Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
Add more filters










Publication year range
1.
Mamm Genome ; 33(1): 230-240, 2022 03.
Article in English | MEDLINE | ID: mdl-34476572

ABSTRACT

Swine biomedical models have been gaining in popularity over the last decade, particularly for applications in oncology research. Swine models for cancer research include pigs that have severe combined immunodeficiency for xenotransplantation studies, genetically modified swine models which are capable of developing tumors in vivo, as well as normal immunocompetent pigs. In recent years, there has been a low success rate for the approval of new oncological therapeutics in clinical trials. The two leading reasons for these failures are either due to toxicity and safety issues or lack of efficacy. As all therapeutics must be tested within animal models prior to clinical testing, there are opportunities to expand the ability to assess efficacy and toxicity profiles within the preclinical testing phases of new therapeutics. Most preclinical in vivo testing is performed in mice, canines, and non-human primates. However, swine models are an alternative large animal model for cancer research with similarity to human size, genetics, and physiology. Additionally, tumorigenesis pathways are similar between human and pigs in that similar driver mutations are required for transformation. Due to their larger size, the development of orthotopic tumors is easier than in smaller rodent models; additionally, porcine models can be harnessed for testing of new interventional devices and radiological/surgical approaches as well. Taken together, swine are a feasible option for preclinical therapeutic and device testing. The goals of this resource are to provide a broad overview on regulatory processes required for new therapeutics and devices for use in the clinic, cross-species differences in oncological therapeutic responses, as well as to provide an overview of swine oncology models that have been developed that could be used for preclinical testing to fulfill regulatory requirements.


Subject(s)
Neoplasms , Translational Research, Biomedical , Animals , Disease Models, Animal , Dogs , Mice , Neoplasms/genetics , Neoplasms/therapy , Primates , Swine , Transplantation, Heterologous
5.
Front Immunol ; 11: 510, 2020.
Article in English | MEDLINE | ID: mdl-32296428

ABSTRACT

Severe combined immunodeficiency (SCID) is described as the lack of functional T and B cells. In some cases, mutant genes encoding proteins involved in the process of VDJ recombination retain partial activity and are classified as hypomorphs. Hypomorphic activity in the products from these genes can function in the development of T and B cells and is referred to as a leaky phenotype in patients and animals diagnosed with SCID. We previously described two natural, single nucleotide variants in ARTEMIS (DCLR1EC) in a line of Yorkshire pigs that resulted in SCID. One allele contains a splice site mutation within intron 8 of the ARTEMIS gene (ART16), while the other mutation is within exon 10 that results in a premature stop codon (ART12). While initially characterized as SCID and lacking normal levels of circulating lymphoid cells, low levels of CD3ε+ cells can be detected in most SCID animals. Upon further assessment, we found that ART16/16, and ART12/12 SCID pigs had abnormally small populations of CD3ε+ cells, but not CD79α+ cells, in circulation and lymph nodes. Newborn pigs (0 days of age) had CD3ε+ cells within lymph nodes prior to any environmental exposure. CD3ε+ cells in SCID pigs appeared to have a skewed CD4α+CD8α+CD8ß- T helper memory phenotype. Additionally, in some pigs, rearranged VDJ joints were detected in lymph node cells as probed by PCR amplification of TCRδ V5 and J1 genomic loci, as well as TCRß V20 and J1.1, providing molecular evidence of residual Artemis activity. We additionally confirmed that TCRα and TCRδ constant region transcripts were expressed in the thymic and lymph node tissues of SCID pigs; although the expression pattern was abnormal compared to carrier animals. The leaky phenotype is important to characterize, as SCID pigs are an important tool for biomedical research and this additional phenotype may need to be considered. The pig model also provides a relevant model for hypomorphic human SCID patients.


Subject(s)
DNA-Binding Proteins/genetics , Disease Models, Animal , Endonucleases/genetics , Severe Combined Immunodeficiency/genetics , Severe Combined Immunodeficiency/immunology , T-Lymphocyte Subsets/immunology , Animals , CD3 Complex , Swine
6.
Front Immunol ; 11: 100, 2020.
Article in English | MEDLINE | ID: mdl-32117254

ABSTRACT

Pigs with severe combined immunodeficiency (SCID) are an emerging biomedical animal model. Swine are anatomically and physiologically more similar to humans than mice, making them an invaluable tool for preclinical regenerative medicine and cancer research. One essential step in further developing this model is the immunological humanization of SCID pigs. In this work we have generated T- B- NK- SCID pigs through site directed CRISPR/Cas9 mutagenesis of IL2RG within a naturally occurring DCLRE1C (ARTEMIS)-/- genetic background. We confirmed ART-/-IL2RG-/Y pigs lacked T, B, and NK cells in both peripheral blood and lymphoid tissues. Additionally, we successfully performed a bone marrow transplant on one ART-/-IL2RG-/Y male SCID pig with bone marrow from a complete swine leukocyte antigen (SLA) matched donor without conditioning to reconstitute porcine T and NK cells. Next, we performed in utero injections of cultured human CD34+ selected cord blood cells into the fetal ART-/-IL2RG-/Y SCID pigs. At birth, human CD45+ CD3ε+ cells were detected in cord and peripheral blood of in utero injected SCID piglets. Human leukocytes were also detected within the bone marrow, spleen, liver, thymus, and mesenteric lymph nodes of these animals. Taken together, we describe critical steps forwards the development of an immunologically humanized SCID pig model.


Subject(s)
Bone Marrow Transplantation , Interleukin Receptor Common gamma Subunit/genetics , Severe Combined Immunodeficiency/genetics , Animals , Animals, Genetically Modified , Antigens, CD34 , CRISPR-Cas Systems , Cell Differentiation , Chimera , DNA-Binding Proteins/deficiency , Disease Models, Animal , Gene Targeting , Genetic Engineering , Graft Survival , Host vs Graft Reaction , Humans , Killer Cells, Natural , Models, Animal , Swine , T-Lymphocytes/metabolism , Transplantation, Heterologous
11.
Radiol Imaging Cancer ; 2(6): e200058, 2020 11.
Article in English | MEDLINE | ID: mdl-33778750

ABSTRACT

Patients with cancer have been negatively impacted during the coronavirus disease 2019 (COVID-19) pandemic, as many of these individuals may be immunosuppressed and of older age. Additionally, cancer follow-up or imaging appointments have been delayed in many clinics around the world. Postponement of routine screening exams will result in delays in new cancer diagnoses. Clinics are continuing to monitor and adapt their appointment schedules based on local outbreaks of COVID-19. Studies on COVID-19 in patients with cancer are limited, but consistently indicate that this population is at risk for more severe COVID-19 illness. Data from recent studies also suggest that pediatric patients with cancer have a lower risk of severe COVID-19 illness compared to adults. Certain features of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection detected by lung, brain, and gastrointestinal imaging may confound radiologists' interpretation of cancer diagnosis, staging, and treatment response. Lastly, as clinics begin to re-open for routine appointments, protocols have been put in place to reduce SARS-CoV-2 exposure to patients during their visits. This review details different perspectives on the impact of the COVID-19 pandemic on patients with cancer and on cancer imaging. Keywords: Abdomen/GI, Cardiac, Infection, Nervous-Peripheral.


Subject(s)
COVID-19/complications , Diagnostic Imaging/methods , Neoplasms/complications , Neoplasms/diagnostic imaging , Patient Care/methods , Humans , Pandemics , SARS-CoV-2
14.
Front Oncol ; 9: 884, 2019.
Article in English | MEDLINE | ID: mdl-31572678

ABSTRACT

Prostate cancer (PCa) is the most common cancer in men, and the second leading cause of cancer related death in men in Western countries. The standard therapy for metastatic PCa is androgen suppression therapy (AST). Men undergoing AST eventually develop metastatic castration-resistant prostate cancer (mCRPC), of which there are limited treatment options available. Immunotherapy has presented substantial benefits for many types of cancer, but only a marginal benefit for mCRPC, at least in part, due to the immunosuppressive tumor microenvironment (TME). Current clinical trials are investigating monotherapies or combination therapies involving adoptive cellular therapy, viral, DNA vaccines, oncolytic viruses, and immune checkpoint inhibitors (ICI). Immunotherapies are also being combined with chemotherapy, radiation, and AST. Additionally, preclinical investigations show promise with the recent description of alternative ways to circumvent the immunosuppressive nature of the prostate tumor microenvironment, including harnessing the immune stimulatory NKG2D pathway, inhibiting myeloid derived suppressor cells, and utilizing immunomodulatory oncolytic viruses. Herein we provide an overview of recent preclinical and clinical developments in cancer immunotherapies and discuss the perspectives for future immunotherapies in PCa.

15.
Comp Med ; 69(2): 123-129, 2019 04 01.
Article in English | MEDLINE | ID: mdl-30755290

ABSTRACT

Swine are a commonly used animal model for biomedical research. One research application of swine models is the in utero injection of human or pig cells into the fetal liver (FL) or intraperitoneal space. In utero injections can be accomplished through laparotomy procedures in pregnant swine. In this study, we aimed to establish comprehensive laparotomy protocols for ultrasound-guided injections into fetuses. Two pregnant gilts, with a total of 16 fetuses, underwent laparotomy at 41 and 42 d of gestation. During surgery, we attempted to inject half of the fetuses in the FL or intraperitoneal space with saline and titanium wire for radiographic imaging after birth. After the laparotomy and fetal injections, both gilts maintained pregnancy throughout gestation and initiated labor at full term. Of the 16 fetuses present at the time of laparotomy, 12 were liveborn, 2 were stillborn, and the remaining 2 were mummies. A total of 7 fetuses from the 2 litters were known to have been injected with a wire during the surgery. After farrowing, piglets were radiographed, and 6 piglets were identified to have wire within the abdominal space. Livers were dissected, and additional radiographs were obtained. It was determined that one piglet had wire within the liver, whereas the other 5 had wire within the intraperitoneal space. Overall, we describe in-depth laparotomy surgery protocols, ultrasound-guided injection of saline and titanium wire into the FL or intraperitoneal space, postoperative monitoring protocols, and information on radiographic detection of titanium wire after piglet birth. These protocols can be followed by other research groups intending to inject cells of interest into either the intraperitoneal space or FL of fetal piglets.


Subject(s)
Fetal Therapies/veterinary , Injections, Intraperitoneal/veterinary , Laparotomy/methods , Animals , Female , Liver/diagnostic imaging , Models, Animal , Pregnancy , Swine , Ultrasonography, Interventional
16.
Front Oncol ; 9: 9, 2019.
Article in English | MEDLINE | ID: mdl-30723704

ABSTRACT

Ovarian cancer (OvCa) is the most lethal gynecologic malignancy, with two-thirds of patients having late-stage disease (II-IV) at diagnosis. Improved diagnosis and therapies are needed, yet preclinical animal models for ovarian cancer research have primarily been restricted to rodents, for data on which can fail to translate to the clinic. Thus, there is currently a need for a large animal OvCa model. Therefore, we sought to determine if pigs, being more similar to humans in terms of anatomy and physiology, would be a viable preclinical animal model for OvCa. We injected human OSPC-ARK1 cells, a chemotherapy-resistant primary ovarian serous papillary carcinoma cell line, into the neck muscle and ear tissue of four severe combined immune deficient (SCID) and two non-SCID pigs housed in novel biocontainment facilities to study the ability of human OvCa cells to form tumors in a xenotransplantation model. Tumors developed in ear tissue of three SCID pigs, while two SCID pigs developed tumors in neck tissue; no tumors were detected in non-SCID control pigs. All tumor masses were confirmed microscopically as ovarian carcinomas. The carcinomas in SCID pigs were morphologically similar to the original ovarian carcinoma and had the same immunohistochemical phenotype based on expression of Claudin 3, Claudin 4, Cytokeratin 7, p16, and EMA. Confirmation that OSPC-ARK1 cells form carcinomas in SCID pigs substantiates further development of orthotopic models of OvCa in pigs.

17.
Xenotransplantation ; 26(2): e12466, 2019 03.
Article in English | MEDLINE | ID: mdl-30311702

ABSTRACT

BACKGROUND: Severe combined immunodeficient (SCID) pigs are an emerging animal model being developed for biomedical and regenerative medicine research. SCID pigs can successfully engraft human-induced pluripotent stem cells and cancer cell lines. The development of a humanized SCID pig through xenotransplantation of human hematopoietic stem cells (HSCs) would be a further demonstration of the value of such a large animal SCID model. Xenotransplantation success with HSCs into non-obese diabetic (NOD)-derived SCID mice is dependent on the ability of NOD mouse signal regulatory protein alpha (SIRPA) to bind human CD47, inducing higher phagocytic tolerance than other mouse strains. Therefore, we investigated whether porcine SIRPA binds human CD47 in the context of developing a humanized SCID pig. METHODS: Peripheral blood mononuclear cells (PBMCs) were collected from SCID and non-SCID pigs. Flow cytometry was used to assess whether porcine monocytes could bind to human CD47. Porcine monocytes were isolated from PBMCs and were subjected to phagocytosis assays with pig, human, and mouse red blood cell (RBC) targets. Blocking phagocytosis assays were performed by incubating human RBCs with anti-human CD47 blocking antibody B6H12, non-blocking antibody 2D3, and nonspecific IgG1 antibody and exposing to human or porcine monocytes. RESULTS: We found that porcine SIRPA binds to human CD47 in vitro by flow cytometric assays. Additionally, phagocytosis assays were performed, and we found that porcine monocytes phagocytose human and porcine RBCs at significantly lower levels than mouse RBCs. When human RBCs were preincubated with CD47 antibodies B6H12 or 2D3, phagocytosis was induced only after B6H12 incubation, indicating the lower phagocytic activity of porcine monocytes with human cells requires interaction between porcine SIRPA and human CD47. CONCLUSIONS: We have shown the first evidence that porcine monocytes can bind to human CD47 and are phagocytically tolerant to human cells, suggesting that porcine SCID models have the potential to support engraftment of human HSCs.


Subject(s)
CD47 Antigen/immunology , Hematopoietic Stem Cell Transplantation , Monocytes/immunology , Animals , Hematopoietic Stem Cell Transplantation/methods , Humans , Leukocytes, Mononuclear/immunology , Macrophages/immunology , Mice, Inbred NOD/immunology , Mice, SCID , Phagocytosis/immunology , Receptors, Immunologic/immunology , Swine , Transplantation, Heterologous/methods
18.
Front Oncol ; 8: 559, 2018.
Article in English | MEDLINE | ID: mdl-30560086

ABSTRACT

Within the last decade there have been several severe combined immunodeficient (SCID) pig models discovered or genetically engineered. The animals have mutations in ARTEMIS, IL2RG, or RAG1/2 genes, or combinations thereof, providing SCID pigs with NK cells, but deficient in T and B cells, or deficient in NK, T, and B cells for research studies. Biocontainment facilities and positive pressure isolators are developed to limit pathogen exposure and prolong the life of SCID pigs. Raising SCID pigs in such facilities allows for completion of long-term studies such as xenotransplantation of human cells. Ectopically injected human cancer cell lines develop into tumors in SCID pigs, thus providing a human-sized in vivo model for evaluating imaging methods to improve cancer detection and therapeutic research and development. Immunocompromised pigs have the potential to be immunologically humanized by xenotransplantation with human hematopoietic stem cells, peripheral blood leukocytes, or fetal tissue. These cells can be introduced through various routes including injection into fetal liver or the intraperitoneal (IP) space, or into piglets by intravenous, IP, and intraosseous administration. The development and maintenance of transplanted human immune cells would be initially (at least) dependent on immune signaling from swine cells. Compared to mice, swine share higher homology in immune related genes with humans. We hypothesize that the SCID pig may be able to support improved engraftment and differentiation of a wide range of human immune cells as compared to equivalent mouse models. Humanization of SCID pigs would thus provide a valuable model system for researchers to study interactions between human tumor and human immune cells. Additionally, as the SCID pig model is further developed, it may be possible to develop patient-derived xenograft models for individualized therapy and drug testing. We thus theorize that the individualized therapeutic approach would be significantly improved with a humanized SCID pig due to similarities in size, metabolism, and physiology. In all, porcine SCID models have significant potential as an excellent preclinical animal model for therapeutic testing.

19.
Lab Anim ; 52(4): 402-412, 2018 Aug.
Article in English | MEDLINE | ID: mdl-29325489

ABSTRACT

Severe combined immunodeficiency (SCID) is defined by the lack of an adaptive immune system. Mutations causing SCID are found naturally in humans, mice, horses, dogs, and recently in pigs, with the serendipitous discovery of the Iowa State University SCID pigs. As research models, SCID animals are naturally tolerant of xenotransplantation and offer valuable insight into research areas such as regenerative medicine, cancer therapy, as well as immune cell signaling mechanisms. Large-animal biomedical models, particularly pigs, are increasingly essential to advance the efficacy and safety of novel regenerative therapies on human disease. Thus, there is a need to create practical approaches to maintain hygienic severe immunocompromised porcine models for exploratory medical research. Such research often requires stable genetic lines for replication and survival of healthy SCID animals for months post-treatment. A further hurdle in the development of the ISU SCID pig as a biomedical model involved the establishment of facilities and protocols necessary to obtain clean SPF piglets from the conventional pig farm on which they were discovered. A colony of homozygous SCID boars and SPF carrier sows has been created and maintained through selective breeding, bone marrow transplants, innovative husbandry techniques, and the development of biocontainment facilities.


Subject(s)
Disease Models, Animal , Housing, Animal , Severe Combined Immunodeficiency , Specific Pathogen-Free Organisms , Swine , Animal Husbandry , Animals , Female , Male
20.
Front Immunol ; 8: 813, 2017.
Article in English | MEDLINE | ID: mdl-28747915

ABSTRACT

After the discovery of naturally occurring severe combined immunodeficiency (SCID) within a selection line of pigs at Iowa State University, we found two causative mutations in the Artemis gene: haplotype 12 (ART12) and haplotype 16 (ART16). Bone marrow transplants (BMTs) were performed to create genetically SCID and phenotypically immunocompetent breeding animals to establish a SCID colony for further characterization and research utilization. Of nine original BMT transfer recipients, only four achieved successful engraftment. At approximately 11 months of age, both animals homozygous for the ART16 mutation were diagnosed with T cell lymphoma. One of these ART16/ART16 recipients was a male who received a transplant from a female sibling; the tumors in this recipient consist primarily of Y chromosome-positive cells. The other ART16/ART16 animal also presented with leukemia in addition to T cell lymphoma, while one of the ART12/ART16 compound heterozygote recipients presented with a nephroblastoma at a similar age. Human Artemis SCID patients have reported cases of lymphoma associated with a "leaky" Artemis phenotype. The naturally occurring Artemis SCID pig offers a large animal model more similar to human SCID patients and may offer a naturally occurring cancer model and provides a valuable platform for therapy development.

SELECTION OF CITATIONS
SEARCH DETAIL
...