Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2009 Sep 25.
Published in final edited form as: Vet Comp Oncol. 2007 Mar;5(1):14–30. doi: 10.1111/j.1476-5829.2006.00114.x

Five decades of progress in hematopoietic cell transplantation based on the preclinical canine model

Marilena Lupu 1,2, Rainer Storb 1,3
PMCID: PMC2752055  NIHMSID: NIHMS66946  PMID: 19754798

Abstract

The preclinical canine model has proved valuable for the development of principles and techniques of hematopoietic cell transplantation (HCT) applicable to human patients. Studies in random-bred dogs concerning the impact of histocompatibility barriers on engraftment and graft-versus-host disease, the kinetics of immunological reconstitution, the efficacy of various pre-transplant conditioning regimens, post-transplantation immunosuppression protocols, treatment of malignant diseases, and graft-versus-tumor effects have advanced HCT from an investigational therapy with uncertain clinical benefit half a century ago to an important treatment choice for thousands of patients treated annually in transplantation centers worldwide. More recent preclinical canine studies have resulted in the clinical translation of nonmyeloablative, minimally invasive transplantation protocols that have extended allogeneic HCT to include older human patients with malignant and non-malignant, acquired or inherited hematological disorders, and those with comorbid conditions. Here we review the contributions of the canine model to modern HCT and describe the usefulness of HCT for the treatment of canine hematological disorders.

Keywords: dog, hematopoietic cell transplantation, hematological malignancies

The Beginnings of Hematopoietic Cell Transplantation (HCT)

The history of HCT began with attempts to allay the lethal effects of irradiation that were observed in the wake of the atomic bomb explosions toward the end of World War II. It was soon recognized that bone marrow was the most radiosensitive organ in the body, and that marrow transplantation could rescue radiation victims from the lethal effects of marrow aplasia. This discovery encouraged clinicians and scientists to explore more aggressive protocols in patients with life-threatening hematological malignancies and increase the intensity of cytotoxic therapies beyond levels that were marrow toxic in hopes of eradicating the underlying cancer; rescue from the otherwise lethal marrow aplasia would be accomplished by the intravenous infusion of bone marrow cells.

In 1957, Thomas and colleagues reported studies of marrow grafting in patients with leukemia,1 concluding after two years2 that marrow transplantation was for the most part unsuccessful, with almost all patients dying from either allograft failure or recurrent leukemia. The immunological mechanisms involved in graft rejection and graft-versus-host disease (GVHD) were not well understood at that time. Overall, the early clinical results were disappointing and raised the concern that the allogeneic barrier could never be overcome. Therefore, many clinicians abandoned the idea that marrow transplantation could be used for the treatment of hematological malignancies in human patients,3 even though graft-versus-leukemia effects had already been recognized.4

Preclinical Studies of HCT: Murine and Canine Models

The first studies aimed at elucidating the radiation protection phenomenon were conducted in inbred mice.516 Early research showed that the protection of lethally irradiated mice could be achieved not only by shielding the spleen or one femur with lead during the radiation,6 but also by the intra-peritoneal7 or intra-venous16 infusion of syngeneic spleen or bone marrow after irradiation. Jacobson and colleagues6 hypothesized that the mouse spleen contained a humoral factor capable of stimulating the regeneration of blood-forming tissue, thereby advancing the “humoral” theory of hematopoietic reconstitution after irradiation. As an alternative to the humoral hypothesis, Barnes and colleagues8 suggested that the active principle in the spleen and bone marrow suspensions might be living cells that would act at least temporarily as a tissue graft, advancing the “cellular” theory which received little support at that time. The cellular hypothesis received strong support by Main’s and Prehn’s observation9 in mice that marrow donor skin grafts survived indefinitely in allogenic marrow recipients. Subsequently, Trentin and colleagues5 showed that the skin graft tolerance was specific for the donor strain. In 1956, three independent groups1012 confirmed the cellular theory by showing marrow repopulation through donor cells using various blood genetic markers. Further studies in inbred mice addressed other fundamental rules of marrow transplantation biology, including the observations that the mechanisms of graft rejection and GVHD13 were controlled by genetic factors14 that were in turn governed by the major histocompatibility complex (MHC).15 However, in 1967, the experimental hematologist Dirk W. van Bekkum drew attention to the fact that the first clinical trials in human patients with hematological malignancies failed mainly because “the clinical applications were undertaken too soon, most of them before even the minimum of basic knowledge required to bridge the gap between mouse and patient had been obtained.”17

In the following years, scientists directed their efforts at identifying more suitable preclinical animal models. Dogs appeared especially useful owing to their random-bred nature, large body size, longer life span, wide genetic diversity, and well-mixed gene pool; they are the only mammalian species besides humans to possess these qualities.18 Further, short gestation time and large litter sizes allowed studies of donor-recipient combinations that were matched or mismatched for MHC antigens, which are known as the dog leukocyte antigen (DLA) system, simulating human leukocyte antigen (HLA) matched donor-recipient pairs. Early experiments in the canine model using DLA-matched donor-recipient pairs and postgrafting immunosuppression19,20 renewed a sense of confidence towards treating human patients with hematological diseases by allogeneic marrow transplantation. As a consequence, in late 1960s, the results of the preclinical canine HCT studies began to be translated to patients with aplastic anemia and hematological malignancies using HLA-matched sibling marrow donors.21,22

Not surprisingly, studies in canines have provided an excellent basis for most of the HCT principles and techniques23 that have been directly translated over the past decades to the clinical setting for a wide spectrum of human diseases.16 Dog models of total body irradiation (TBI),17,2431 chemical3235 and radioimmunological36,37 myeloablation, cell dose requirements,3841 in vivo4247 and in vitro4851 graft manipulation, engraftment and GVHD,5254 as well as graft-versus-tumor effects5557 have been essential in order to solve the existing clinical problems of human HCT.

DLA Typing

Early canine allogeneic marrow transplantation studies showed occasional long-term stable engraftment, even though most animals succumbed to problems with GVHD and graft rejection.58 From these experiments it was concluded that major efforts would need to be directed towards understanding and defining histocompatibility barriers between donors and recipients.

The use of canine species in the experimental HCT field involved a rigorous study of the DLA system.19,5968 In 1968, Epstein and colleagues59 described a serologic canine histocompatibility typing system and demonstrated that compatibility between littermate donors and recipients played a fundamental role in reducing the risks of both graft rejection and GVHD. Further, long-term donor engraftment was shown to occur in both unrelated and related recipients, which were matched either by serologic typing alone or by serology and mixed leukocyte culture non-reactivity.19,61 Thus, the dog was the first random-bred species in which the impact of in vitro donor-recipient matching on HCT outcomes was demonstrated.19

Antigenic canine histocompatibility polymorphisms were first studied by serological59,61 and cellular typing in mixed leukocyte culture systems.60,61 It was not until later that the term DLA (dog leukocyte antigen) was introduced, and with it, the recognition that the histocompatibility complex could be divided into class I and class II regions. Subsequently, understanding of the molecular organization of the DLA region provided tools for molecular histocompatibility typing, which was facilitated by identification of convenient microsatellite polymorphisms within class I and class II regions that were inherited in a Mendelian fashion.68 As a consequence, molecular assessment of DLA class I and class II microsatellite marker polymorphisms,62,63 combined with DLA class II DRB1 allele sequencing,65,67 enabled high resolution histocompatibility testing of canine families and rapid selection of DLA-identical donors.

Graft Collection

Initial canine HCT studies involved the use of bone marrow as the source of hematopoietic progenitor cells obtained by aspiration from the humeral and femoral bones.23 Marrow cells stored at −80° C in dimethyl sulfoxide were capable of recovering 80% of the hematopoietic colony forming units in vitro69 and induced hematopoietic reconstitution of lethally irradiated dogs.20,48

Subsequent experiments in the canine model showed that either autologous40,49,70 or allogeneic50,51,71,72 peripheral blood stem cells could be successfully used as substitutes for marrow grafts, leading to recovery of normal hematopoiesis after otherwise lethal marrow ablation. Quantitative studies of peripheral blood mononuclear cell (PBMC) grafting demonstrated that approximately ten-fold more PBMCs than marrow cells were necessary to ensure stable engraftment.39,40 To induce expansion and/or peripheralization of hematopoietic progenitors from the marrow space into the peripheral blood, various mobilizing agents, such as cyclophosphamide (Cy);42 recombinant cytokines, including recombinant canine (rc) granulocyte colony-stimulating factor (G-CSF),43,45 rc stem cell factor (SCF, c-kit ligand),43,45 rc granulocyte-macrophage colony-stimulating factor,44 or the recently described CXCR4 antagonist AMD3100,47 were evaluated in the canine model. In addition, to facilitate PBMC collection, numerous apheresis-based techniques have been adapted to canines.19,23,7278

Currently, over 90% of all autologous HCT protocols for adult human patients with hematological malignancies include the use of mobilized PBMC grafts as a source of stem cells because of higher cell yields, decreased procedural risks, faster engraftment, and shorter hospitalization compared to the use of marrow grafts. However, human patients with aplastic anemia79 and those given grafts from unrelated donors80 showed better outcomes after allogeneic marrow rather than PBMC grafts. Although the major disadvantage of allogeneic PBMC grafts was associated with a higher incidence of chronic GVHD,81 an advantage in survival was seen in human patients with advanced hematological cancers.82

Conditioning Regimens

Conditioning regimens given before HCT serve to suppress the recipient’s immune system for graft acceptance while eradicating the underlying disease. Extensive preclinical canine studies investigating various conditioning regimens of chemotherapy, TBI, monoclonal/polyclonal antibody administration, or combinations thereof have set the stage for successful HCT trials in human patients.

Chemotherapy

In 1957, Thomas and colleagues established that irradiation itself did not eliminate leukemia in human patients; it was postulated that eradication of underlying malignancy might by possible by addition of chemotherapy.1

Encouraging results of pre-transplant conditioning with Cy in mice and rats83 prompted investigation of high-dose Cy conditioning regimens in dogs and rhesus monkeys, in place of TBI.32,33,83 It has been shown that dogs given a single Cy dose of 100 mg/kg could be rescued from lethal immunosuppression by administration of either autologous33 or allogeneic32 marrow grafts from DLA-matched donors, leading to sustained engraftment and mixed donor-recipient hematopoietic chimerism. High dose Cy conditioning regimens were subsequently translated to human patients with aplastic anemia undergoing allogeneic HCT.79

Following studies with busulfan in the rat,83 pre-HCT conditioning with dymethyl myleran (DMM), an intravenously injectable homologue of busulfan known for its marked myelosuppressive activities, was extensively evaluated in both healthy dogs34,35 and in dogs with malignant lymphoma.84 In healthy dogs, DMM given at a dose of 7.5 mg/kg induced profound marrow aplasia which was reversed by autologous marrow grafting.34 In the allogeneic HCT setting, despite the achievement of a mixed chimeric state, the level of immunosuppression using DMM at a dose of 10 mg/kg as a single-agent led to successful engraftment in 50% of the dogs, while the reminder rejected their grafts and died from pancytopenia; however, more consistent engraftment was achieved when either antithymocyte serum (ATS), produced by immunization of rabbits with puppy thymocytes, or a combination of ATS and procarbazine was given in addition to DMM.35 In dogs with malignant lymphoma, DMM was less effective in inducing complete remissions compared to TBI conditioning at similar marrow toxic doses.84,85

Total body irradiation

Conditioning regimens using TBI have been extensively studied in both healthy17,2431,86 and tumor-bearing dogs56,57,85,8789 as part of engraftment, toxicity, and malignant disease treatment studies. It was recognized that bone marrow hematopoietic stem cell progenitors were very vulnerable and reacted uniformly to the harmful effects of irradiation. Therefore, for the treatment of lymphoid malignancies, TBI conditioning provided the advantage of eradicating non-cycling malignant cells as effectively as eradicating cycling cells.

Thomas and colleagues were the first to demonstrate prompt recovery of bone marrow and lymphoid tissues in lethally irradiated dogs given intravenous infusions of freshly isolated or cryopreserved autologous or allogeneic marrow cells.24,86,90 All dogs that were not given a marrow infusion after a TBI dose of 400 cGy died from complications of marrow failure.24

In the absence of post-grafting immunosuppression, stable allogenic DLA-identical littermate marrow grafting and full donor chimerism were consistently obtained only after conditioning with TBI doses higher than 900 cGy, delivered at a dose rate of 7 cGy/min.29 However, engraftment of DLA-identical littermate marrow was consistently achieved with a single dose of 450 cGy when the exposure rate was increased to 70 cGy/min.91 Several studies investigating the impacts of dose rate on engraftment and toxicity showed that the exposure rate and total dose were the most important parameters for acute toxicity associated with TBI. It was determined that the overall TBI dose tolerated by healthy dogs given autologous marrow grafts could be increased from 10 to 14 Gy by reducing the exposure rate from 10 to 5 cGy/min. Generally, acute toxicity correlated with the TBI dose rate, while chronic toxicity was related to the total TBI dose.26,27

In order to lower TBI-related toxicity rates seen in human patients, but to retain a strong anti-tumor effect and promote sustained engraftment, a new approach was investigated in the canine model by using higher TBI doses given in a fractionated fashion.30,31,91 This strategy was based on the principle that hematopoietic cells were less capable of undergoing DNA repair after multiple, fractionated TBI doses compared to other tissues that would thus be affected to a lesser extent. As a consequence, long-term complications were significantly less in dogs given fractionated versus single TBI doses. When total doses of 12 to 21 Gy were given in multiple fractions of 1.5 to 2 Gy at intervals of 3 to 6 hours, with dose rates ranging from 2 to 20 cGy/min, an advantage of fractionated doses regarding acute toxicities was noted only at the highest rate of 20 cGy/min.30

Other studies investigated the effect of post-TBI administration of hematopoietic growth factors on engraftment of DLA-identical littermate marrow grafts.92,93 Post-TBI administration of recombinant human G-CSF or rcSCF significantly accelerated recovery of neutrophils and monocytes without altering platelet recovery and without significantly increasing the risks of graft failure or GVHD; however, lymphocyte recovery was more rapid in G-CSF-treated dogs. Although growth factors did not improve engraftment of DLA-identical grafts, they slightly improved survival.

Sustained engraftment of DLA-nonidentical grafts was seen only after preparative regimens with higher TBI doses than those used in the DLA-identical HCT setting. TBI doses of 1500 to 1800 cGy were generally tolerated only when given in fractions.26,28,94,95 When a TBI dose of 1800 cGy was given in three fractions of 600 cGy delivered at 2.1 cGy/min at 48 hours intervals, the immunosuppression achieved was sufficient to allow sustained engraftment of unrelated DLA-incompatible marrow grafts.95 Furthermore, attainment of sustained engraftment of histoincompatible marrow grafts following infusion of donor buffy coat cells or lymphocytes19,96 was an important observation that promoted the use of PBMC grafts. Other manipulations, such as the addition of monoclonal antibodies to TBI,97100 have been also successful in assuring sustained engraftment of DLA-nonidentical grafts, as described below.

Monoclonal and polyclonal antibodies

The efforts to minimize TBI-related toxicities prompted the investigation of novel conditioning regimens in the canine model, including the use of monoclonal antibodies, polyclonal antibodies, or fusion peptides targeted towards specific T-cell populations or T-cell costimulatory pathways.

A beneficial effect in overcoming resistance to DLA-mismatched marrow grafts was noted in 50% of the dogs given anti-class II monoclonal antibodies in addition to TBI and post-grafting immunosuppression.97 In the same model, the administration of S5 monoclonal antibody directed against the CD44 antigen led to sustained engraftment in 75% of the transplanted dogs.100 Further canine studies explored the immunosuppressive effects of radio-labeled monoclonal antibodies, such as the 131I-labeled/S536 or bismuth-213-labeled anti-CD45101/anti-TCRαβ37 conjugates, capable of inducing lethal marrow suppression36 or selective T-cell ablation,37 respectively.

A targeted nonmyeloablative strategy focused on induction of T-cell anergy was tested in dogs given DLA-identical littermate marrow grafts after conditioning with CTLA4-Ig, a fusion peptide that blocked T-cell costimulation through the B7-CD28 signaling pathway, and an otherwise inadequate TBI dose of 100 cGy; when this approach was combined with post-grafting immunosuppression by MMF and CSP, stable mixed hematopoietic chimerism was achieved in 70% of the dogs.102 These results have encouraged further efforts in the preclinical canine model towards conditioning strategies that would further reduce the TBI dosing or even eliminate the need for TBI, thereby further reducing or even avoiding both short- and long-term side effects from radiation.

Immunosuppression after HCT to Prevent GVHD

The preclinical canine model has served to investigate various post-grafting immunosuppressive regimens consisting of single drugs or combinations of immunosuppressive agents;5254,102117 these therapies might suppress the immune system, either by affecting all immune reactive tissues in general or by specifically affecting only replicating immune reactive cells. Parameters such as the clinical effectiveness, engraftment, GVHD prevention, immune reconstitution, and undesirable side effects in long-term survivors have all been used to determine whether a given immunosuppressive regimen could be safely translated from the preclinical setting to clinical protocols.

The main factors that influenced the success in crossing the immunological barriers between donor and recipient were the degree of histocompatibility matching between donor and recipient, and the efficacy of pre-transplant conditioning and post-transplant immunosuppression regimens. In both dogs and humans, even when donors and recipients were rigorously MHC-matched, GVHD prevention remained a difficult goal; marrow grafting from MHC-identical donors carried the risk of acute and/or chronic GVHD, despite the administration of post-grafting immunosuppression.52,103 It was first recognized in dogs that GVHD was due to sensitization of donor T lymphocytes not only to disparate major, but also minor histocompatibility antigens of the recipients.28 Once activated, lymphocytes attacked host tissues, predominantly the hematopoietic system, skin, gut, and liver.52,118 Omission of immunosuppression after grafting has been associated with a high incidence of acute GVHD and adverse effects on survival.59

Studies in unrelated DLA-nonidentical dogs have resulted in valuable strategies for GVHD prevention53,54,103,106109,111,113 and treatment.104,105 In this model, in the absence of post-grafting immunosuppression, acute and rapidly fatal GVHD occurred within 2 weeks of myeloablative HCT.53 Significant delay in the onset of GVHD and prolongation of survival were obtained when postgrafting immunosuppression was given as single-agent therapy with methotrexate (MTX),103 cyclosporine (CSP),106 azathioprine,53 succinylacetone,108 FK-506 (tacrolimus),109 or as combinations of these agents.107,109,111,119 Based on canine studies, post-HCT immunosuppression regimens consisting of either MTX, CSP, or a brief course of MTX combined with either CSP or FK506, were successfully translated to human patients for GVHD prophylaxis; antithymocyte globulin (ATG) was used to treat acute GVHD once it was established.52,54 Furthermore, it was found that postgrafting immunosuppression could be discontinued generally after 3–6 months of treatment due to instauration of mutual graft-versus-host tolerance.

Clear evidence of therapeutic graft-versus-tumor effects1,56 mediated by allogeneic effector T-cells that destroy recipient’s tumor cells after recognizing and reacting to disparate tumor-associated antigens, prompted the exploration in the canine model of less aggressive, nonmyeloablative HCT regimens or so called “minitransplants”. These regimens relied solely upon host immunosuppression to facilitate stable mixed chimerism, prevention of both allograft rejection and GVHD, and eradication of hematological malignant disease via a graft-versus-tumor response, following sublethal, minimal-invasive TBI conditioning.102,110,115 Mycophenolate mofetil (MMF),117 a purine synthesis inhibitor, was used in combination with other immunosuppressive drugs,54 leading to reduced frequency and severity of acute allograft rejection and improved survival in dogs given either DLA-identical102,110,112 or unrelated DLA-nonidentical53,111 marrow grafts. In the DLA-identical model, pre-transplant conditioning with a nonmyeloablative dose of 200 cGy TBI and post-HCT immunosuppression with MMF and CSP induced stable mixed chimerism and prevented both allograft rejection and GVHD.110 This protocol has been successfully translated to human patients who were not eligible for conventional myeloablative transplantation due to advanced age or comorbidities.120

The Impact of Preceding Blood Transfusions on HCT Outcomes

Early canine HCT studies drew attention to the fact that transfusions given before marrow grafts might jeopardize engraftment; it was hypothesized that this phenomenon might be associated to sensitization of recipients to non-DLA-associated polymorphic minor histocompatibility antigens.121 It was furthermore shown that DLA-identical recipients that received either three whole blood transfusions from the marrow donor or nine whole blood unrelated transfusions before TBI conditioning rejected 100% and 40%, respectively, of the marrow grafts.28 These studies demonstrated that several minor polymorphic loci, which were undetected by the usual in vitro histocompatibility typing, were involved in the occurrence of transfusion-induced sensitization. It was almost two decades later when the sensitizing cells responsible for transfusion-associated graft rejection were identified as being dendritic cells contained in the transfusion product.122 These observations prompted the exploration of treatments designed to eliminate or inactivate the cells responsible for the induction of this phenomenon. The incidence of graft rejection was lessened by reducing antigen-presenting mononuclear cells through the use of buffy coat-poor blood transfusion products; transfusion-induced sensitization was successfully overcome by using a combination of an alkylating agent, procarbazine, and ATS as pre-HCT conditioning, or prevented by in vitro treatment of blood transfusions with ultraviolet light or 2000 cGy gamma radiation.28,123,124 These findings were then translated into the clinic, leading to improved management of the multiply-transfused patients with aplastic anemia or other nonmalignant diseases who were candidates for marrow transplantation.125127

Hematopoietic Reconstitution and Side Effects after HCT

Hematopoietic reconstitution

Following myeloablative HCT, granulocyte counts achieved normal levels approximately by days 12; during the early post-irradiation period, dogs might require whole blood or platelet transfusions. However, following nonmyelablative HCT, life threatening declines of peripheral blood cell counts generally did not occur.110 Although dogs with successful engraftment were profoundly immunodeficient for 200 to 300 days after myeloablative HCT, long-term survivors recovered their immune function and were not susceptible to increased incidences of infection.128

Conditioning regimen-induced side effects

The main long-term side effects after high-dose TBI conditioning in dogs were pancreatic insufficiency and atrophy leading to maldigestion and malnutrition, keratitis, pneumonitis, change in coat color, cataracts, and sterility; in addition, a five-fold increased incidence of spontaneous carcinomas and sarcomas was reported. These findings were not seen in a smaller number of dogs conditioned with either Cy or busulfan.129

Acute side effects were associated to Cy administration, including anorexia, hematuria, vomiting, and diarrhea. Based on long-term surveillance for more than ten years after HCT, canine recipients conditioned with Cy regained fertility and sired normal litters.130

Side effects induced by immunosuppression after HCT

The limiting toxicity of MTX in dogs was gastrointestinal, as evidenced by diarrhea and vomiting; however, mouth ulceration or so-called mucositis, which is a major side effect in human patients, was rarely seen in dogs. The side effects associated to MMF administration in dogs were gastrointestinal, consisting mainly of diarrhea.131

The administration of calcineurin inhibitors in dogs was also associated with gastrointestinal side effects, including an increased incidence of intussusception. CSP also caused liver and kidney function changes, although these appeared to be less common in dogs than in humans at therapeutic levels. Following long-term CSP administration, dogs exhibited problems with papilloma infection; in addition, dogs presented changes in the skin and gums, as well as increased hair growth and blood pressure, all of which were reversible upon discontinuation of CSP administration.132 In order to reduce toxicities, CSP blood levels should be monitored and the dosing adjusted to keep levels within the therapeutic range.

HCT for Canine Malignant Diseases

Another advantage of the dog model consisted of the availability of animals with spontaneous malignant diseases, resembling closely those found in humans. In 1974, Weiden and colleagues established that dogs with malignant lymphoma had significant impairments of the humoral and/or cellular immune reactivity, similar to those seen in human patients.133 In addition, it was shown that canine malignant lymphoma was responsive to combination chemotherapy.134 Many other malignant conditions of the dog have well recognized counterparts in humans.135 These observations heightened the value of the canine cancer model in the investigational setting of HCT, including graft-versus-tumor effects, in a randomly bred species.

The attempts to cure spontaneous canine malignancies by marrow transplantation were pioneered by the Seattle marrow transplantation group.5557,84,85,8789,136. The dogs involved into those studies were referred by practicing veterinarians with the consent of the dogs’ owners and the survivors returned to their owners at the completion of the studies. Autologous grafts were given experimentally in dogs with malignant lymphoma,57,84,85,8789,136 leukemia,85 and solid tumors;85 the latter included mammary gland, adenocarcinoma, squamous cell carcinoma, undifferentiated carcinoma, mastocytoma, leiomyosarcoma, osteosarcoma, and melanoma. These early preclinical trials investigated different conditioning regimens consisting of either TBI,85 chemotherapy,84 or combinations thereof.57,8789,136 Studies showed that 25% of the dogs with malignant lymphoma in chemotherapy-induced remission given high doses of TBI, and either autologous cryopreserved marrow57,85,87 or freshly isolated PBMC136 grafts, became disease-free long-term survivors; however, the solid tumor-bearing dogs showed little antitumor responses, consistent with the known greater radioresistance of solid non-hematological tumors.85

Three important allogeneic marrow transplantation studies were carried out by the Seattle transplantation group in dogs with malignant lymphoma,5557 leukemia,56 and miscellaneous solid tumors.55,56 The graft-versus-lymphoma effect of allogeneic grafts was demonstrated by prolonged disease-free survivals (p < 0.005) after allogeneic compared to autologous HCT using similar conditioning regimens.56 However, allogeneic grafts were complicated by high rates of fatal GVHD55 and toxicities related to postgrafting immunosuppression.57 No suggestion of significant graft-versus-tumor effects in dogs with non-hematological solid tumors was obtained, and all transplanted dogs died with evidence of persistent tumors.56

Overall, it was established that the mortality rate in dogs with hematological malignancies given either autologous or allogeneic HCT was adversely affected by a compromised clinical status at the time of transplantation.56,85 From these studies it was predicted that HCT could be more effective if performed in patients not only with minimal tumor burden, but before deterioration of their general condition; clinical translation of these observations resulted in improved transplantation outcomes.137

HCT for Canine Nonmalignant Diseases

The therapeutic potential of allogeneic DLA-identical HCT has been investigated for a wide range of canine non-malignant hematological and non-hematological disorders, such as congenital immunological and enzymatic deficiencies, and Duchenne muscular dystrophy.

Allogeneic marrow grafts from DLA-identical littermates corrected cyclic neutropenia (Grey Collie disease)138 and X-linked severe combined immunodeficiency139 with full reconstitution of neutrophil, and T and B cell functions, respectively. Allogeneic HCT studies were also performed in Basenji dogs with hemolytic anemia due to pyruvate kinase deficiency; these studies demonstrated reversal of iron overload after successful transplantation140 and determined the level of donor chimerism needed to prevent hemolysis.141,142 Using DLA-identical marrow grafts for the treatment of lysosomal storage diseases, such as α-L-iduronidase deficiency,143 mucopolysaccharidosis I,144 and fucosidase deficiency,145 the disease-related pathologies were partially corrected. However, the outcomes in dogs with ceroid lipofuscinosis (Batten’s disease),146 GM1 gangliosidosis,147 hemophilia,148 and Duchenne muscular dystrophy149 were not improved following allogeneic HCT. For the treatment of Duchenne muscular dystrophy, stable hematopoietic chimerism induced tolerance in the immune system of recipients to both dystrophin and cells from HCT donors, thereby setting the stage for ongoing investigations of additional therapeutic strategies such as gene and muscle stem cell therapies.

HCT as a Therapy for Pet Dogs with Hematological Malignancies

Cancer is one of the leading causes of death in dogs, showing a one-log higher incidence than in humans.150 Among canine cancers, hematological malignancies are rarely curable by radio/chemotherapy.151 Although more than 90% response rates were achievable after various combination chemotherapy regimens,152 remissions were short due to the development of multi-drug resistance and recurrence of disseminated disease.151 Other therapeutic alternatives for canine malignant lymphoma, such as the use of high dose half body irradiation given either alone153 or after preceding remission inductions with chemotherapy,154 led to measurable decreases in tumors size; however, besides radiation-induced side effects, the median remission durations were not significantly prolonged compared to those achieved following conventional combination chemotherapy.154

The preclinical research developed in dogs and summarized in this review has the potential for improving cancer therapies in pet dogs. Hematopoietic stem cell transplantation can cure canine malignant hematological conditions that have been considered fatal.57,87,136,155,156 Molecular DLA typing methods offer a high-resolution technology for donor selection among single- and multi-generation canine families,63,65,156 which is a pre-requisite for lower GVHD rates and improved transplantation outcomes. The canine genome map18,64,157 has contributed to identification of new microsatellite repeat sequences that have extended the panel of markers for evaluation of donor chimerism.158 The availability of safe PBMC mobilizing agents and apheresis devices for dogs offers a minimally invasive alternative for graft collection compared to the traditional bone marrow harvest. Less toxic nonmyeloablative HCT regimens have been recently developed in the canine model, which permit graft-versus-tumor effects while controlling GVHD, and which could be applied to veterinary patients.

The continuous advances in veterinary medical oncology, radiation oncology, and transfusion medicine, the expansion of canine blood bank networks, and rapid progress in canine immunogenetics, all promise to improve HCT-based therapies for canine patients with cancer. In the clinical veterinary field, the treatment of canine malignant lymphoma in chemotherapy-induced remission by autologous marrow transplantation has already been reported.155 We have recently described the successful treatment of a pet dog with malignant T-cell lymphoma in chemotherapy-induced remission by TBI and allogeneic rcG-CSF-mobilized PBMC transplantation.156

From Past to Present and Future

As a consequence of the numerous experimental studies in random-bred dogs, HCT has advanced from an investigational therapy that was declared without perspective in the 1960s, to an important treatment option that produces amazing clinical results. In 1990, Dr. Donnall E. Thomas was awarded the Nobel Prize in Medicine for his pioneering work in marrow transplantation. He emphasized that “marrow grafting could not have reached clinical application without animal research, first in inbred rodents and then in outbred species, particularly the dog.”159

The efforts continue in the preclinical canine model to improve the HCT regimens for direct translation to human patients. Research has been directed towards designing specific antibodies against costimulatory molecules with key roles in the development of the immunological reactions after allogeneic HCT.160 Potential antileukemic effects of donor-derived natural killer cells161 are currently explored in the nonmyeloablative HCT setting. The observation that in some human patients graft-versus-tumor effects are associated with GVHD, whereas in others remission can be achieved without GVHD, has led to investigation of strategies to separate graft-versus-tumor effects from GVHD through adoptive transfer of selected T cell populations.162 The use of adoptive immunotherapy could make allogeneic HCT more effective to recognize tumor-specific antigens163 and perhaps extend its application to the treatment of metastatic and non-hematopoietic tumors that arise in organs such as the breast, prostate, pancreas, or colon. In this regard, dogs with spontaneous tumors could become ideal candidates for preclinical trials of anti-tumor vaccination. Furthermore, research involving the recently recognized immune regulatory T cells that arise after HCT could lead to a better understanding of mechanisms of graft rejection and GVHD, and improve strategies to overcome these immunological barriers.162 Promising preclinical results of kidney grafting in a canine mixed hematopoietic chimera model164 suggest that HCT may serve in the future as an immunologic platform for solid organ grafting without the need for lifelong immunosuppression. Finally, the approach of transferring cloned genes into the hematopoietic stem cells to correct specific genetic defects may be a promising avenue for the therapy of genetic diseases.165

Not only in human history but also in the history of medicine, the dog has been loyal to human beings. It is now well recognized in the HCT field that the dog has contributed to a legacy that saves thousands of patients annually, proving once again the paradigm of “man’s best friend”. The remarkable progress in the clinical applications of preclinical HCT protocols established in canines, and the rapid development of the biological sciences predict sustained advances over the coming years in both the human and veterinary oncology fields.

Acknowledgment

M.L. was supported by a fellowship in Oncology and Transplantation Biology from National Institutes of Health, Bethesda, MD.

References

  • 1.Thomas ED, Lochte HL, Jr, Lu WC, Ferrebee JW. Intravenous infusion of bone marrow in patients receiving radiation and chemotherapy. New England Journal of Medicine. 1957;257:491–496. doi: 10.1056/NEJM195709122571102. [DOI] [PubMed] [Google Scholar]
  • 2.Thomas ED, Lochte HL, Jr, Cannon JH, Sahler OD, Ferrebee JW. Supralethal whole body irradiation and isologous marrow transplantation in man. Journal of Clinical Investigation. 1959;38:1709–1716. doi: 10.1172/JCI103949. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Bortin MM. A compendium of reported human bone marrow transplants. Transplantation. 1970;9:571–587. doi: 10.1097/00007890-197006000-00006. [DOI] [PubMed] [Google Scholar]
  • 4.Barnes DWH, Loutit JF. Treatment of murine leukaemia with x-rays and homologous bone marrow: II. British Journal of Haematology. 1957;3:241–252. doi: 10.1111/j.1365-2141.1957.tb05793.x. [DOI] [PubMed] [Google Scholar]
  • 5.Trentin JJ. Mortality and skin transplantability in x-irradiated mice receiving isologous or heterologous bone marrow. Proceedings of the Society for Experimental Biology and Medicine. 1956;92:688–693. doi: 10.3181/00379727-92-22582. [DOI] [PubMed] [Google Scholar]
  • 6.Jacobson LO, Marks EK, Robson MJ, Gaston EO, Zirkle RE. Effect of spleen protection on mortality following x-irradiation. Journal of Laboratory and Clinical Medicine. 1949;34:1538–1543. [Google Scholar]
  • 7.Jacobson LO, Simmons EL, Marks EK, Eldredge JH. Recovery from radiation injury. Science. 1951;113:510–511. doi: 10.1126/science.113.2940.510. [DOI] [PubMed] [Google Scholar]
  • 8.Barnes DWH, Loutit JF. What is the recovery factor in spleen [Letter]? Nucleonics. 1954;12:68–71. [Google Scholar]
  • 9.Main JM, Prehn RT. Successful skin homografts after the administration of high dosage X radiation and homologous bone marrow. Journal of the National Cancer Institute. 1955;15:1023–1029. [PubMed] [Google Scholar]
  • 10.Ford CE, Hamerton JL, Barnes DWH, Loutit JF. Cytological identification of radiation-chimaeras. Nature. 1956;177:452–454. doi: 10.1038/177452a0. [DOI] [PubMed] [Google Scholar]
  • 11.Nowell PC, Cole LJ, Habermeyer JG, Roan PL. Growth and continued function of rat marrow cells in x-radiated mice. Cancer Research. 1956;16:258–261. [PubMed] [Google Scholar]
  • 12.Vos O, Davids JAG, Weyzen WWH, van Bekkum DW. Evidence for cellular hypothesis in radiation protection by bone marrow cells. Acta Physiologica et Pharmacologica Neerlandica. 1956;4:482–486. [PubMed] [Google Scholar]
  • 13.Barnes DWH, Corp MJ, Loutit JF, Neal FE. Treatment of murine leukaemia with x-rays and homologous bone marrow. Preliminary communication. British Medical Journal. 1956;2:626–627. doi: 10.1136/bmj.2.4993.626. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Uphoff DE. Genetic factors influencing irradiation protection by bone marrow. I. The F1 hybrid effect. Journal of the National Cancer Institute. 1957;19:123–125. [PubMed] [Google Scholar]
  • 15.Snell GD. The Nobel Lectures in Immunology. Lecture for the Nobel Prize for Physiology or Medicine, 1980: Studies in histocompatibility. (Review) Scandinavian Journal of Immunology. 1992;36:513–526. doi: 10.1111/j.1365-3083.1992.tb03218.x. [DOI] [PubMed] [Google Scholar]
  • 16.Blume KG, Forman SJ, Appelbaum FR, editors. Thomas' Hematopoietic Cell Transplantation. Third edn. Oxford, UK: Blackwell Publishing Ltd.; 2004. [Google Scholar]
  • 17.van Bekkum DW, de Vries MJ. Radiation chimaeras. London: Logos Press Limited; 1967. [Google Scholar]
  • 18.Ostrander EA, Wayne RK. The canine genome (Review) Genome Research. 2005;15:1706–1716. doi: 10.1101/gr.3736605. [DOI] [PubMed] [Google Scholar]
  • 19.Storb R, Epstein RB, Bryant J, Ragde H, Thomas ED. Marrow grafts by combined marrow and leukocyte infusions in unrelated dogs selected by histocompatibility typing. Transplantation. 1968;6:587–593. doi: 10.1097/00007890-196807000-00011. [DOI] [PubMed] [Google Scholar]
  • 20.Storb R, Epstein RB, LeBlond RF, Rudolph RH, Thomas ED. Transplantation of allogeneic canine bone marrow stored at −80OC in dimethyl sulfoxide. (Brief report) Blood. 1969;33:918–923. [PubMed] [Google Scholar]
  • 21.Thomas ED, Buckner CD, Rudolph RH, Fefer A, Storb R, Neiman PE, Bryant JI, Chard RL, Clift RA, Epstein RB, Fialkow PJ, Funk DD, Giblett ER, Lerner KG, Reynolds FA, Slichter S. Allogeneic marrow grafting for hematologic malignancy using HL-A matched donor-recipient sibling pairs. Blood. 1971;38:267–287. [PubMed] [Google Scholar]
  • 22.Thomas ED, Storb R, Clift RA, Fefer A, Johnson FL, Neiman PE, Lerner KG, Glucksberg H, Buckner CD. Bone-marrow transplantation. New England Journal of Medicine. 1975;292:832–843. 895–902. doi: 10.1056/NEJM197504172921605. [DOI] [PubMed] [Google Scholar]
  • 23.Deeg HJ, Storb R. Bone marrow transplantation in dogs. In: Makowka L, Cramer DV, Podesta LG, editors. Handbook of Animal Models in Transplantation Research. Boca Raton, Florida: CRC Press, Inc.; 1994. pp. 255–285. [Google Scholar]
  • 24.Thomas ED, LeBlond R, Graham T, Storb R. Marrow infusions in dogs given midlethal or lethal irradiation. Radiation Research. 1970;41:113–124. [PubMed] [Google Scholar]
  • 25.Bull MI, Herzig GP, Graw RG, Jr, Krueger GR, Bowles CA. Canine allogeneic bone marrow transplantation. Technique and variables influencing engraftment. Transplantation. 1976;22:150–159. doi: 10.1097/00007890-197608000-00010. [DOI] [PubMed] [Google Scholar]
  • 26.Kolb HJ, Rieder I, Bodenberger U, Netzel B, Schäffer E, Kolb H, Thierfelder S. Dose rate and dose fractionation studies in total body irradiation of dogs. Pathologie Biologie. 1979;27:370–372. [PubMed] [Google Scholar]
  • 27.Deeg HJ, Storb R, Weiden PL, Schumacher D, Shulman H, Graham T, Thomas ED. High-dose total-body irradiation and autologous marrow reconstitution in dogs: Dose-rate-related acute toxicity and fractionation-dependent long-term survival. Radiation Research. 1981;88:385–391. [PubMed] [Google Scholar]
  • 28.Storb R, Deeg HJ. Failure of allogeneic canine marrow grafts after total body irradiation: Allogeneic “resistance” vs transfusion induced sensitization. Transplantation. 1986;42:571–580. doi: 10.1097/00007890-198612000-00001. [DOI] [PubMed] [Google Scholar]
  • 29.Storb R, Raff RF, Appelbaum FR, Schuening FW, Sandmaier BM, Graham TC, Thomas ED. What radiation dose for DLA-identical canine marrow grafts? Blood. 1988;72:1300–1304. [PubMed] [Google Scholar]
  • 30.Deeg HJ, Storb R, Longton G, Graham TC, Shulman HM, Appelbaum F, Thomas ED. Single dose or fractionated total body irradiation and autologous marrow transplantation in dogs: Effects of exposure rate, fraction size and fractionation interval on acute and delayed toxicity. International Journal of Radiation Oncology, Biology, Physics. 1988;15:647–653. doi: 10.1016/0360-3016(88)90307-0. [DOI] [PubMed] [Google Scholar]
  • 31.Storb R, Raff RF, Appelbaum FR, Graham TC, Schuening FG, Sale G, Pepe M. Comparison of fractionated to single-dose total body irradiation in conditioning canine littermates for DLA-identical marrow grafts. Blood. 1989;74:1139–1143. [PubMed] [Google Scholar]
  • 32.Storb R, Epstein RB, Rudolph RH, Thomas ED. Allogeneic canine bone marrow transplantation following cyclophosphamide. Transplantation. 1969;7:378–386. doi: 10.1097/00007890-196905000-00007. [DOI] [PubMed] [Google Scholar]
  • 33.Epstein RB, Storb R, Clift RA, Thomas ED. Autologous bone marrow grafts in dogs treated with lethal doses of cyclophosphamide. Cancer Research. 1969;29:1072–1075. [PubMed] [Google Scholar]
  • 34.Kolb HJ, Storb R, Weiden PL, Ochs HD, Kolb H, Graham TC, Floersheim GL, Thomas ED. Immunologic, toxicologic and marrow transplantation studies in dogs given dimethyl myleran. Biomedicine. 1974;20:341–351. [PubMed] [Google Scholar]
  • 35.Storb R, Weiden PL, Graham TC, Lerner KG, Nelson N, Thomas ED. Hemopoietic grafts between DLA-identical canine littermates following dimethyl myleran. Evidence for resistance to grafts not associated with DLA and abrogated by antithymocyte serum. Transplantation. 1977;24:349–357. doi: 10.1097/00007890-197711000-00006. [DOI] [PubMed] [Google Scholar]
  • 36.Appelbaum FR, Brown P, Sandmaier B, Badger C, Schuening F, Graham TC, Storb R. Antibody-radionuclide conjugates as part of a myeloblative preparative regimen for marrow transplantation. Blood. 1989;73:2202–2208. [PubMed] [Google Scholar]
  • 37.Bethge WA, Wilbur DS, Storb R, Hamlin DK, Santos EB, Brechbiel MW, Fisher DR, Sandmaier BM. Selective T-cell ablation with bismuth-213-labeled anti-TCRαβ as nonmyeloablative conditioning for allogeneic canine marrow transplantation. Blood. 2003;101:5068–5075. doi: 10.1182/blood-2002-12-3867. [DOI] [PubMed] [Google Scholar]
  • 38.Sullivan RD, Stecher G, Sternberg SS. Value of bone marrow and spleen cell suspensions for survival of lethally irradiated dogs. Journal of the National Cancer Institute. 1959;23:367–383. [PubMed] [Google Scholar]
  • 39.Appelbaum FR, Herzig GP, Graw RG, Bull MI, Bowles C, Gorin NC, Deisseroth AB. Study of cell dose and storage time on engraftment of cryopreserved autologous bone marrow in a canine model. Transplantation. 1978;26:245–248. doi: 10.1097/00007890-197810000-00008. [DOI] [PubMed] [Google Scholar]
  • 40.Appelbaum FR. Hemopoietic reconstitution following autologous bone marrow and peripheral blood mononuclear cell infusions. Experimental Hematology. 1979;7(Suppl5):7–11. [PubMed] [Google Scholar]
  • 41.Bodenberger U, Kolb HJ, Rieder I, Netzel B, Schäffer E, Kolb H, Thierfelder S. Fractionated total body irradiation and autologous bone marrow transplantation in dogs: hemopoietic recovery after various marrow cell doses. Experimental Hematology. 1980;8:384–394. [PubMed] [Google Scholar]
  • 42.Abrams RA, McCormack K, Bowles C, Deisseroth AB. Cyclophosphamide treatment expands the circulating hematopoietic stem cell pool in dogs. Journal of Clinical Investigation. 1981;67:1392–1399. doi: 10.1172/JCI110167. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.de Revel T, Appelbaum FR, Storb R, Schuening F, Nash R, Deeg J, McNiece I, Andrews R, Graham T. Effects of granulocyte colony stimulating factor and stem cell factor, alone and in combination, on the mobilization of peripheral blood cells that engraft lethally irradiated dogs. Blood. 1994;83:3795–3799. [PubMed] [Google Scholar]
  • 44.Nash RA, Schuening FG, Seidel K, Appelbaum FR, Boone T, Deeg HJ, Graham TC, Hackman R, Sullivan-Pepe M, Storb R. Effect of recombinant canine granulocyte-macrophage colony-stimulating factor on hematopoietic recovery after otherwise lethal total body irradiation. Blood. 1994;83:1963–1970. [PubMed] [Google Scholar]
  • 45.Sandmaier BM, Storb R, Santos EB, Krizanac-Bengez L, Lian T, McSweeney PA, Yu C, Schuening FG, Deeg HJ, Graham T. Allogeneic transplants of canine peripheral blood stem cells mobilized by recombinant canine hematopoietic growth factors. Blood. 1996;87:3508–3513. [PubMed] [Google Scholar]
  • 46.Yunusov MY, Georges GE, Storb R, Moore P, Hagglund H, Affolter V, Lesnikova M, Gass MJ, Little M-T, Loken M, McKenna H, Storer B, Nash RA. FLT3 ligand promotes engraftment of allogeneic hematopoietic stem cells without significant graft-versus-host disease. Transplantation. 2003;75:933–940. doi: 10.1097/01.TP.0000057831.93385.7D. [DOI] [PubMed] [Google Scholar]
  • 47.Burroughs L, Mielcarek M, Little M-T, Bridger G, MacFarland R, Fricker S, LaBrecque J, Sandmaier BM, Storb R. Durable engraftment of AMD3100-mobilized autologous and allogeneic peripheral blood mononuclear cells in a canine transplantation model. Blood. 2005;106:4002–4008. doi: 10.1182/blood-2005-05-1937. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Cavins JA, Kasakura S, Thomas ED, Ferrebee JW. Recovery of lethally irradiated dogs following infusion of autologous marrow stored at low temperature in dimethyl-sulphoxide. Blood. 1962;20:730–734. [PubMed] [Google Scholar]
  • 49.Cavins JA, Scheer SC, Thomas ED, Ferrebee JW. The recovery of lethally irradiated dogs given infusions of autologous leukocytes preserved at −80 C. Blood. 1964;23:38–43. [PubMed] [Google Scholar]
  • 50.Körbling M, Fliedner TM, Calvo W, Ross WM, Nothdurft W, Steinbach I. Albumin density gradient purification of canine hemopoietic blood stem cells (HBSC): long-term allogeneic engraftment without GVH-reaction. Experimental Hematology. 1979;7:277–288. [PubMed] [Google Scholar]
  • 51.Gerhartz HH, Nothdurft W, Carbonell F, Fliedner TM. Allogeneic transplantation of blood stem cells concentrated by density gradients. Experimental Hematology. 1985;13:136–142. [PubMed] [Google Scholar]
  • 52.Storb R, Thomas ED. Graft-versus-host disease in dog and man: the Seattle experience (Review) Immunological Reviews. 1985;88:215–238. doi: 10.1111/j.1600-065x.1985.tb01160.x. [DOI] [PubMed] [Google Scholar]
  • 53.Storb R, Kolb HJ, Deeg HJ, Weiden PL, Appelbaum F, Graham TC, Thomas ED. Prevention of graft-versus-host disease by immunosuppressive agents after transplantation of DLA-nonidentical canine marrow. Bone Marrow Transplantation. 1986;1:167–177. [PubMed] [Google Scholar]
  • 54.Storb R, Deeg HJ, Raff R, Schuening F, Yu C, Sandmaier BM, Graham T. Prevention of graft-versus-host disease: Studies in a canine model (Review) Annals of the New York Academy of Sciences. 1995;770:149–164. doi: 10.1111/j.1749-6632.1995.tb31052.x. [DOI] [PubMed] [Google Scholar]
  • 55.Epstein RB, Graham TC, Storb R, Thomas ED. Studies of marrow transplantation, chemotherapy and cross-circulation in canine lymphosarcoma. Blood. 1971;37:349–359. [PubMed] [Google Scholar]
  • 56.Weiden PL, Storb R, Sale GE, Graham TC, Thomas ED. Allogeneic hematopoietic grafts after total-body irradiation in dogs with spontaneous tumors. Journal of the National Cancer Institute. 1978;61:353–357. [PubMed] [Google Scholar]
  • 57.Appelbaum FR, Deeg HJ, Storb R, Self S, Graham TC, Sale GE, Weiden PL. Marrow transplant studies in dogs with malignant lymphoma. Transplantation. 1985;39:499–504. doi: 10.1097/00007890-198505000-00008. [DOI] [PubMed] [Google Scholar]
  • 58.Thomas ED, Collins JA, Herman EC, Jr, Ferrebee JW. Marrow transplants in lethally irradiated dogs given methotrexate. Blood. 1962;19:217–228. [PubMed] [Google Scholar]
  • 59.Epstein RB, Storb R, Ragde H, Thomas ED. Cytotoxic typing antisera for marrow grafting in littermate dogs. Transplantation. 1968;6:45–58. doi: 10.1097/00007890-196801000-00005. [DOI] [PubMed] [Google Scholar]
  • 60.Rudolph RH, Hered B, Epstein RB, Thomas ED. Canine mixed leukocyte reactivity and transplantation antigens. Transplantation. 1969;8:141–146. doi: 10.1097/00007890-196908000-00006. [DOI] [PubMed] [Google Scholar]
  • 61.Storb R, Rudolph RH, Thomas ED. Marrow grafts between canine siblings matched by serotyping and mixed leukocyte culture. Journal of Clinical Investigation. 1971;50:1272–1275. doi: 10.1172/JCI106605. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Burnett RC, Francisco LV, DeRose SA, Storb R, Ostrander EA. Identification and characterization of a highly polymorphic microsatellite marker within the canine MHC class I region. Mammalian Genome. 1995;6:684–685. doi: 10.1007/BF00352386. [DOI] [PubMed] [Google Scholar]
  • 63.Wagner JL, Burnett RC, DeRose SA, Francisco LV, Storb R, Ostrander EA. Histocompatibility testing of dog families with highly polymorphic microsatellite markers. Transplantation. 1996;62:876–877. doi: 10.1097/00007890-199609270-00032. [DOI] [PubMed] [Google Scholar]
  • 64.Francisco LV, Langston AA, Mellersh CS, Neal CL, Ostrander EA. A class of highly polymorphic tetranucleotide repeats for canine genetic mapping. Mammalian Genome. 1996;7:359–362. doi: 10.1007/s003359900104. [DOI] [PubMed] [Google Scholar]
  • 65.Wagner JL, Works JD, Storb R. DLA-DRB1 and DLA-DQB1 histocompatibility typing by PCR-SSCP and sequencing (Brief Communication) Tissue Antigens. 1998;52:397–401. doi: 10.1111/j.1399-0039.1998.tb03063.x. [DOI] [PubMed] [Google Scholar]
  • 66.Graumann MB, DeRose SA, Ostrander EA, Storb R. Polymorphism analysis of four canine MHC class I genes. Tissue Antigens. 1998;51:374–381. doi: 10.1111/j.1399-0039.1998.tb02976.x. [DOI] [PubMed] [Google Scholar]
  • 67.Wagner JL, Burnett RC, Works JD, Storb R. Molecular analysis of DLA-DRBB1 polymorphism. Tissue Antigens. 1996;48:554–561. doi: 10.1111/j.1399-0039.1996.tb02669.x. [DOI] [PubMed] [Google Scholar]
  • 68.Wagner JL, Sarmiento UM, Storb R. Cellular, serological, and molecular polymorphism of the class I and class II loci of the canine major histocompatibility complex. Tissue Antigens. 2002;59:205–210. doi: 10.1034/j.1399-0039.2002.590304.x. [DOI] [PubMed] [Google Scholar]
  • 69.Debelak-Fehir KM, Catchatourian R, Epstein RB. Hemopoietic colony forming units in fresh and cryopreserved peripheral blood cells of canines and man. Experimental Hematology. 1975;3:109–116. [PubMed] [Google Scholar]
  • 70.Storb R, Epstein RB, Thomas ED. Marrow repopulating ability of peripheral blood cells compared to thoracic duct cells. Blood. 1968;32:662–667. [PubMed] [Google Scholar]
  • 71.Epstein RB, Graham TC, Buckner CD, Bryant J, Thomas ED. Allogeneic marrow engraftment by cross circulation in lethally irradiated dogs. Blood. 1966;28:692–707. [PubMed] [Google Scholar]
  • 72.Storb R, Epstein RB, Ragde H, Thomas ED. Marrow engraftment by allogeneic leukocytes in lethally irradiated dogs. Blood. 1967;30:805–811. [PubMed] [Google Scholar]
  • 73.Buckner D, Eisel R, Perry S. Blood cell separation in the dog by continuous flow centrifugation. Blood. 1968;31:653–672. [PubMed] [Google Scholar]
  • 74.Epstein RB, Clift RA, Thomas ED. The effect of leukocyte transfusions on experimental bacteremia in the dog. Blood. 1969;34:782–790. [PubMed] [Google Scholar]
  • 75.Herbst EW, Fliedner TM, Calvo W, Schnappauf H, Meyer H. Collection of hematopoietic stem cells from canine peripheral blood and their ability to regenerate hematopoiesis [German] Blut. 1975;30:265–276. doi: 10.1007/BF01633820. [DOI] [PubMed] [Google Scholar]
  • 76.Zander AR, Gray KN, Ben-Ze'ev A, Johnston DA, Spitzer G, Raulston GL, McCredie KB, Jardine JH, Wu J, Gleiser C, Cardiff J, Dicke KA. Rescue by peripheral blood mononuclear cells in dogs from bone marrow failure after total-body irradiation. Transfusion. 1984;24:42–45. doi: 10.1046/j.1537-2995.1984.24184122560.x. [DOI] [PubMed] [Google Scholar]
  • 77.Ladiges WC, Storb R, Graham T, Thomas ED. Experimental techniques used to study the immune system of dogs and other large animals. In: Gay WI, Heavener JE, editors. Methods of Animal Experimentation. New York, NY: Academic Press; 1989. pp. 103–133. [Google Scholar]
  • 78.Lee R, Storb R, Little M-T, Joslyn A, Spector M, Kuhr CS. Percutaneous central dual-lumen catheter for apheresis in the canine. Journal of Investigative Surgery. 2002;15:337–341. doi: 10.1080/08941930290086155. [DOI] [PubMed] [Google Scholar]
  • 79.Storb R, Prentice RL, Thomas ED. Marrow transplantation for treatment of aplastic anemia. An analysis of factors associated with graft rejection. New England Journal of Medicine. 1977;296:61–66. doi: 10.1056/NEJM197701132960201. [DOI] [PubMed] [Google Scholar]
  • 80.Ringden O, Remberger M, Runde V, Bornhauser M, Blau IW, Basara N, Holig K, Beelen DW, Hagglund H, Basu O, Ehninger G, Fauser AA. Peripheral blood stem cell transplantation from unrelated donors: a comparison with marrow transplantation. Blood. 1999;94:455–464. [PubMed] [Google Scholar]
  • 81.MacDonald KP, Rowe V, Filippich C, Johnson D, Morris ES, Clouston AD, Ferrara JL, Hill GR. Chronic graft-versus-host disease after granulocyte colony-stimulating factor-mobilized allogeneic stem cell transplantation: the role of donor T-cell dose and differentiation. Biology of Blood and Marrow Transplantation. 2004;10:373–385. doi: 10.1016/j.bbmt.2004.02.002. [DOI] [PubMed] [Google Scholar]
  • 82.Bensinger WI, Martin PJ, Storer B, Clift R, Forman SJ, Negrin R, Kashyap A, Flowers MED, Lilleby K, Chauncey TR, Storb R, Appelbaum FR. Transplantation of bone marrow as compared with peripheral-blood cells from HLA-identical relatives in patients with hematologic cancers. New England Journal of Medicine. 2001;344:175–181. doi: 10.1056/NEJM200101183440303. [DOI] [PubMed] [Google Scholar]
  • 83.Thomas ED. High-dose therapy and bone marrow transplantation. Seminars in Oncology. 1986;12:15–20. [PubMed] [Google Scholar]
  • 84.Weiden PL, Storb R, Shulman H, Graham TC. Dimethyl myleran and autologous marrow grafting for the treatment of spontaneous canine lymphoma. European Journal of Cancer and Clinical Oncology. 1977;13:1411–1415. doi: 10.1016/0014-2964(77)90154-2. [DOI] [PubMed] [Google Scholar]
  • 85.Weiden PL, Storb R, Lerner KG, Kao GF, Graham TC, Thomas ED. Treatment of canine malignancies by 1200 R total body irradiation and autologous marrow grafts. Experimental Hematology. 1975;3:124–134. [PubMed] [Google Scholar]
  • 86.Ferrebee JW, Lochte HL, Jr, Jaretzki A, III, Sahler OD, Thomas ED. Successful marrow homograft in the dog after radiation. Surgery. 1958;43:516–520. [PubMed] [Google Scholar]
  • 87.Weiden PL, Storb R, Deeg HJ, Graham TC, Thomas ED. Prolonged disease-free survival in dogs with lymphoma after total-body irradiation and autologous marrow transplantation consolidation of combination-chemotherapy-induced remissions. Blood. 1979;54:1039–1049. [PubMed] [Google Scholar]
  • 88.Weiden PL, Storb R, Deeg HJ, Graham TC. Total body irradiation and autologous marrow transplantation as consolidation therapy for spontaneous canine lymphoma in remission. Experimental Hematology. 1979;7(Suppl5):160–163. [PubMed] [Google Scholar]
  • 89.Deeg HJ, Appelbaum FR, Weiden PL, Hackman RC, Graham TC, Storb R. Autologous marrow transplantation as consolidation therapy for canine lymphoma: Efficacy and toxicity of various regimens of total body irradiation. American Journal of Veterinary Research. 1985;46:2016–2018. [PubMed] [Google Scholar]
  • 90.Mannick JA, Lochte HL, Jr, Ashley CA, Thomas ED, Ferrebee JW. Autografts of bone marrow in dogs after lethal total-body radiation. Blood. 1960;15:255–266. [PubMed] [Google Scholar]
  • 91.Storb R, Raff RF, Graham T, Appelbaum FR, Deeg HJ, Schuening FG, Shulman H, Pepe M. Marrow toxicity of fractionated versus single dose total body irradiation is identical in a canine model. International Journal of Radiation Oncology, Biology, Physics. 1993;26:275–283. doi: 10.1016/0360-3016(93)90207-c. [DOI] [PubMed] [Google Scholar]
  • 92.Schuening FG, Storb R, Goehle S, Graham TC, Hackman R, Mori M, Sousa LM, Appelbaum FR. Recombinant human granulocyte colony-stimulating factor accelerates hematopoietic recovery after DLA-identical littermate marrow transplants in dogs. Blood. 1990;76:636–640. [PubMed] [Google Scholar]
  • 93.Schuening FG, von Kalle C, Kiem H-P, Appelbaum FR, Deeg HJ, Pepe M, Gooley T, Graham TC, Hackman R, Storb R. Effect of recombinant canine stem cell factor, a c-kit ligand, on hematopoietic recovery after DLA-identical littermate marrow transplants in dogs. Experimental Hematology. 1997;25:1240–1245. [PubMed] [Google Scholar]
  • 94.Vriesendorp HM, Klapwijk WM, van Kessel AMC, Zurcher C, van Bekkum DW. Lasting engraftment of histoincompatible bone marrow cells in dogs. Transplantation. 1981;31:347–352. doi: 10.1097/00007890-198105010-00009. [DOI] [PubMed] [Google Scholar]
  • 95.Deeg HJ, Storb R, Shulman HM, Weiden PL, Graham TC, Thomas ED. Engraftment of DLA-nonidentical unrelated canine marrow after high-dose fractionated total body irradiation. Transplantation. 1982;33:443–446. doi: 10.1097/00007890-198204000-00021. [DOI] [PubMed] [Google Scholar]
  • 96.Deeg HJ, Storb R, Weiden PL, Shulman HM, Graham TC, Torok-Storb BJ, Thomas ED. Abrogation of resistance to and enhancement of DLA-nonidentical unrelated marrow grafts in lethally irradiated dogs by thoracic duct lymphocytes. Blood. 1979;53:552–557. [PubMed] [Google Scholar]
  • 97.Deeg HJ, Sale GE, Storb R, Graham TC, Schuening F, Appelbaum FR, Thomas ED. Engraftment of DLA-nonidentical bone marrow facilitated by recipient treatment with anti-class II monoclonal antibody and methotrexate. Transplantation. 1987;44:340–345. doi: 10.1097/00007890-198709000-00003. [DOI] [PubMed] [Google Scholar]
  • 98.Schuening F, Storb R, Goehle S, Meyer J, Graham TC, Deeg HJ, Appelbaum FR, Sale GE, Graf L, Loughran TP., Jr Facilitation of engraftment of DLA-nonidentical marrow by treatment of recipients with monoclonal antibody directed against marrow cells surviving radiation. Transplantation. 1987;44:607–613. doi: 10.1097/00007890-198711000-00004. [DOI] [PubMed] [Google Scholar]
  • 99.Sandmaier BM, Storb R, Appelbaum FR, Gallatin WM. An antibody that facilitates hematopoietic engraftment recognizes CD44. Blood. 1990;76:630–635. [PubMed] [Google Scholar]
  • 100.Sandmaier BM, Storb R, Bennett KL, Appelbaum FR, Santos EB. Epitope specificity of CD44 for monoclonal antibody dependent facilitation of marrow engraftment in a canine model. Blood. 1998;91:3494–3502. [PubMed] [Google Scholar]
  • 101.Sandmaier BM, Bethge WA, Wilbur DS, Hamlin DK, Santos EB, Brechbiel MW, Fisher DR, Storb R. Bismuth 213-labeled anti-CD45 radioimmunoconjugate to condition dogs for nonmyeloablative allogeneic marrow grafts. Blood. 2002;100:318–326. doi: 10.1182/blood-2001-12-0322. [DOI] [PubMed] [Google Scholar]
  • 102.Storb R, Yu C, Zaucha JM, Deeg HJ, Georges G, Kiem H-P, Nash RA, McSweeney PA, Wagner JL. Stable mixed hematopoietic chimerism in dogs given donor antigen, CTLA4Ig, and 100 cGy total body irradiation before and pharmacologic immunosuppression after marrow transplant. Blood. 1999;94:2523–2529. [PubMed] [Google Scholar]
  • 103.Storb R, Epstein RB, Graham TC, Thomas ED. Methotrexate regimens for control of graft-versus-host disease in dogs with allogeneic marrow grafts. Transplantation. 1970;9:240–246. doi: 10.1097/00007890-197003000-00007. [DOI] [PubMed] [Google Scholar]
  • 104.Kolb HJ, Storb R, Graham TC, Kolb H, Thomas ED. Antithymocyte serum and methotrexate for control of graft-versus-host disease in dogs. Transplantation. 1973;16:17–23. doi: 10.1097/00007890-197307000-00004. [DOI] [PubMed] [Google Scholar]
  • 105.Storb R, Kolb HJ, Graham TC, Kolb H, Weiden PL, Thomas ED. Treatment of established graft-versus-host disease in dogs by antithymocyte serum or prednisone. Blood. 1973;42:601–609. [PubMed] [Google Scholar]
  • 106.Deeg HJ, Storb R, Weiden PL, Graham T, Atkinson K, Thomas ED. Cyclosporin-A: effect on marrow engraftment and graft-versus-host disease in dogs. Transplantation Proceedings. 1981;13:402–409. [PubMed] [Google Scholar]
  • 107.Deeg HJ, Storb R, Weiden PL, Raff RF, Sale GE, Atkinson K, Graham TC, Thomas ED. Cyclosporin A and methotrexate in canine marrow transplantation: engraftment, graft-versus-host disease, and induction of tolerance. Transplantation. 1982;34:30–35. doi: 10.1097/00007890-198207000-00006. [DOI] [PubMed] [Google Scholar]
  • 108.Raff RF, Storb R, Graham T, Fidler JM, Sale GE, Johnston B, Deeg HJ, Pepe M, Schuening F, Appelbaum FR, Bauer RJ, Young JD, Marafino BJ, Ando DG, Braude IA. Pharmacologic, toxicologic and marrow transplantation studies in dogs given succinyl acetone. Transplantation. 1992;54:813–820. doi: 10.1097/00007890-199211000-00009. [DOI] [PubMed] [Google Scholar]
  • 109.Storb R, Raff RF, Appelbaum FR, Deeg HJ, Fitzsimmons W, Graham TC, Pepe M, Pettinger M, Sale G, Van Der Jagt R, Schuening FG. FK506 and methotrexate prevent graft-versus-host disease in dogs given 9.2 Gy total body irradiation and marrow grafts from unrelated DLA-nonidentical donors. Transplantation. 1993;56:800–807. doi: 10.1097/00007890-199310000-00005. [DOI] [PubMed] [Google Scholar]
  • 110.Storb R, Yu C, Wagner JL, Deeg HJ, Nash RA, Kiem H-P, Leisenring W, Shulman H. Stable mixed hematopoietic chimerism in DLA-identical littermate dogs given sublethal total body irradiation before and pharmacological immunosuppression after marrow transplantation. Blood. 1997;89:3048–3054. [PubMed] [Google Scholar]
  • 111.Yu C, Seidel K, Nash RA, Deeg HJ, Sandmaier BM, Barsoukov A, Santos E, Storb R. Synergism between mycophenolate mofetil and cyclosporine in preventing graft-versus-host disease among lethally irradiated dogs given DLA-nonidentical unrelated marrow grafts. Blood. 1998;91:2581–2587. [PubMed] [Google Scholar]
  • 112.Storb R, Yu C, Barnett T, Wagner JL, Deeg HJ, Nash RA, Kiem H-P, McSweeney P, Seidel K, Georges G, Zaucha JM. Stable mixed hematopoietic chimerism in dog leukocyte antigen-identical littermate dogs given lymph node irradiation before and pharmacologic immunosuppression after marrow transplantation. Blood. 1999;94:1131–1136. [PubMed] [Google Scholar]
  • 113.Yu C, Linsley P, Seidel K, Sale G, Deeg HJ, Nash RA, Storb R. Cytotoxic T lymphocyte antigen 4-immunoglobulin fusion protein combined with methotrexate/cyclosporine as graft-versus-host disease prevention in a canine dog leukocyte antigen-nonidentical marrow transplant model. Transplantation. 2000;69:450–454. doi: 10.1097/00007890-200002150-00027. [DOI] [PubMed] [Google Scholar]
  • 114.Lee RS, Kuhr CS, Sale GE, Zellmer E, Hogan WJ, Storb R, Little M-T. FTY720 does not abrogate acute graft-versus-host disease in the DLA-nonidentical unrelated canine model. Transplantation. 2003;76:1155–1158. doi: 10.1097/01.TP.0000083891.14089.B8. [DOI] [PubMed] [Google Scholar]
  • 115.Hogan WJ, Little M-T, Zellmer E, Friedetzky A, Diaconescu R, Gisburne S, Lee R, Kuhr C, Storb R. Postgrafting immunosuppression with sirolimus and cyclosporine facilitates stable mixed hematopoietic chimerism in dogs given sublethal total body irradiation before marrow transplantation from DLA-identical littermates. Biology of Blood and Marrow Transplantation. 2003;9:489–495. doi: 10.1016/s1083-8791(03)00148-4. [DOI] [PubMed] [Google Scholar]
  • 116.Kuhr CS, Lupu M, Little M-T, Zellmer E, Sale GE, Storb R. RDP58 does not prevent graft-versus-host disease after dog leukocyte antigen-nonidentical canine hematopoietic cell transplantation. Transplantation. 2006;81:1460–1462. doi: 10.1097/01.tp.0000203323.82681.7d. [DOI] [PubMed] [Google Scholar]
  • 117.Lupu M, McCune JS, Kuhr CS, Gooley T, Storb R. Pharmacokinetics of oral mycophenolate mofetil in dog: bioavailability studies and the impact of antibiotic therapy (Letter to the Editor) Biology of Blood and Marrow Transplantation. doi: 10.1016/j.bbmt.2006.07.009. (in press) [DOI] [PubMed] [Google Scholar]
  • 118.Kolb H, Sale GE, Lerner KG, Storb R, Thomas ED. Pathology of acute graft-versus-host disease in the dog. An autopsy study of ninety-five dogs. American Journal of Pathology. 1979;96:581–594. [PMC free article] [PubMed] [Google Scholar]
  • 119.Yu C, Storb R, Deeg HJ, Graham TC, Schuening FG, Huss R, Seidel K, Fitzsimmons WE. Tacrolimus (FK506) and methotrexate regimens to prevent graft-versus-host disease after unrelated dog leukocyte antigen (DLA) nonidentical marrow transplantation. Bone Marrow Transplantation. 1996;17:649–653. [PubMed] [Google Scholar]
  • 120.McSweeney PA, Niederwieser D, Shizuru JA, Sandmaier BM, Molina AJ, Maloney DG, Chauncey TR, Gooley TA, Hegenbart U, Nash RA, Radich J, Wagner JL, Minor S, Appelbaum FR, Bensinger WI, Bryant E, Flowers MED, Georges GE, Grumet FC, Kiem H-P, Torok-Storb B, Yu C, Blume KG, Storb RF. Hematopoietic cell transplantation in older patients with hematologic malignancies: replacing high-dose cytotoxic therapy with graft-versus-tumor effects. Blood. 2001;97:3390–3400. doi: 10.1182/blood.v97.11.3390. [DOI] [PubMed] [Google Scholar]
  • 121.Storb R, Epstein RB, Rudolph RH, Thomas ED. The effect of prior transfusion on marrow grafts between histocompatible canine siblings. Journal of Immunology. 1970;105:627–633. [PubMed] [Google Scholar]
  • 122.Deeg HJ, Aprile J, Storb R, Graham TC, Hackman R, Appelbaum FR, Schuening F. Functional dendritic cells are required for transfusion-induced sensitization in canine marrow graft recipients. Concise Report. Blood. 1988;71:1138–1140. [PubMed] [Google Scholar]
  • 123.Deeg HJ, Aprile J, Graham TC, Appelbaum FR, Storb R. Ultraviolet irradiation of blood prevents transfusion-induced sensitization and marrow graft rejection in dogs. Concise Report. Blood. 1986;67:537–539. [PubMed] [Google Scholar]
  • 124.Bean MA, Storb R, Graham T, Raff R, Sale GE, Schuening F, Appelbaum FR. Prevention of transfusion-induced sensitization to minor histocompatibility antigens on DLA-identical canine marrow grafts by gamma irradiation of marrow donor blood. Transplantation. 1991;52:956–960. doi: 10.1097/00007890-199112000-00004. [DOI] [PubMed] [Google Scholar]
  • 125.Storb R, Thomas ED . for the Seattle Marrow Transplant Team. Marrow transplantation for treatment of aplastic anaemia. In: Thomas ED, editor. Clinics in Haematology. London: W.B.Saunders Company Ltd.; 1978. pp. 597–609. [PubMed] [Google Scholar]
  • 126.Storb R, Thomas ED, Buckner CD, Clift RA, Deeg HJ, Fefer A, Goodell BW, Sale GE, Sanders JE, Singer J, Stewart P, Weiden PL. Marrow transplantation in thirty “untransfused” patients with severe aplastic anemia. Annals of Internal Medicine. 1980;92:30–36. doi: 10.7326/0003-4819-92-1-30. [DOI] [PubMed] [Google Scholar]
  • 127.Storb R, Doney KC, Thomas ED, Appelbaum F, Buckner CD, Clift RA, Deeg HJ, Goodell BW, Hackman R, Hansen JA, Sanders J, Sullivan K, Weiden PL, Witherspoon RP. Marrow transplantation with or without donor buffy coat cells for 65 transfused aplastic anemia patients. Blood. 1982;59:236–246. [PubMed] [Google Scholar]
  • 128.Ochs HD, Storb R, Thomas ED, Kolb H-J, Graham TC, Mickelson E, Parr M, Rudolph RH. Immunologic reactivity in canine marrow graft recipients. Journal of Immunology. 1974;113:1039–1057. [Google Scholar]
  • 129.Deeg HJ, Prentice R, Fritz TE, Sale GE, Lombard LS, Thomas ED, Storb R. Increased incidence of malignant tumors in dogs after total body irradiation and marrow transplantation. International Journal of Radiation Oncology, Biology, Physics. 1983;9:1505–1511. doi: 10.1016/0360-3016(83)90325-5. [DOI] [PubMed] [Google Scholar]
  • 130.Hager EB, Thomas ED, Ferrebee JW. Fertility of dogs after recovery from “lethal” exposure to radiation. Radiobiologia Radiotherapia. 1962;3:1–3. [PubMed] [Google Scholar]
  • 131.Chanda SM, Sellin JH, Torres CM, Yee JP. Comparative gastrointestinal effects of mycophenolate mofetil capsules and enteric-coated tablets of sodium-mycophenolic acid in beagle dogs. Transplantation Proceedings. 2002;34:3387–3392. doi: 10.1016/s0041-1345(02)03601-1. [DOI] [PubMed] [Google Scholar]
  • 132.Ladiges WC, Storb R, Thomas ED. Canine models of bone marrow transplantation. Laboratory Animal Science. 1990;40:11–15. [PubMed] [Google Scholar]
  • 133.Weiden PL, Storb R, Kolb H-J, Ochs HD, Graham TC, Tsoi M-S, Schroeder M-L, Thomas ED. Immune reactivity in dogs with spontaneous malignancy. Journal of the National Cancer Institute. 1974;53:1049–1056. doi: 10.1093/jnci/53.4.1049. [DOI] [PubMed] [Google Scholar]
  • 134.Madewell BR. Chemotherapy for canine lymphosarcoma. American Journal of Veterinary Research. 1975;36:1525–1528. [PubMed] [Google Scholar]
  • 135.Owen LN, Bostock DE, Betton GR, Onions DE, Holmes J, Yoxall A, Gorman N. The role of spontaneous canine tumours in the evaluation of the aetiology and therapy of human cancer. Journal of Small Animal Practice. 1975;16:155–162. doi: 10.1111/j.1748-5827.1975.tb05729.x. [DOI] [PubMed] [Google Scholar]
  • 136.Appelbaum FR, Deeg HJ, Storb R, Graham TC, Charrier K, Bensinger W. Cure of malignant lymphoma in dogs with peripheral blood stem cell transplantation. Transplantation. 1986;42:19–22. doi: 10.1097/00007890-198607000-00004. [DOI] [PubMed] [Google Scholar]
  • 137.Thomas ED, Buckner CD, Banaji M, Clift RA, Fefer A, Flournoy N, Goodell BW, Hickman RO, Lerner KG, Neiman PE, Sale GE, Sanders JE, Singer J, Stevens M, Storb R, Weiden PL. One hundred patients with acute leukemia treated by chemotherapy, total body irradiation, and allogeneic marrow transplantation. Blood. 1977;49:511–533. [PubMed] [Google Scholar]
  • 138.Weiden P, Robinett B, Graham TC, Adamson JW, Storb R. Canine cyclic neutropenia. A stem cell defect. Journal of Clinical Investigation. 1974;53:950–953. doi: 10.1172/JCI107636. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139.Felsburg PJ, Somberg RL, Hartnett BJ, Suter SF, Henthorn PS, Moore PF, Weinberg KI, Ochs HD. Full immunologic reconstitution following nonconditioned bone marrow transplantation for canine X-linked severe combined immunodeficiency. Blood. 1997;90:3214–3221. [PubMed] [Google Scholar]
  • 140.Weiden PL, Hackman RC, Deeg HJ, Graham TC, Thomas ED, Storb R. Long-term survival and reversal of iron overload after marrow transplantation in dogs with congenital hemolytic anemia. Blood. 1981;57:66–70. [PubMed] [Google Scholar]
  • 141.Zaucha JM, Yu C, Lothrop CD, Jr, Nash RA, Sale G, Georges G, Kiem H-P, Niemeyer GP, Dufresne M, Cao Q, Storb R. Severe canine hereditary hemolytic anemia treated by nonmyeloablative marrow transplantation. Biology of Blood and Marrow Transplantation. 2001;7:14–24. doi: 10.1053/bbmt.2001.v7.pm11215693. [DOI] [PubMed] [Google Scholar]
  • 142.Takatu A, Nash RA, Zaucha JM, Little M-T, Georges GE, Sale GE, Zellmer E, Kuhr CS, Lothrop CD, Jr, Storb R. Adoptive immunotherapy to increase the level of donor hematopoietic chimerism after nonmyeloablative marrow transplantation for severe canine hereditary hemolytic anemia. Biology of Blood and Marrow Transplantation. 2003;9:674–682. doi: 10.1016/j.bbmt.2003.08.005. [DOI] [PubMed] [Google Scholar]
  • 143.Shull RM, Breider MA, Constantopoulos GC. Long-term neurological effects of bone marrow transplantation in a canine lysosomal storage disease. Pediatric Research. 1988;24:347–352. doi: 10.1203/00006450-198809000-00015. [DOI] [PubMed] [Google Scholar]
  • 144.Breider MA, Shull RM, Constantopoulos G. Long-term effects of bone marrow transplantation in dogs with mucopolysaccharidosis I. American Journal of Pathology. 1989;134:677–692. [PMC free article] [PubMed] [Google Scholar]
  • 145.Taylor RM, Farrow BR, Stewart GJ. Amelioration of clinical disease following bone marrow transplantation in fucosidase-deficient dogs. American Journal of Medical Genetics. 1992;42:628–632. doi: 10.1002/ajmg.1320420439. [DOI] [PubMed] [Google Scholar]
  • 146.Deeg HJ, Shulman HM, Albrechtsen D, Graham TC, Storb R, Koppang N. Batten's disease: Failure of allogeneic bone marrow transplantation to arrest disease progression in a canine model. Clinical Genetics. 1990;37:264–270. doi: 10.1111/j.1399-0004.1990.tb04188.x. [DOI] [PubMed] [Google Scholar]
  • 147.O'Brien JS, Storb R, Raff RF, Harding J, Appelbaum F, Morimoto S, Kishimoto Y, Graham T, Ahern-Rindell A, O'Brien SL. Bone marrow transplantation in canine GM1 gangliosidosis. Clinical Genetics. 1990;38:274–280. doi: 10.1111/j.1399-0004.1990.tb03581.x. [DOI] [PubMed] [Google Scholar]
  • 148.Storb R, Marchioro TL, Graham TC, Willemin M, Hougie C, Thomas ED. Canine hemophilia and hemopoietic grafting. Blood. 1972;40:234–238. [PubMed] [Google Scholar]
  • 149.Dell'Agnola C, Wang Z, Storb R, Tapscott SJ, Kuhr CS, Hauschka SD, Lee RS, Sale GE, Zellmer E, Gisburne S, Bogan J, Kornegay JN, Cooper BJ, Gooley TA, Little M-T. Hematopoietic stem cell transplantation does not restore dystrophin expression in Duchenne muscular dystrophy dogs. Blood. 2004;104:4311–4318. doi: 10.1182/blood-2004-06-2247. [DOI] [PubMed] [Google Scholar]
  • 150.Hahn KA, Bravo L, Adams WH, Frazier DL. Naturally occurring tumors in dogs as comparative models for cancer therapy research (Review) In Vivo. 1994;8:133–143. [PubMed] [Google Scholar]
  • 151.Vail DM. Lymphoma. In: Feldman BF, Zinkl JG, Jain NC, editors. Schalm's Veterinary Hematology. 5th edn. Philadelphia, PA: Lippincott Williams & Wilkins; 2000. pp. 601–630. [Google Scholar]
  • 152.Vonderhaar MA, Morrison WB. Cancer in Dogs and Cats: Medical and Surgical Management. Baltimore, MD: Williams & Wilkins; 1998. Lymphosarcoma; pp. 667–695. [Google Scholar]
  • 153.Laing EJ, Fitzpatrick PJ, Binnington AG, Norris AM, Mosseri A, Rider WD, Valli VE, Baur A. Half-body radiotherapy in the treatment of canine lymphoma. Journal of Veterinary Internal Medicine. 1989;3:102–108. doi: 10.1111/j.1939-1676.1989.tb03087.x. [DOI] [PubMed] [Google Scholar]
  • 154.Williams LE, Johnson JL, Hauck ML, Ruslander DM, Price GS, Thrall DE. Chemotherapy followed by half-body radiation therapy for canine lymphoma. Journal of Veterinary Internal Medicine. 2004;18:703–709. doi: 10.1892/0891-6640(2004)18<703:cfbhrt>2.0.co;2. [DOI] [PubMed] [Google Scholar]
  • 155.Frimberger AE, Moore AS, Quesenberry PJ. Bone marrow transplantation-based experimental therapies for canine lymphoma patients. In: Modiano JF, editor. 1st Meeting on Genes, Dogs, and Cancer: Emerging Concepts in Molecular Diagnosis and Therapy, 2001 - Keystone, CO, USA. Ithaca, NY: International Veterinary Information Service; 2001. ( www.ivis.org), Document No. P0316.0501. [Google Scholar]
  • 156.Lupu M, Sullivan EW, Westfall TE, Little M-T, Weigler BJ, Moore PF, Stroup PA, Zellmer E, Kuhr C, Storb R. Use of multigeneration-family molecular dog leukocyte antigen typing to select a hematopoietic cell transplant donor for a dog with T-cell lymphoma. Journal of the American Veterinary Medical Association. 2006;228:728–732. doi: 10.2460/javma.228.5.728. [DOI] [PubMed] [Google Scholar]
  • 157.Mellersh CS, Langston AA, Acland GM, Fleming MA, Ray K, Wiegand NA, Francisco LV, Gibbs M, Aguirre GD, Ostrander EA. A linkage map of the canine genome. Genomics. 1997;46:326–336. doi: 10.1006/geno.1997.5098. [DOI] [PubMed] [Google Scholar]
  • 158.Yu C, Ostrander E, Bryant E, Burnett R, Storb R. Use of (CA)n polymorphisms to determine the origin of blood cells after allogeneic canine marrow grafting. Transplantation. 1994;58:701–706. [PubMed] [Google Scholar]
  • 159.Thomas ED. Bone marrow transplantation - past, present and future: Nobel lecture, December 8, 1990. In: Frängsmyr T, editor. Les Prix Nobel: The Nobel Prizes 1990. Stockholm, Sweden: Nobel Foundation; 1990. pp. 222–230. [Google Scholar]
  • 160.Lee J-H, Joo Y-D, Yim D, Lee R, Ostrander EA, Loretz C, Little M-T, Storb R, Kuhr CS. Molecular cloning and characterization of canine ICOS. Genomics. 2004;84:730–736. doi: 10.1016/j.ygeno.2004.06.009. [DOI] [PubMed] [Google Scholar]
  • 161.Ruggeri L, Capanni M, Casucci M, Volpi I, Tosti A, Perruccio K, Urbani E, Negrin RS, Martelli MF, Velardi A. Role of natural killer cell alloreactivity in HLA-mismatched hematopoietic stem cell transplantation. Blood. 1999;94:333–339. [PubMed] [Google Scholar]
  • 162.Fowler DH, Gress RE. Th2 and Tc2 cells in the regulation of GVHD, GVL, and graft rejection: considerations for the allogeneic transplantation therapy of leukemia and lymphoma [Review] Leukemia and Lymphoma. 2000;38:221–234. doi: 10.3109/10428190009087014. [DOI] [PubMed] [Google Scholar]
  • 163.Appelbaum FR. Haematopoietic cell transplantation as immunotherapy. Nature. 2001;411:385–389. doi: 10.1038/35077251. [DOI] [PubMed] [Google Scholar]
  • 164.Kuhr CS, Allen MD, Junghanss C, Zaucha JM, Marsh CL, Yunusov M, Zellmer E, Little M-T, Torok-Storb B, Storb R. Tolerance to vascularized kidney grafts in canine mixed hematopoietic chimeras. Transplantation. 2002;73:1487–1493. doi: 10.1097/00007890-200205150-00020. [DOI] [PubMed] [Google Scholar]
  • 165.Trobridge G, Beard BC, Kiem H-P. Hematopoietic stem cell transduction and amplification in large animal models. Human Gene Therapy. 2005;16:1355–1366. doi: 10.1089/hum.2005.16.1355. [DOI] [PubMed] [Google Scholar]

RESOURCES