Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2011 Apr 1.
Published in final edited form as: Cytokine Growth Factor Rev. 2010 Mar 7;21(2-3):169–175. doi: 10.1016/j.cytogfr.2010.02.010

Oncolytic viral purging of leukemic hematopoietic stem and progenitor cells with Myxoma virus

Masmudur M Rahman 1, Gerard J Madlambayan 2, Christopher R Cogle 2, Grant McFadden 1,*
PMCID: PMC2881168  NIHMSID: NIHMS181384  PMID: 20211576

Abstract

High-dose chemotherapy and radiation followed by autologous blood and marrow transplantation (ABMT) has been extensively used for the treatment of certain cancers that are refractory to standard therapeutic regimes. However, a major challenge with ABMT for patients with hematologic malignancies is disease relapse, mainly due to either contamination with cancerous hematopoietic stem and progenitor cells (HSPCs) within the autograft or the persistence of residual therapy-resistant disease niches within the patient. Oncolytic viruses represent a promising therapeutic approach to prevent cancer relapse by eliminating tumor-initiating cells that contaminate the autograft. Here we summarize an ex vivo “purging” strategy with oncolytic myxoma virus (MYXV) to remove cancer-initiating cells from patient autografts prior to transplantation. MYXV, a novel oncolytic poxvirus with potent anti-cancer properties in a variety of in vivo tumor models, can specifically eliminate cancerous stem and progenitor cells from samples obtained from acute myelogenous leukemia (AML) patients, while sparing normal CD34+ hematopoietic stem and progenitor cells capable of rescuing hematopoiesis following high dose conditioning. We propose that a broader subset of patients with intractable hematologic malignancies who have failed standard therapy could become eligible for ABMT when the treatment schema is coupled with ex vivo oncolytic therapy.

Introduction

Hematologic malignancies such as acute myelogenous leukemia (AML), myelodysplastic syndromes, lymphomas and multiple myeloma together represent about 9.5% of the total new cancer cases diagnosed in the United States. High-dose chemotherapy and radiotherapy to eliminate cancer cells in the patient followed by autologous blood and marrow transplantation (ABMT) has been used as an effective therapeutic modality to treat hematologic malignancies and solid tumors including neuroblastoma (1, 2). Success with ABMT relies on the autograft being free of cancer cells, to minimize the chance of disease recurrence. Generally, relapse of disease can be either caused by residual therapy-resistant cancer niches in the patient or by contaminating cancer cells within the autograft. Hematopoietic stem and progenitor cells (HSPCs) harvested from patients with advanced stage cancer often are contaminated with neoplastic cells, and thus can directly contribute to relapse following transplant (3).

In order to eliminate contaminating cancer cells from autografts prior to ABMT, several ex vivo cancer cell “purging” strategies have been tested over the last three decades. The main objective of purging is twofold: (a) to eliminate the number of contaminating cancer cells capable of initiating new tumor growth following transplant, and (b) to protect the normal HSPCs that are necessary for subsequent immune reconstitution. Different purging strategies have been evaluated, including ex vivo chemotherapy with drugs like mafosfamide and 4-hydroperoxycyclophosphamide (4, 5), CD34+ (stem) cell enrichment using immunomagnetic selection (6), exploitation of immunotoxins or hybrid cytotoxic proteins designed to selectively kill cancer cells (7), immunomagnetic removal of tumor cells (8), and the use of oncolytic viruses (9). In many cases, these ex vivo purging methods were applied as combination therapy to make them more effective as an adjunct to ABMT. Examples of this include high-dose chemotherapy followed by CD34+ cell enrichment in the case of multiple myeloma (10), CD34+ cell enrichment followed by killing of residual cancer cells using immunotoxins (11), or a combination of low-dose chemotherapy and monoclonal antibodies (12). A summary of the different purging methods is presented in Table 1.

Table 1. Purging strategies to eliminate cancer cells from graft samples.

Method of
purging
Example of purging agents Tumor type Reference
Ex vivo
Chemotherapy 4-
hydroperoxycyclophosphamide
(4-HC) and mafosfamide
Breast cancer (70)
Monoclonal
antibodies
Alemtuzumab, Rituximab Chronic
lymphocytic
leukemia (CLL), B-
cell lymphoma
(71)
Positive
hematopoietic
stem cell
(CD34+)
selection
Immunomagnetic
enrichment/selection
B cell non-
Hodgkin’s
lymphoma
(72)
Photodynamic
purging process
Rhodamine Depletion of T cells
in allogenic stem
cell transplant
(73)
Immunotoxins Heregulin (HRG)-
Pseudomonas exotoxin (PE) 40
toxin
Breast cancer cell
lines MDA-MB-361,
ZR-75-1, MCF-7
(74)
Oncolytic
viruses
Herpes simplex virus type 1
(HSV-1)
Adenovirus
Reovirus

Myxoma
Breast cancer cells

Breast cancer cells
Lymphocytic
leukemia and
lymphoma
Acute myelogenous
leukemia
(23, 25, 27, 65)
In vivo
Chemotherapy Idarubicin, Cytarabine and
etoposide
Chronic
myelogenous
leukemia
(75)
Monoclonal Ab Rituximab Chronic
myelogenous
leukemia
(76)

All of these purging methods have certain limitations and therefore an unmet medical need exists for effective methods that will lead to complete eradication of contaminating tumor-initiating cells within autografts prior to ABMT. For example, purging strategies that utilize high doses of chemotherapy might damage normal HSPCs, which may delay or prevent normal hematopoietic cell engraftment. On the other hand, antibody-based cell enrichment methods are technically demanding, expensive, and are frequently incomplete at cancer cell purging. The success of ex vivo purging methods depends on the number and type of contaminating tumor cells within the autograft, as well as the biology of the individual cancers from which they are derived (13). A successful ex vivo purging method has to be technically feasible for routine clinical practice and applicable for a wide variety of cancer cell types, including so-called “cancer stem cells” that might reside within the patient autograft (14). In this review, we highlight the use of oncolytic viruses, particularly Myxoma virus, as a novel ex vivo purging agent for hematologic malignancies.

Virus-mediated oncolysis and cancer therapy

The use of viruses to target and kill cancer cells dates back almost a century (15). However, the real potential of oncolytic virotherapy for different cancers has gained particular attention over the past 15 years, as some of the candidate oncolytic viruses have entered clinical trials, or, in the case of one derivative of adenovirus, have reached clinical practice. The major oncolytic viruses include adenovirus, Herpes simplex virus (HSV), measles, vaccinia, reovirus and Newcastle disease virus (NDV) (16). In some cases, oncolytic viruses have been tested in combination with other anti-cancer drugs. For example, one Adenovirus construct (ONYX-015) was more effective when combined with cisplatin and 5-fluorouracil chemotherapy for treatment of head and neck cancer (17). Eventually, the E1B gene-deleted variant of adenovirus H101 was approved in China for clinical use, after a phase III study (18). These experiences provide a clinical paradigm for other oncolytic viruses for their progression from preclinical animal models, through clinical trials, to approved usage for cancer treatment in patients. However, in many cases the efficacy in patients has been limited and there have been continued concerns about safety in some patients, particularly those that are immunosuppressed.

An oncolytic virus candidate must meet several criteria to become an effective cancer therapy (19). The virus should preferentially replicate within cancer cells, spread within tumor tissues, avoid infecting normal cells and tissues, be amenable for genetic manipulation to improve its safety and efficacy, and be nonpathogenic to humans and preferably other animal hosts as well. In some cases, oncolytic viruses have been genetically engineered to express immune stimulatory molecules within the tumor, for example Vaccinia JX-594 encodes human GM-CSF (20), or to express gene products that will enhance tumor cell killing, for example adenovirus Ad5-CD/TKrep-encoding a cytosine deaminase/thymine kinase (CD/TK) fusion gene (21). However, these genetic alterations in viruses should not result in toxic effects on normal somatic cells or tissues. Virus-mediated oncolysis as an ex vivo cancer cell purging strategy

Oncolytic viruses, with or without genetic modifications, have been tested for ex vivo elimination of cancer cells, as previous studies have indicated that normal bone marrow cells are particularly resistant to infection by many oncolytic virus candidates. A recombinant HSV-1 vector (JS1/34.5-/47-) with deletions of the ICP34.5 and ICP47 genes, permitting tumor-selective replication and enhanced antigen presentation in HSV-infected cells, was reported to successfully infect breast cancer cells, without affecting normal bone marrow cells in vitro (22). Similar results were found using another recombinant HSV-1 (HSV-1/G207) in an in vitro infection model (23). However, neither virus has been tested in vivo to evaluate the actual purging efficiency for ABMT. The purging potential of vesicular stomatitis virus (VSV), a negative strand RNA virus, was tested using interferon-sensitive VSV variants AV1, AV2 and a heat-resistant strain of VSV. These viruses were able to selectively eliminate a leukemic cell line with only minimal effects on the colony-forming ability of normal peripheral blood progenitor cells (PBPCs) (24). Another potential oncolytic virus, reovirus, a double-stranded RNA virus, has also been tested for its purging ability of cancer cells. In an in vitro purging strategy, reovirus eliminated myeloma cells or breast cancer cells without affecting the subsequent colony-forming potential of normal CD34+ stem cells (25, 26). All of these oncolytic viruses have thus shown promise in the in vitro purging of tumor cells with minimal toxicity on normal CD34+ stem cells. However, none of these studies directly demonstrated ex vivo cancer cell purging, because the virus-treated cells were never tested for their in vivo engraftment potential in immunocompromised hosts.

One oncolytic DNA virus, an adenovirus derivative, and its genetically engineered variants, such as virus expressing cytosine deaminase (Ad-CD) or carrying wild-type p53 as a transgene, have been tested in several ex vivo purging models of breast, myeloma and neuroblastoma tumor cells. In addition to ex vivo purging, adenovirus had no toxic effects on engraftment of normal human hematopoietic cells when the virus-purged cells were subsequently injected in a non-obese diabetic/severe combined immunodeficient (NOD/scid) immunocompromised mouse model (27, 28). Adenovirus has also been tested for ex vivo elimination of cancer cells in combination with other treatment methods. For example, breast cancer cells treated with Ad-CD and 5-fluorocytosine (5-FC) were unable to form tumors after transplantation in mice (29). Although these viruses were effective at ex vivo purging of several human cancer cell lines, they have not been tested for ex vivo purging of primary human cancer cells derived directly from patients.

Oncolytic poxviruses: Myxoma virus, a new oncolytic virus candidate

Poxviruses are all large, double-stranded DNA viruses that infect either invertebrates (Entomopoxvirinae) or vertebrates (Chordopoxvirinae) and replicate exclusively in the cytoplasm of infected cells. Vaccinia virus, the prototypic member of the genus orthopoxvirus and used as the vaccine to eradicate smallpox, has been tested in various cancer models, and attenuated variants are currently in different phases of clinical trial (30-33). Based on the success with smallpox vaccination, vaccinia and related poxviruses such as fowlpox and canarypox (genus avipoxvirus), which are genetically modified to express foreign/tumor antigens, have been used in many vaccine trials for protection against many infectious diseases and for treatment of established cancers (34, 35).

Several inherent features of poxviruses make certain specific members attractive candidates for oncolytic virotherapy. Poxvirus DNA is never integrated into the host genome, as these viruses exclusively replicate in the cytoplasm of infected cells. Most poxviruses can infect a broad variety of cell types, at least in vitro, because no cell-specific binding/entry receptor has yet been described. But at the host organism level, infection is usually severely restricted to a small number of susceptible host species (36-38). The large genomes of poxviruses have made them amenable for genetic manipulation and use for vaccination, gene therapy, and cancer therapy (30). Poxviruses induce strong cellular and humoral immune responses that can be detected many decades later, as reported for vaccinia in smallpox eradication (39). A number of newly developed antiviral agents such as ST-246, cidofovir, and derivatives of cidofovir are available to treat orthopoxvirus infections, such as in cases of pathogenicity associated with generalized vaccinia infections (40). Over the last 40 years, both natural vaccine strains and genetically engineered vaccinia viruses (such as JX-594) have been tested in preclinical cancer models and in clinical trials (15, 30).

Another poxvirus family member, called Myxoma virus (MYXV), has been recently developed as a novel oncolytic virus candidate (41). MYXV is the prototypic member of the Leporipoxvirus genus of Poxviridae family of viruses, which causes lethal disease myxomatosis in European rabbits (Oryctolagus cuniculus), but is completely restricted to rabbits as its host species. The South American tapeti (Sylvilagus brasilensis) and the North American brush rabbit (Sylvilagus bruneii) are the natural reservoirs for MYXV, where the virus causes only a benign infection (42). This suggests that even within related lagomorphs, host genetic variation can dictate viral pathogenicity. Other than lagomorphs, MYXV has not been reported to infect any other vertebrate species, including humans or mice (36, 43). The genome of MYXV strain Lausanne has been sequenced, and many of the encoded immunomodulatory proteins that regulate tissue or cellular tropism of virus have been characterized (43-46).

At the cellular level, the rabbit-specific host restriction of MYXV replication has been studied in vitro and in vivo. For example, MYXV is unable to replicate in primary mouse fibroblasts because MYXV infection induces a robust type I IFN signaling pathway response that aborts the virus infection (47). In contrast, in primary human fibroblasts MYXV is permissive unless blocked by a synergistic anti-viral response mediated by type I IFN plus TNF (48). Since many human cell types, such as primary macrophages, robustly produce these two cytokines in response to MYXV infection, any tissues that possess cells capable of expressing IFN plus TNF can protect all the resident primary human cells in a paracrine-like fashion (48, 49). In contrast, MYXV infection is fully permissive in a wide variety of different cancer cells derived from either mice or humans. In vitro, MYXV can selectively replicate within and kill the majority of human cancer cell lines listed in National Cancer Institute (NCI) reference (50). The mechanistic basis for this selective killing of cancer cells by MYXV is still being investigated, but two significant contributing factors are 1) the fact that most human cancer cells lack normal type I IFN and TNF synergy responses (51), and 2) many cancer cells possess excessive levels of activated Akt, which facilitates MYXV replication (52). It does seem clear that the selective cancer cell tropism of MYXV is essentially tied to the signaling environment of the cell and is independent of the origin of the tumor tissue from which the cancer cells are derived. Like cancer cells of human origin, many mouse cancer cells are also fully permissive to MYXV infection, even though mice (like humans) are not permissive hosts for the virus (53).

At the molecular level, there are several indications that MYXV distinguishes permissive versus nonpermissive cells by virtue of their variant innate signaling capacities, and not by any receptor differences at the cell surfaces. For example, compared to normal primary human somatic cells, all human cancer cells tested to date lack the ability to induce the synergistic IFN + TNF signaling responses and are not able to induce a sufficiently robust anti-viral state to block MYXV replication completely (51). As mentioned above, other cellular signaling molecules have been shown to also govern the cancer cell tropism of MYXV in human cancer cells. For example, the activation state of cellular Akt/PKB, a serine threonine kinase that plays an important role in cell survival, proliferation and cell death, also regulates MYXV tropism in cancer cells (52). Akt signaling is frequently dysregulated in a wide spectrum of human cancer cells (54), and the increased level of cellular Akt activation is directly linked to MYXV tropism in these cells (52). Screening of several MYXV host-range gene knockout viruses in human cancer cell lines has identified one particular viral host range gene, called M-T5, that regulates the ability of MYXV to productively infect different human cancer cells (50). The viral M-T5 protein interacts directly with the cellular Akt, and this protein/protein interaction regulates MYXV permissiveness in a particular class of human cancer cells called Type II (52). Cancer cells that possess naturally high levels of endogenous activated Akt (called Type I cells) are permissive for MYXV replication, whether the virus expresses M-T5 or not. A second type of human cancer cells (Type II cells) also exist, where Akt is directly activated by M-T5, and MYXV can infect those cells only when M-T5 is expressed. Thus, wild-type MYXV is permissive in type II cancer cells, but the M-T5 knockout variant of MYXV is nonpermissive in these same cells. The third type of cancer cells (called Type III) possess only low or undetectable levels of Akt activation, and MYXV cannot infect these cells in vitro or cause the activation of Akt even in the presence of M-T5 (52, 55). The requirement of activated Akt for productive MYXV infection is independent of origin of the tumor tissue (53).

Although MYXV infection in cancer cells depends on activated Akt, at least in vitro, it is still not known why MYXV specifically requires this hyper-phosphorylated form of Akt. The activated Akt-mediated signaling apparently makes transformed cancer cells a better environment for MYXV replication, possibly by either blocking key anti-viral pathways or by inducing cellular factors that are required for optimal MYXV replication. Importantly, however, the cellular Akt activation level can be manipulated with certain anti-cancer drugs that can allow MYXV replication even in nonpermissive cancer cells. For example, rapamycin increases MYXV tropism for certain cancer cells in vitro and in vivo (53, 56, 57). However, it is also possible that other cellular signaling pathways might also regulate MYXV tropism for cancer cells. In the case of vaccinia virus, viral replication and spread depends on activation of epidermal growth factor receptor (EGFR)-Ras signaling pathway, and in most cancer cells EGFR-Ras pathway is highly activated (58-60). In order to make vaccinia virus more effective as an oncolytic therapeutic, the virus has been genetically modified to delete viral genes that might compromise infected cell killing or to reduce viral virulence. Examples include vaccinia viruses that have been engineered to lack the viral thymidine kinase, vaccinia growth factor that are deleted for self-protective genes that block cellular apoptosis (like serpins), or anti-viral response (such as inhibitors of type I IFN) (30).

Preclinical animal models of MYXV oncolysis

The potential of MYXV as an oncolytic virus has been tested in several preclinical cancer models. In vivo efficacy of MYXV was first demonstrated in an orthotopic model of human glioblastoma in immunocompromised mice (61). The results demonstrated that intratumoral (i.t.) injection of MYXV decreased the xenografted tumor size and prolonged the survival of mice. However, the virus was able to effectively kill only the cells within the tumor where it was directly injected and did not migrate to an implanted tumor in the contralateral hemisphere (61). This could be because of type I IFN-mediated clearance of MYXV outside of the tumor and/or the lack of a permissive migratory cell to carry the virus to a distant tumor site. In another study, MYXV injected by I.T. route was able to successfully reduce the burden of xenografted human teratoid/rhabdoid tumors, a highly aggressive pediatric tumor implanted by subcutaneous (S.C.) injection in CD-1 nude mice (62). In this model, MYXV reduced the tumor and increased the survival of mice only when injected intracranially but not by intravenous (I.V.) injection. MYXV was also tested in a syngeneic metastatic B16F10 tumor model in immunocompetent mice. In this model, both I.T. and systemic administration of MYXV reduced lung tumor burden (53). Additionally, a combination therapy of MYXV plus rapamycin reduced both size and number of lung metastases (53). In another study, combination therapy with rapamycin plus MYXV also increased the in vivo oncolysis in an orthotopic medulloblastoma tumor model, the most common malignant brain tumor in children. This combination therapy reduced spinal cord and ventricle metastases in a xenograft mouse model (57). All these studies clearly suggest that MYXV has the ability to target and kill cancer cells in situ even without any genetic manipulation of the virus and without exhibiting any detectable toxicity to the animals. Also, MYXV did not replicate within any noncancerous host tissues, even following systemic injection into severely immunocompromised mice.

Purging of leukemic stem and progenitor cells with Myxoma virus

Apart from its natural rabbit host, MYXV has never been isolated or shown to replicate in humans, non-human primates or any other animal species outside of lagomorphs. The safety record of MYXV can be supported by the fact that even after its deliberate release in Australia in the 1950’s (or later in Europe) to control the feral rabbit population, there is no report so far that the released virus caused any disease in humans or in any other animal species other than rabbits (63). Another advantage with MYXV is that, like other DNA viruses, genetic manipulation is possible by insertion of multiple therapeutic transgenes in the large genome of MYXV. Additionally, targeted deletions of MYXV virulence genes can render the virus nonpathogenic even for rabbits, while leaving its ability to infect and kill human cancer cells intact (64).

The inherent ability of MYXV to selectively target cancer cells and spare normal cells makes it a suitable oncolytic virus candidate for ex vivo purging of human cancer cells prior to autografts. The advantage of this specific ex vivo infection strategy for ABMT is that the protocol can be modified to ensure that every potential cancer cell within an autograft is exposed to virus, setting up the possibility of infecting and eliminating contaminating cancer cells in the graft. The potential exploitation of MYXV for purging of hematologic cancers has recently been tested with primary human AML cells and compared to normal CD34+ HSPCs. The purpose of this study was to investigate 1) whether MYXV infection ex vivo could purge AML cells and prevent their engraftment into immunodeficient mice in vivo, and 2) to determine if MYXV treatment had any adverse effect on normal human donor HSPC functions (65). AML is a hematologic malignancy characterized by uncontrolled proliferation and accumulation of clonal neoplastic cells due to mutations which occur in response to various cytogenetic and microenvironmental abnormalities. AML accounts for approximately 80% of all adult leukemias and its overall incidence is increasing over the last 15-20 years. Certain types of AML are high risk for relapse and death due to resistance of conventional chemotherapy (66-68).

In this study, MYXV safety with normal human donor CD34+ HSPCs was tested in vitro and in vivo. MYXV did not infect or alter the differentiation functions of virus-treated CD34+ HSPC populations, even at a high multiplicity of infection (MOI) of 10. As tested by an in vitro colony-forming cell (CFC) assay, MYXV-treated normal HSPCs formed all the expected types of leukocyte colonies similar to the mock-treated CD34+ control HSPCs (65). In contrast to the normal HSPCs, primary leukemic AML cells were highly infected by MYXV when tested in vitro. It has been reported that PI3K/Akt signaling is frequently activated in AML patient blasts, which contribute to the proliferation, survival and drug resistance of these cells (69). In the case of AML, the activation of PI3K/Akt signaling is probably due to mutations in FLT3, Ras or c-Kit. More importantly, MYXV productively infected AML cells in vitro, and the infected cells lost their ability to form colonies of leukemic blast cells (65) .

The oncolytic purging potential of MYXV was also tested in vivo using an immunocompromised mouse model. The safety and ability of normal human bone marrow-derived HSPCs (CD34+ selected cells) to engraft in the bone marrow was tested in severely immunocompromised NOD/scid/IL2-R□ −/− (NOG) mice. When these cells were treated with MYXV ex vivo and transplanted in sublethally irradiated NOG mice, their engraftment property remained unchanged, as confirmed by analysis of mouse bone marrow for the engraftment of human CD45+/HLA-abc+ cells (65). The level of normal human cell engraftment in the transplanted NOG mice was essentially the same in the case of mock- and MYXV-treated cells. These results confirmed that both in vitro and in vivo MYXV does not alter the engraftment properties of normal human HSPCs. The use of NOG mice and absence of any toxicity after analyzing different tissues from mice also confirmed the safety of MYXV, even in the immunocompromised animals.

In vivo MYXV purging efficacy was then tested using primary AML cells that possess a diagnostic FLT3 internal tandem duplication (ITD). The MYXV-treated AML FLT3 ITD+ mononuclear cells were transplanted in sublethally irradiated NOG mice. Analysis of BM samples for the presence of human CD45+/HLA-abc+ engraftment by FACS analysis demonstrated that MYXV was able to efficiently purge the leukemic cells, as 90% of the transplanted mice had no detectable engraftment (65). Conversely, mock-treated AML samples demonstrated robust leukemia engraftment in all transplanted mice. This suggests that MYXV is a promising oncolytic virus candidate for ex vivo purging, as it is safe for normal HSPCs and successfully eliminates leukemic cells from patient autografts before transplant. Future studies with this ex vivo purging protocol using MYXV will next focus on patient HSPC donor samples with detectable levels of cancerous cells that can be tracked and distinguished from their normal HSPC counterparts following engraftment.

Concluding remarks

Oncolytic viruses have shown great promise for the treatment of certain human tumors, but their potential as anti-cancer therapeutics is only beginning to be exploited. Although oncolytic viruses preferentially infect cancer cells compared to normal somatic cells, they might be even more effective for certain specific procedures where delivery of the virus to the target cancer cells can be rigorously controlled, such as ex vivo purging of cancer cells that contaminate autografts used for ABMT. Among the oncolytic poxviruses, MYXV has great potential because of its natural ability to selectively kill cancer cells, the absence of deleterious effects on normal/stem cell functions, and lack of toxicity, as demonstrated using even extremely immunocompromised animal models. Ex vivo purging of hematologic cancer cells using various oncolytic viruses has been tested by different groups in the past, but none of these studies have been extended to include in vivo engraftment studies with primary patient cancer cells, as reported here. In this ex vivo delivery strategy, one major concern is the safety of these viruses in humans, particularly for severely immunocompromised patients. MYXV could be an ideal candidate for both ex vivo and in vivo purging of cancer cells that contaminate autografts or reside in patient tissue niches. In the future, MYXV host range mutants generated by the targeted deletion of viral genes, like M063KO and M135KO, can also be tested for purging, as they are even safer for rabbits and to some extent may even possess superior oncolytic properties (64). Our future goal would be production of clinical grade MYXV that can be exploited for human trials for this promising new therapeutic.

Acknowledgements

This study was supported by start-up funding to GM from University of Florida, College of Medicine, NIH R01 AI080607 and CA138541 to GM, and a grant from the Leukemia Lymphoma Society to CRC.

Biography

graphic file with name nihms-181384-b0001.gif Masmudur M. Rahman, Ph.D. Dr. Rahman is a Research Assistant Professor in the Department of Molecular Genetics and Microbiology, University of Florida. He received his Ph.D. in 2004 from the Indian Institute of Science, Bangalore, India, where he studied the biotechnological application of Bombyx mori nucleopolyhedrovirus and its interaction with host. His Postdoctoral training was at Roberts Research Institute, London, Ontario, Canada in the areas of poxvirus-encoded immunomodulatory proteins. Dr. Rahman’s current research focuses on understanding the mechanism of poxvirus-mediated manipulation of host innate immune system and cancer therapy using oncolytic poxviruses. Dr. Rahman has authored several peer-reviewed original papers, review articles and book chapters.

graphic file with name nihms-181384-b0002.gif Gerard J. Madlambayan, Ph.D. Dr. Madlambayan is a Research Assistant Professor in the Department of Medicine, Division of Hematology/Oncology. He obtained a Ph.D. degree from the University of Toronto and a Masters degree from the University of Michigan in the areas of Chemical and Biomedical Engineering. Dr. Madlambayan is recognized for his translational research efforts in the area of stem cell engineering and cancer biology. Dr. Madlambayan has industry experience as the Senior Scientist and Laboratory Director of Insception Biosciences, Canada’s leading stem cell research company. Dr. Madlambayan has developed and patented a bioprocess for the growth of cord blood-derived stem cells, which has been approved for use in clinical trials. Dr. Madlambayan has authored peer-reviewed original papers, review articles and book chapters in the areas of stem cell engineering and cancer biology and has presented his research at several international conferences.

graphic file with name nihms-181384-b0003.gif Christopher R. Cogle, MD Dr. Cogle is an Assistant Professor of Medicine at the University of Florida, Division of Hematology/Oncology, and is the Research Director of the Shands Hospital Stem Cell Laboratory. He is recognized for his adult hemangioblast translational research, with peer-reviewed grant support from the NIH, Florida Department of Health and Leukemia Lymphoma Society. Dr. Cogle’s clinical expertise is in leukemias, myelodysplastic syndromes and blood & marrow transplantation. His laboratory focus is in human hemangioblast activity of normal and malignant adult stem and progenitor cells. His translational research focus is in translating hemangioblast therapies into the clinic. Dr. Cogle’s lab uses pre-clinical animal models of human hematologic malignancies to study dependent pathways of hematologic malignancies. He has translated these discoveries into phase I and II clinical trials testing novel agents for treatment of leukemia and myelodysplastic syndromes.

graphic file with name nihms-181384-b0004.gif Grant McFadden, Ph.D. Dr. McFadden is a professor in the Department of Molecular Genetics and Microbiology, University of Florida. He received his Ph.D. from McGill University in Montreal, Canada, in 1975. Dr. McFadden held several academic positions in Canada until 2006, when he relocated to the University of Florida, USA. His laboratory studies how viruses interact with the host immune system, in particular the mechanisms that determine the tropism and host range of poxviruses. The lab is also developing specific poxviruses as novel virotherapy candidates for the oncolytic treatment of human cancers. The lab has investigated the molecular basis for the host and cellular tropism specificity of poxviruses, using the myxoma virus model system as a novel platform for replication-restricted vaccine vectors and as an oncolytic virus. His lab also studies how viruses in general can occasionally leap from a long-term evolutionary host species to cause zoonotic infections in humans. The McFadden lab is also investigating whether myxoma virus can selectively infect and kill primary human cancer stem cells but leave normal human stem cells untouched.

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

References

  • 1.Kvalheim G. Purging of autografts: methods and clinical significance. Ann Med. 1996;28:167–173. doi: 10.3109/07853899609092942. [DOI] [PubMed] [Google Scholar]
  • 2.Gribben JG, Nadler LM. Bone marrow purging for autologous bone marrow transplantation. Leuk Lymphoma. 1993;11(Suppl 2):141–148. doi: 10.3109/10428199309064274. [DOI] [PubMed] [Google Scholar]
  • 3.Rill DR, Santana VM, Roberts WM, et al. Direct demonstration that autologous bone marrow transplantation for solid tumors can return a multiplicity of tumorigenic cells. Blood. 1994;84:380–383. [PubMed] [Google Scholar]
  • 4.Motta MR, Mangianti S, Rizzi S, et al. Pharmacological purging of minimal residual disease from peripheral blood stem cell collections of acute myeloblastic leukemia patients: preclinical studies. Exp Hematol. 1997;25:1261–1269. [PubMed] [Google Scholar]
  • 5.Strauss G, Westhoff MA, Fischer-Posovszky P, et al. 4-hydroperoxy-cyclophosphamide mediates caspase-independent T-cell apoptosis involving oxidative stress-induced nuclear relocation of mitochondrial apoptogenic factors AIF and EndoG. Cell Death Differ. 2008;15:332–343. doi: 10.1038/sj.cdd.4402272. [DOI] [PubMed] [Google Scholar]
  • 6.Abonour R, Scott KM, Kunkel LA, et al. Autologous transplantation of mobilized peripheral blood CD34+ cells selected by immunomagnetic procedures in patients with multiple myeloma. Bone Marrow Transplant. 1998;22:957–963. doi: 10.1038/sj.bmt.1701473. [DOI] [PubMed] [Google Scholar]
  • 7.Spyridonidis A, Schmidt M, Bernhardt W, et al. Purging of mammary carcinoma cells during ex vivo culture of CD34+ hematopoietic progenitor cells with recombinant immunotoxins. Blood. 1998;91:1820–1827. [PubMed] [Google Scholar]
  • 8.Gee A, Moss T, Mansour V, et al. Large-scale immunomagnetic separation system for the removal of tumor cells from bone marrow. Prog Clin Biol Res. 1992;377:181–187. [PubMed] [Google Scholar]
  • 9.Thirukkumaran CM, Russell JA, Stewart DA, Morris DG. Viral purging of haematological autografts: should we sneeze on the graft? Bone Marrow Transplant. 2007;40:1–12. doi: 10.1038/sj.bmt.1705668. [DOI] [PubMed] [Google Scholar]
  • 10.Stewart AK, Vescio R, Schiller G, et al. Purging of autologous peripheral-blood stem cells using CD34 selection does not improve overall or progression-free survival after high-dose chemotherapy for multiple myeloma: results of a multicenter randomized controlled trial. J Clin Oncol. 2001;19:3771–3779. doi: 10.1200/JCO.2001.19.17.3771. [DOI] [PubMed] [Google Scholar]
  • 11.Kvalheim G, Wang MY, Pharo A, et al. Purging of tumor cells from leukapheresis products: experimental and clinical aspects. J Hematother. 1996;5:427–436. doi: 10.1089/scd.1.1996.5.427. [DOI] [PubMed] [Google Scholar]
  • 12.Zhong RK, Donnenberg AD, Rubin J, Ball ED. Differential effect of 4-hydroperoxycyclophosphamide and antimyeloid monoclonal antibodies on T and natural killer cells during bone marrow purging. Blood. 1994;83:2345–2351. [PubMed] [Google Scholar]
  • 13.Shimoni A, Korbling M. Tumor cell contamination in re-infused stem cell autografts: does it have clinical significance? Crit Rev Oncol Hematol. 2002;41:241–250. doi: 10.1016/s1040-8428(01)00160-3. [DOI] [PubMed] [Google Scholar]
  • 14.Cripe TP, Wang PY, Marcato P, Mahller YY, Lee PW. Targeting cancer-initiating cells with oncolytic viruses. Mol Ther. 2009;17:1677–1682. doi: 10.1038/mt.2009.193. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Liu TC, Galanis E, Kirn D. Clinical trial results with oncolytic virotherapy: a century of promise, a decade of progress. Nat Clin Pract Oncol. 2007;4:101–117. doi: 10.1038/ncponc0736. [DOI] [PubMed] [Google Scholar]
  • 16.Roberts MS, Lorence RM, Groene WS, Bamat MK. Naturally oncolytic viruses. Curr Opin Mol Ther. 2006;8:314–321. [PubMed] [Google Scholar]
  • 17.Khuri FR, Nemunaitis J, Ganly I, et al. a controlled trial of intratumoral ONYX-015, a selectively-replicating adenovirus, in combination with cisplatin and 5-fluorouracil in patients with recurrent head and neck cancer. Nat Med. 2000;6:879–885. doi: 10.1038/78638. [DOI] [PubMed] [Google Scholar]
  • 18.Garber K. China approves world’s first oncolytic virus therapy for cancer treatment. J Natl Cancer Inst. 2006;98:298–300. doi: 10.1093/jnci/djj111. [DOI] [PubMed] [Google Scholar]
  • 19.Liu TC, Kirn D. Systemic efficacy with oncolytic virus therapeutics: clinical proof-of-concept and future directions. Cancer Res. 2007;67:429–432. doi: 10.1158/0008-5472.CAN-06-2871. [DOI] [PubMed] [Google Scholar]
  • 20.Mastrangelo MJ, Maguire HC, Jr., Eisenlohr LC, et al. Intratumoral recombinant GM-CSF-encoding virus as gene therapy in patients with cutaneous melanoma. Cancer Gene Ther. 1999;6:409–422. doi: 10.1038/sj.cgt.7700066. [DOI] [PubMed] [Google Scholar]
  • 21.Freytag SO, Khil M, Stricker H, et al. Phase I study of replication-competent adenovirus-mediated double suicide gene therapy for the treatment of locally recurrent prostate cancer. Cancer Res. 2002;62:4968–4976. [PubMed] [Google Scholar]
  • 22.Hu JC, Booth MJ, Tripuraneni G, et al. A novel HSV-1 virus, JS1/34.5-/47-, purges contaminating breast cancer cells from bone marrow. Clin Cancer Res. 2006;12:6853–6862. doi: 10.1158/1078-0432.CCR-06-1228. [DOI] [PubMed] [Google Scholar]
  • 23.Wu A, Mazumder A, Martuza RL, et al. Biological purging of breast cancer cells using an attenuated replication-competent herpes simplex virus in human hematopoietic stem cell transplantation. Cancer Res. 2001;61:3009–3015. [PubMed] [Google Scholar]
  • 24.Lichty BD, Stojdl DF, Taylor RA, et al. Vesicular stomatitis virus: a potential therapeutic virus for the treatment of hematologic malignancy. Hum Gene Ther. 2004;15:821–831. doi: 10.1089/hum.2004.15.821. [DOI] [PubMed] [Google Scholar]
  • 25.Thirukkumaran CM, Luider JM, Stewart DA, et al. Reovirus oncolysis as a novel purging strategy for autologous stem cell transplantation. Blood. 2003;102:377–387. doi: 10.1182/blood-2002-08-2508. [DOI] [PubMed] [Google Scholar]
  • 26.Thirukkumaran CM, Luider JM, Stewart DA, et al. Biological purging of breast cancer cell lines using a replication-competent oncolytic virus in human stem cell autografts. Bone Marrow Transplant. 2005;35:1055–1064. doi: 10.1038/sj.bmt.1704931. [DOI] [PubMed] [Google Scholar]
  • 27.Lillo R, Ramirez M, Alvarez A, et al. Efficient and nontoxic adenoviral purging method for autologous transplantation in breast cancer patients. Cancer Res. 2002;62:5013–5018. [PubMed] [Google Scholar]
  • 28.Wagner LM, Guichard SM, Burger RA, et al. Efficacy and toxicity of a virus-directed enzyme prodrug therapy purging method: preclinical assessment and application to bone marrow samples from neuroblastoma patients. Cancer Res. 2002;62:5001–5007. [PubMed] [Google Scholar]
  • 29.Wolff G, Korner IJ, Schumacher A, Arnold W, Dorken B, Mapara MY. Ex vivo breast cancer cell purging by adenovirus-mediated cytosine deaminase gene transfer and short-term incubation with 5-fluorocytosine completely prevents tumor growth after transplantation. Hum Gene Ther. 1998;9:2277–2284. doi: 10.1089/hum.1998.9.15-2277. [DOI] [PubMed] [Google Scholar]
  • 30.Kirn DH, Thorne SH. Targeted and armed oncolytic poxviruses: a novel multi-mechanistic therapeutic class for cancer. Nat Rev Cancer. 2009;9:64–71. doi: 10.1038/nrc2545. [DOI] [PubMed] [Google Scholar]
  • 31.Shen Y, Nemunaitis J. Fighting cancer with vaccinia virus: teaching new tricks to an old dog. Mol Ther. 2005;11:180–195. doi: 10.1016/j.ymthe.2004.10.015. [DOI] [PubMed] [Google Scholar]
  • 32.Thorne SH, Bartlett DL, Kirn DH. The use of oncolytic vaccinia viruses in the treatment of cancer: a new role for an old ally? Curr Gene Ther. 2005;5:429–443. doi: 10.2174/1566523054546215. [DOI] [PubMed] [Google Scholar]
  • 33.Worschech A, Haddad D, Stroncek DF, Wang E, Marincola FM, Szalay AA. The immunologic aspects of poxvirus oncolytic therapy. Cancer Immunol Immunother. 2009;58:1355–1362. doi: 10.1007/s00262-009-0686-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Gilbert PA, McFadden G. Poxvirus cancer therapy. Recent Pat Antiinfect Drug Discov. 2006;1:309–321. doi: 10.2174/157489106778777592. [DOI] [PubMed] [Google Scholar]
  • 35.Guo ZS, Thorne SH, Bartlett DL. Oncolytic virotherapy: molecular targets in tumor-selective replication and carrier cell-mediated delivery of oncolytic viruses. Biochim Biophys Acta. 2008;1785:217–231. doi: 10.1016/j.bbcan.2008.02.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.McFadden G. Poxvirus tropism. Nat Rev Microbiol. 2005;3:201–213. doi: 10.1038/nrmicro1099. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Mercer J, Helenius A. Vaccinia virus uses macropinocytosis and apoptotic mimicry to enter host cells. Science. 2008;320:531–535. doi: 10.1126/science.1155164. [DOI] [PubMed] [Google Scholar]
  • 38.Moss B. Poxvirus entry and membrane fusion. Virology. 2006;344:48–54. doi: 10.1016/j.virol.2005.09.037. [DOI] [PubMed] [Google Scholar]
  • 39.Putz MM, Midgley CM, Law M, Smith GL. Quantification of antibody responses against multiple antigens of the two infectious forms of Vaccinia virus provides a benchmark for smallpox vaccination. Nat Med. 2006;12:1310–1315. doi: 10.1038/nm1457. [DOI] [PubMed] [Google Scholar]
  • 40.Vora S, Damon I, Fulginiti V, et al. Severe eczema vaccinatum in a household contact of a smallpox vaccinee. Clin Infect Dis. 2008;46:1555–1561. doi: 10.1086/587668. [DOI] [PubMed] [Google Scholar]
  • 41.Stanford MM, McFadden G. Myxoma virus and oncolytic virotherapy: a new biologic weapon in the war against cancer. Expert Opin Biol Ther. 2007;7:1415–1425. doi: 10.1517/14712598.7.9.1415. [DOI] [PubMed] [Google Scholar]
  • 42.Fenner F. Adventures with poxviruses of vertebrates. FEMS Microbiol Rev. 2000;24:123–133. doi: 10.1016/S0168-6445(00)00027-9. [DOI] [PubMed] [Google Scholar]
  • 43.Stanford MM, Werden SJ, McFadden G. Myxoma virus in the European rabbit: interactions between the virus and its susceptible host. Vet Res. 2007;38:299–318. doi: 10.1051/vetres:2006054. [DOI] [PubMed] [Google Scholar]
  • 44.Barrett JW, Cao JX, Hota-Mitchell S, McFadden G. Immunomodulatory proteins of myxoma virus. Semin Immunol. 2001;13:73–84. doi: 10.1006/smim.2000.0298. [DOI] [PubMed] [Google Scholar]
  • 45.Kerr P, McFadden G. Immune responses to myxoma virus. Viral Immunol. 2002;15:229–246. doi: 10.1089/08828240260066198. [DOI] [PubMed] [Google Scholar]
  • 46.Zuniga MC. A pox on thee! Manipulation of the host immune system by myxoma virus and implications for viral-host co-adaptation. Virus Res. 2002;88:17–33. doi: 10.1016/s0168-1702(02)00118-1. [DOI] [PubMed] [Google Scholar]
  • 47.Wang F, Ma Y, Barrett JW, et al. Disruption of Erk-dependent type I interferon induction breaks the myxoma virus species barrier. Nat Immunol. 2004;5:1266–1274. doi: 10.1038/ni1132. [DOI] [PubMed] [Google Scholar]
  • 48.Wang F, Gao X, Barrett JW, et al. RIG-I mediates the co-induction of tumor necrosis factor and type I interferon elicited by myxoma virus in primary human macrophages. PLoS Pathog. 2008;4:e1000099. doi: 10.1371/journal.ppat.1000099. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Bartee E, Mohamed MR, Lopez MC, Baker HV, McFadden G. The addition of tumor necrosis factor plus beta interferon induces a novel synergistic antiviral state against poxviruses in primary human fibroblasts. J Virol. 2009;83:498–511. doi: 10.1128/JVI.01376-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Sypula J, Wang F, Ma Y, Bell J, McFadden G. Myxoma virus tropism in human tumor cells. Gene Ther Mol Biol. 2004;8:103–114. [Google Scholar]
  • 51.Bartee E, McFadden G. Human cancer cells have specifically lost the ability to induce the synergistic state caused by tumor necrosis factor plus interferon-beta. Cytokine. 2009;47:199–205. doi: 10.1016/j.cyto.2009.06.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Wang G, Barrett JW, Stanford M, et al. Infection of human cancer cells with myxoma virus requires Akt activation via interaction with a viral ankyrin-repeat host range factor. Proc Natl Acad Sci U S A. 2006;103:4640–4645. doi: 10.1073/pnas.0509341103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Stanford MM, Shaban M, Barrett JW, et al. Myxoma virus oncolysis of primary and metastatic B16F10 mouse tumors in vivo. Mol Ther. 2008;16:52–59. doi: 10.1038/sj.mt.6300348. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Tokunaga E, Oki E, Egashira A, et al. Deregulation of the Akt pathway in human cancer. Curr Cancer Drug Targets. 2008;8:27–36. doi: 10.2174/156800908783497140. [DOI] [PubMed] [Google Scholar]
  • 55.Werden SJ, McFadden G. The role of cell signaling in poxvirus tropism: the case of the M-T5 host range protein of myxoma virus. Biochim Biophys Acta. 2008;1784:228–237. doi: 10.1016/j.bbapap.2007.08.001. [DOI] [PubMed] [Google Scholar]
  • 56.Stanford MM, Barrett JW, Nazarian SH, Werden S, McFadden G. Oncolytic virotherapy synergism with signaling inhibitors: Rapamycin increases myxoma virus tropism for human tumor cells. J Virol. 2007;81:1251–1260. doi: 10.1128/JVI.01408-06. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Lun XQ, Zhou H, Alain T, et al. Targeting human medulloblastoma: oncolytic virotherapy with myxoma virus is enhanced by rapamycin. Cancer Res. 2007;67:8818–8827. doi: 10.1158/0008-5472.CAN-07-1214. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Katsafanas GC, Moss B. Vaccinia virus intermediate stage transcription is complemented by Ras-GTPase-activating protein SH3 domain-binding protein (G3BP) and cytoplasmic activation/proliferation-associated protein (p137) individually or as a heterodimer. J Biol Chem. 2004;279:52210–52217. doi: 10.1074/jbc.M411033200. [DOI] [PubMed] [Google Scholar]
  • 59.Yang H, Kim SK, Kim M, et al. Antiviral chemotherapy facilitates control of poxvirus infections through inhibition of cellular signal transduction. J Clin Invest. 2005;115:379–387. doi: 10.1172/JCI23220. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Soares JA, Leite FG, Andrade LG, et al. Activation of the PI3K/Akt pathway early during vaccinia and cowpox virus infections is required for both host survival and viral replication. J Virol. 2009;83:6883–6899. doi: 10.1128/JVI.00245-09. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Lun X, Yang W, Alain T, et al. Myxoma virus is a novel oncolytic virus with significant antitumor activity against experimental human gliomas. Cancer Res. 2005;65:9982–9990. doi: 10.1158/0008-5472.CAN-05-1201. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Wu Y, Lun X, Zhou H, et al. Oncolytic efficacy of recombinant vesicular stomatitis virus and myxoma virus in experimental models of rhabdoid tumors. Clin Cancer Res. 2008;14:1218–1227. doi: 10.1158/1078-0432.CCR-07-1330. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Fenner F, Ratcliffe F. Myxomatosis. University Press; Cambridge: 1965. [Google Scholar]
  • 64.Barrett JW, Alston LR, Wang F, et al. Identification of host range mutants of myxoma virus with altered oncolytic potential in human glioma cells. J Neurovirol. 2007;13:549–560. doi: 10.1080/13550280701591526. [DOI] [PubMed] [Google Scholar]
  • 65.Kim M, Madlambayan GJ, Rahman MM, Smallwood SE, Meacham AM, Hosaka K, Scott EW, Cogle CR, McFadden G. Myxoma virus targets primary human leukemia stem and progenitor cells while sparing normal hematopoietic stem and progenitor cells. Leukemia. 2009;23:2313–2317. doi: 10.1038/leu.2009.219. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Kiyoi H, Naoe T, Nakano Y, et al. Prognostic implication of FLT3 and N-RAS gene mutations in acute myeloid leukemia. Blood. 1999;93:3074–3080. [PubMed] [Google Scholar]
  • 67.Thiede C, Steudel C, Mohr B, et al. Analysis of FLT3-activating mutations in 979 patients with acute myelogenous leukemia: association with FAB subtypes and identification of subgroups with poor prognosis. Blood. 2002;99:4326–4335. doi: 10.1182/blood.v99.12.4326. [DOI] [PubMed] [Google Scholar]
  • 68.Schnittger S, Schoch C, Dugas M, et al. Analysis of FLT3 length mutations in 1003 patients with acute myeloid leukemia: correlation to cytogenetics, FAB subtype, and prognosis in the AMLCG study and usefulness as a marker for the detection of minimal residual disease. Blood. 2002;100:59–66. doi: 10.1182/blood.v100.1.59. [DOI] [PubMed] [Google Scholar]
  • 69.Martelli AM, Nyakern M, Tabellini G, et al. Phosphoinositide 3-kinase/Akt signaling pathway and its therapeutical implications for human acute myeloid leukemia. Leukemia. 2006;20:911–928. doi: 10.1038/sj.leu.2404245. [DOI] [PubMed] [Google Scholar]
  • 70.Shpall EJ, Jones RB, Bast RC, Jr., et al. 4-Hydroperoxycyclophosphamide purging of breast cancer from the mononuclear cell fraction of bone marrow in patients receiving high-dose chemotherapy and autologous marrow support: a phase I trial. J Clin Oncol. 1991;9:85–93. doi: 10.1200/JCO.1991.9.1.85. [DOI] [PubMed] [Google Scholar]
  • 71.Cheson BD. Monoclonal antibody therapy for B-cell malignancies. Semin Oncol. 2006;33:S2–14. doi: 10.1053/j.seminoncol.2006.01.024. [DOI] [PubMed] [Google Scholar]
  • 72.Mohr M, Dalmis F, Hilgenfeld E, et al. Simultaneous immunomagnetic CD34+ cell selection and B-cell depletion in peripheral blood progenitor cell samples of patients suffering from B-cell non-Hodgkin’s lymphoma. Clin Cancer Res. 2001;7:51–57. [PubMed] [Google Scholar]
  • 73.Goggins TF, Chao N. Depletion of host reactive T cells by photodynamic cell purging and prevention of graft versus host disease. Leuk Lymphoma. 2003;44:1871–1879. doi: 10.1080/1042819031000119226. [DOI] [PubMed] [Google Scholar]
  • 74.Keir M, Fiddes R, Biggs JC, Kearney PP. Sensitivity of c-erbB positive cells to a ligand toxin and its utility in purging breast cancer cells from peripheral blood stem cell (PBSC) collections. Stem Cells. 2000;18:422–427. doi: 10.1634/stemcells.18-6-422. [DOI] [PubMed] [Google Scholar]
  • 75.Carella AM, Lerma E, Corsetti MT, et al. Autografting with philadelphia chromosome-negative mobilized hematopoietic progenitor cells in chronic myelogenous leukemia. Blood. 1999;93:1534–1539. [PubMed] [Google Scholar]
  • 76.Bhatia S, Sather HN, Heerema NA, Trigg ME, Gaynon PS, Robison LL. Racial and ethnic differences in survival of children with acute lymphoblastic leukemia. Blood. 2002;100:1957–1964. doi: 10.1182/blood-2002-02-0395. [DOI] [PubMed] [Google Scholar]

RESOURCES