutic responses in individuals with Parkinson's disease (PD). Possession of the variant Met allele results in decreased activity-dependent release of BDNF. Given the resurgent worldwide interest in neural transplantation for PD and the biological relevance of BDNF, the current studies examined the effects of the rs6265 SNP on therapeutic efficacy and side-effect development following primary dopamine (DA) neuron transplantation. Considering the significant reduction in BDNF release associated with rs6265, we hypothesized that rs6265-mediated dysfunctional BDNF signaling contributes to the limited clinical benefit observed in a subpopulation of PD patients despite robust survival of grafted DA neurons, and further, that this mutation contributes to the development of aberrant graft-induced dyskinesias (GID). To this end, we generated a CRISPR knock-in rat model of the rs6265 BDNF SNP to examine for the first time the influence of a common genetic polymorphism on graft survival, functional efficacy, and side-effect liability, comparing these parameters between wild-type (Val/Val) rats and those homozygous for the variant Met allele (Met/Met). Counter to our hypothesis, the current research indicates that Met/Met rats show enhanced graft-associated therapeutic efficacy and a paradoxical enhancement of graft-derived neurite outgrowth compared to wild-type rats. However, consistent with our hypothesis, we demonstrate that the rs6265 genotype in the host rat is strongly linked to development of GID, and that this behavioral phenotype is significantly correlated with neurochemical signatures of atypical glutamatergic neurotransmission by grafted DA neurons.


Brain-Derived Neurotrophic Factor/genetics , Cell Transplantation/methods , Dopaminergic Neurons/transplantation , Dyskinesias/genetics , Animals , Antiparkinson Agents/adverse effects , Cell Transplantation/adverse effects , Dopaminergic Neurons/metabolism , Dyskinesia, Drug-Induced/etiology , Dyskinesias/etiology , Embryo, Mammalian , Gene Knock-In Techniques , Levodopa/adverse effects , Mesencephalon/cytology , Oxidopamine/toxicity , Parkinson Disease, Secondary/chemically induced , Rats , Sympatholytics/toxicity , Vesicular Glutamate Transport Protein 2/metabolism
12.
Biochem Biophys Res Commun ; 534: 765-772, 2021 01 01.
Article En | MEDLINE | ID: mdl-33213838

Modifying T cells to attack tumors using engineered chimeric receptors display powerful new therapeutic capabilities. Unfortunately, the effectiveness of therapeutic T cells is limited due to the inherent T cell responses: certain facets of endogenous response programs may be toxic, and the ability to overcome the immunosuppression in TME is deficient. Here we developed a Notch receptor based synNotch T cell platform that is able to response to target tumor cells and selectively lead to CXCL10 production. Further study showed that the administration of synNotch T cells significantly inhibited the tumor growth in a humanized murine model, accompanied by the increased infiltration of CD3+T cells and elevated level of CXCL10 and IFN-γ in the tumor site. A slightly increased level of CXCL10 and limited IFN-γ were found in the serum in mice received synNotch T cells, suggesting a high security of this treatment. Finally, we demonstrated that CXCL10 is sufficient and indispensable for the synNotch T cells induced anti-tumor effect. This study provided theoretical and experimental bases for the clinical implication of CXCL10 encoding synNotch T cells.


Cell Transplantation/methods , Chemokine CXCL10/metabolism , Lymphocytes, Tumor-Infiltrating/immunology , T-Lymphocytes/immunology , A549 Cells , Animals , Cell Transplantation/adverse effects , Chemokine CXCL10/genetics , Coculture Techniques , Cytokine Release Syndrome/etiology , GPI-Linked Proteins/metabolism , Humans , Interferon-gamma/metabolism , Male , Mesothelin , Mice, Inbred NOD , T-Lymphocytes/metabolism , T-Lymphocytes/transplantation , Xenograft Model Antitumor Assays
13.
Immunol Lett ; 229: 55-61, 2021 01.
Article En | MEDLINE | ID: mdl-33253759

Humanized mice are widely used to study the human immune system in vivo and develop therapies for various human diseases. Human peripheral blood mononuclear cells (PBMC)-engrafted NOD/Shi-scid IL2rγnull (NOG) mice are useful models for characterization of human T cells. However, the development of graft-versus-host disease (GVHD) limits the use of NOG PBMC models. We previously established a NOG-major histocompatibility complex class I/II double knockout (dKO) mouse model. Although humanized dKO mice do not develop severe GVHD, they have impaired reproductive performance and reduced chimerism of human cells. In this study, we established a novel beta-2 microglobulin (B2m) KO mouse model using CRISPR/Cas9. By crossing B2m KO mice with I-Ab KO mice, we established a modified dKO (dKO-em) mouse model. Reproductivity was slightly improved in dKO-em mice, compared with conventional dKO (dKO-tm) mice. dKO-em mice showed no signs of GVHD after the transfer of human PBMCs; they also exhibited high engraftment efficiency. Engrafted human PBMCs survived significantly longer in the peripheral blood and spleens of dKO-em mice, compared with dKO-tm mice. In conclusion, dKO-em mice might constitute a promising PBMC-based humanized mouse model for the development and preclinical testing of novel therapeutics for human diseases.


CRISPR-Cas Systems , Cell Transplantation , Gene Knockout Techniques , Histocompatibility Antigens/genetics , Interleukin Receptor Common gamma Subunit/deficiency , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/metabolism , Animals , Biomarkers , Cell Transplantation/adverse effects , Cell Transplantation/methods , Gene Editing , Gene Targeting , Genetic Loci , Graft Survival , Graft vs Host Disease/diagnosis , Graft vs Host Disease/etiology , Humans , Immunohistochemistry , Immunophenotyping , Mice , Mice, Inbred NOD , Mice, Knockout , Models, Animal , Severity of Illness Index , Spleen/immunology , Spleen/metabolism
14.
Nat Commun ; 11(1): 6265, 2020 12 08.
Article En | MEDLINE | ID: mdl-33293538

Cell and gene therapies offer opportunities for treating disease with potential to restore function, and cure disease. However, they are not without risk and pose complex logistical, economic, ethical and social challenges for health systems. Here we report our systematic review of the current evidence on patient and public knowledge and perspectives of cell and gene therapies, to inform future research, education and awareness raising activities. We screened 10,735 titles and abstracts, and evaluated the full texts of 151 publications. The final selection was 35 publications. Four themes were generated from the narrative synthesis of the study findings namely: (1) Knowledge and understanding of cell and gene therapies, (2) Acceptance of cell and gene therapies (3) Understanding of risk and benefits of therapy, and (4) Information needs and current sources of information. As potential funders or future recipients, it is important that the public and patients are aware of these therapies, understand the issues involved, and can contribute to the debate. This review highlights the need for appropriate patient and public education on the various aspects of cell and gene therapies. High quality studies exploring patient and public opinions and experiences of cell and gene therapy are required. Patient and public perceptions of these therapies, alongside evidence of clinical and cost-effectiveness, will be central to their uptake and use.


Cell Transplantation/methods , Delivery of Health Care/ethics , Genetic Therapy/methods , Public Opinion , Cell Transplantation/adverse effects , Cell Transplantation/economics , Cell Transplantation/ethics , Cost-Benefit Analysis , Delivery of Health Care/economics , Genetic Therapy/adverse effects , Genetic Therapy/economics , Genetic Therapy/ethics , Health Education , Humans , Patient Education as Topic , Qualitative Research
15.
Front Immunol ; 11: 565236, 2020.
Article En | MEDLINE | ID: mdl-33193333

Chimeric Antigen Receptor-T cells (CAR-T) are considered novel biological agents, designed to selectively attack cancer cells expressing specific antigens, with demonstrated clinical activity in patients affected with relapsed/refractory B-cell malignancies. In consideration of their complexity, the use of CAR-T requires dedicated clinical setting and health care practitioners with expertise in the selection, treatment, and management of toxicities and side effects. Such issue appears particularly important when contextualized in the rapid progress of CAR-T cell treatment, translating into a constant need of updating and evolution. Moreover, the clinical grade manufacturing of CAR-T cells is complex and implies articulated regulatory and organizational aspects. The main goal of this review is to summarize and provide an accurate analysis of the clinical, logistic, and regulatory requirements of CAR-T cell centers. Finally, we describe a new occupational figure called "CAR-T specialist" devoted to the establishment and coordination of the required facilities and regulatory landscape in the context of cancer centers.


Antigens, Neoplasm/immunology , Cell Transplantation/adverse effects , Immunotherapy, Adoptive/adverse effects , Neoplasms/therapy , Receptors, Chimeric Antigen/immunology , T-Lymphocytes/immunology , Aftercare/methods , Antigens, CD19/immunology , Blood Donors/legislation & jurisprudence , Cell Transplantation/legislation & jurisprudence , Health Personnel/education , Humans , Immunotherapy, Adoptive/legislation & jurisprudence , Patient Selection , Transplants , Tumor Microenvironment/immunology
16.
Arch Dis Child Fetal Neonatal Ed ; 105(5): 563-568, 2020 Sep.
Article En | MEDLINE | ID: mdl-32253200

Recent decades have seen the rapid progress of neonatal intensive care, and the survival rates of the most preterm infants are improving. This improvement is associated with changing patterns of morbidity and new phenotypes of bronchopulmonary dysplasia and preterm brain injury are recognised. Inflammation and immaturity are known contributors to their pathogenesis. However, a new phenomenon, the exhaustion of progenitor cells is emerging as an important factor. Current therapeutic approaches do not adequately address these new mechanisms of injury. Cell therapy, that is the use of stem and stem-like cells, with its potential to both repair and prevent injury, offers a new approach to these challenging conditions. This review will examine the rationale for cell therapy in the extremely preterm infant, the preclinical and early clinical evidence to support its use in bronchopulmonary dysplasia and preterm brain injury. Finally, it will address the challenges in translating cell therapy from the laboratory to early clinical trials.


Brain Injuries/therapy , Bronchopulmonary Dysplasia/therapy , Cell Transplantation/methods , Infant, Extremely Premature , Infant, Premature, Diseases/therapy , Animals , Cell Transplantation/adverse effects , Clinical Protocols , Clinical Trials as Topic , Cord Blood Stem Cell Transplantation/adverse effects , Cord Blood Stem Cell Transplantation/methods , Fetal Blood , Humans , Infant, Newborn , Infant, Very Low Birth Weight , Intensive Care, Neonatal/methods , Mesenchymal Stem Cell Transplantation/adverse effects , Mesenchymal Stem Cell Transplantation/methods , Time Factors
17.
Spinal Cord ; 58(8): 844-856, 2020 08.
Article En | MEDLINE | ID: mdl-32249830

INTRODUCTION: Spinal cord injury (SCI) is associated with significant and life-long disability. Yet, despite decades of research, no regenerative treatment has reached clinical practice. Cell-based therapies are one possible regenerative strategy beginning to transfer to human trials from a more extensive pre-clinical basis. METHODS: We therefore conducted a scoping review to synthesise all cell-based trials in SCI to consider the current state of the field and the cell transplant type or strategy with greatest promise. A search strategy of MEDLINE returned 1513 results. All clinical trials including adult human patients with acute or chronic, compete or incomplete SCI and a recorded ASIA score were sought. Exclusion criteria included non-traumatic SCI, paediatric patients and animal studies. A total of 43 studies, treating 1061 patients, were identified. Most trials evaluated cells from the bone marrow (22 papers, 660 patients) or the olfactory bulb (10 papers, 245 patients). RESULTS: Cell transplantation does appear to be safe, with no serious adverse effects being reported in the short-term. 86% of trials described efficacy as a primary outcome. However, varying degrees of outcome reporting prevented meta-analysis. No emerging cell type or technique was identified. The majority of trials, 53%, took place in developing countries, which may suggest more stringent regulatory requirements within Western countries. CONCLUSION: We believe cell-based transplantation translation remains in its infancy and that, although further robust clinical research is required, it is an important strategy to consider in the treatment of SCI.


Cell Transplantation , Outcome Assessment, Health Care , Spinal Cord Injuries/therapy , Cell Transplantation/adverse effects , Cell Transplantation/statistics & numerical data , Humans , Outcome Assessment, Health Care/statistics & numerical data
18.
Methods Mol Biol ; 2140: 43-64, 2020.
Article En | MEDLINE | ID: mdl-32207105

3D bioprinting involves engineering live cells into a 3D structure, using a 3D printer to print cells, often together with a compatible 3D scaffold. 3D-printed cells and tissues may be used for a range of purposes including medical research, in vitro drug testing, and in vivo transplantation. The inclusion of living cells and biomaterials in the 3D printing process raises ethical, policy, and regulatory issues at each stage of the bioprinting process that include the source of cells and materials, stability and biocompatibility of cells and materials, disposal of 3D-printed materials, intended use, and long-term effects. This chapter focuses on the ethical issues that arise from 3D bioprinting in the lab-from consideration of the source of cells and materials, ensuring their quality and safety, through to testing of bioprinted materials in animal and human trials. It also provides guidance on where to seek information concerning appropriate regulatory frameworks and guidelines, including on classification and patenting of 3D-bioprinted materials, and identifies regulatory gaps that deserve attention.


Bioprinting/ethics , Printing, Three-Dimensional/ethics , Animal Experimentation/ethics , Animal Experimentation/standards , Animals , Cell Transplantation/adverse effects , Cell Transplantation/ethics , Clinical Trials as Topic/ethics , Evaluation Studies as Topic , Human Experimentation/ethics , Humans , Implants, Experimental/adverse effects , Implants, Experimental/ethics , Intellectual Property , Patents as Topic , Policy , Practice Guidelines as Topic , Stem Cells , Tissue Engineering/ethics , Tissue Scaffolds/adverse effects
19.
J Diabetes Sci Technol ; 14(2): 212-225, 2020 03.
Article En | MEDLINE | ID: mdl-32116026

Islet transplantation is a promising curative treatment option for type 1 diabetes (T1D) as it can provide physiological blood glucose control. The widespread utilization of islet transplantation is limited due to systemic immunosuppression requirements, persisting graft immunodestruction, and poor islet engraftment. Traditional macro- and micropolymeric encapsulation strategies can alleviate the need for antirejection immunosuppression, yet the increased graft volume and diffusional distances imparted by these coatings can be detrimental to graft viability and glucose control. Additionally, systemic administration of pro-engraftment and antirejection therapeutics leaves patients vulnerable to adverse off-target side effects. Nanoscale engineering techniques can be used to immunocamouflage islets, modulate the transplant microenvironment, and provide localized pro-engraftment cues. In this review, we discuss the applications of nanotechnology to advance the clinical potential of islet transplantation, with a focus on cell surface engineering, bioactive functionalization, and use of nanoparticles in T1D cell-based treatments.


Cell Transplantation , Diabetes Mellitus, Type 1/therapy , Immunity , Nanotechnology/methods , Animals , Cell Transplantation/adverse effects , Diabetes Mellitus, Experimental/immunology , Diabetes Mellitus, Experimental/pathology , Diabetes Mellitus, Experimental/therapy , Diabetes Mellitus, Type 1/immunology , Graft Survival/immunology , Humans , Islets of Langerhans/immunology , Islets of Langerhans Transplantation/immunology , Nanotechnology/trends , Transplantation Immunology
20.
J Am Acad Dermatol ; 83(1): 109-116, 2020 Jul.
Article En | MEDLINE | ID: mdl-32147390

BACKGROUND: Few effective treatments are available for male pattern hair loss (MPHL) or, especially, for female pattern hair loss (FPHL). Recently, cell-based therapies using autologous or allogeneic cells have been used clinically. OBJECTIVE: We examined the safety and efficacy of autologous cell-based therapy using dermal sheath cup (DSC) cells to treat MPHL and FPHL. METHODS: DSCs dissected from occipital hair follicles were cultured to manufacture DSC cells. Participants with MPHL or FPHL received single injections of 7.5 × 106, 1.5 × 106, or 3.0 × 105 DSC cells or a placebo in 4 randomized separate regions on the scalp, and hair densities and diameters were measured for 3, 6, 9, and 12 months. RESULTS: Fifty men and 15 women aged 33 to 64 years were injected with DSC cells. Total hair density and cumulative hair diameter at the 3.0 × 105 DSC cells injection site was significantly increased compared with the placebo after 6 and 9 months. Men and women showed similar improvements, and there were no serious adverse events. LIMITATIONS: No lower cell numbers were tested, and the positive effect was temporary until 9 months. CONCLUSION: The results suggest that cell therapy with autologous DSC cells may be useful as a new therapeutic method for treating MPHL and FPHL.


Alopecia/therapy , Cell Transplantation , Hair Follicle/cytology , Adult , Cell Transplantation/adverse effects , Double-Blind Method , Female , Hair/anatomy & histology , Hair/growth & development , Humans , Male , Middle Aged , Proof of Concept Study , Transplantation, Autologous , Treatment Outcome
...