Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2019 Mar 1.
Published in final edited form as: Vet Comp Oncol. 2018 Oct 21;17(1):69–79. doi: 10.1111/vco.12447

Cancer Stem Cell Populations in Lymphoma in Dogs and Impact of Cytotoxic Chemotherapy

Genevieve Hartley *, Robyn Elmslie **, Brent Murphy *, Amanda Guth *, Steven Dow *
PMCID: PMC6364829  NIHMSID: NIHMS1008902  PMID: 30238600

Abstract

Cancer relapse following chemotherapy has been attributed in part to the presence of cancer stem cells (CSC), which drive tumor growth and metastasis and are highly resistant to the effects of cytotoxic chemotherapy. As a result, treatment with cytotoxic chemotherapy selects for drug-resistant CSC populations that eventually drive tumor recurrence. Little is known currently regarding the role of CSC in dogs with lymphoma, nor the impact of chemotherapy on CSC populations. Therefore, we prospectively quantitated CSC populations in dogs with B cell and T cell lymphoma, using tumor aspirates and flow cytometric analysis with a panel of CSC markers. In addition, in vitro studies were done to determine the impact of chemotherapy resistance on the stem cell phenotype and stem cell properties of lymphoma cells. We found that the percentages of tumor cells expressing CSC markers were significantly increased in dogs with B cell lymphoma, compared to B cells from normal lymph nodes. Similar findings were observed in dogs with T cell lymphoma. In vitro studies revealed that lymphoma cells selected for resistance to CHOP chemotherapy had significantly upregulated expression of CSC markers, formed spheroids in culture more readily, and expressed significantly greater aldehyde dehydrogenase activity compared to chemotherapy-sensitive tumor cells. Similar results were observed in tumor samples dogs with relapsed B cell lymphoma. These findings suggest that cytotoxic chemotherapy can lead to a relative enrichment of tumor cells with CSC properties, which may be associated with lymphoma recurrence.

Keywords: dog, immune, tumor, B cell, chemotherapy

Introduction.

Lymphoma accounts for up to 90% of all hematopoietic neoplasms in dogs, with certain breeds showing a higher risk of developing the disease1,2. The tumor is characterized by a clonal expansion of lymphoid cells, and the origin of the malignant cells (B cell versus T cell) is also strongly associated with response to therapy3. Most high-grade canine lymphomas are of B-cell origin, and usually manifest a higher complete response rate to cytotoxic chemotherapy with longer remission and survival times compared to T cell lymphomas in dogs46. Following treatment with standard cytotoxic chemotherapy (typically protocols that include some combination of cyclophosphamide, vincristine, hydroxydoxorubicin, and prednisone7), approximately 85% of dogs with B cell lymphoma (BLC) experience complete tumor remission6. The duration of remission averages less than 12 months for dogs with BCL, and 6 months for dogs with T cell lymphoma (TCL)6. Virtually all treated dogs will relapse and ultimately succumb to the original lymphoma, regardless of treatment protocol8,9. In lymphoma and other hematopoietic neoplasms in humans and in rodent models, tumor relapse and loss of sensitivity to chemotherapy has been attributed to selective expansion and/or enrichment of CSC populations1012.

Cancer stem cells are tumor cells that exhibit properties of self-renewal and asymmetric cell division, which allows them to generate both tumor populating cells as well as new CSC13,14. The CSC population is thought to arise from either normal stem cells or from progenitor cells that have undergone multiple mutations15,16. Alternatively, mutations in adult cells may drive cell de-differentiation to become more stem cell-like1719. Though the concept of CSC is still not universally accepted, there is compelling evidence from certain models that CSC can drive the growth and recurrence of primary tumors, and lead to tumor metastasis13,20,21. In addition, ablation of CSC has been shown experimentally to lead to the cessation of cancer growth, tumor regression, and decreased relapse of aggressive cancer, consistent with their key role in sustaining the overall tumor cell population2225. The unusual nature of CSC cell division, and the presence of metabolic pathways that are associated with drug efflux, render these cells relatively resistant to chemotherapy, compared to cancer daughter cells. Therefore, treatment with cytotoxic chemotherapy has been shown to selectively enrich lymphomas and leukemias for CSC, which eventually leads to disease recurrence2628.

Multiple different populations of CSC have been identified in lymphomas in humans and in rodent models, using cell surface expressed molecules and functional properties to identify CSC within the much larger population of differentiated tumor cells2931. Cell surface molecules used to define CSC in human lymphoma include CD20+/CD27+ B cells for Hodgkin’s lymphoma and CD45+/CD19+ cells in mantle cell lymphoma cells32,33. In human acute myeloid leukemia, CD34+/CD38 cells were identified as leukemia stem cells34, and one study found evidence suggesting that CD34+/CD90- cells were the source of leukemia stem cells35. However, another group reported that CD90 expression was linked to poor prognosis and high risk disease36, and CD90 has been identified as a marker of CSC in other tumor types37,38. Other markers used to identify leukemia stem cells include CD96, CD123, CD47, CD44, and CLL-13943.

A widely-accepted vitro assay to identify CSC in vitro is their ability to form tumor spheres under non-adherent and serum-free or low serum cell culture conditions44,45. The cells that form tumor spheres display CSC properties including resistance to chemotherapy and increased aldehyde dehydrogenase (ALDH) activity32,46. The ALDH family of enzymes is responsible for detoxifying cells, metabolizing chemotherapeutic drugs, and for retinoic acid signaling to maintain the CSC phenotype4749. For Hodgkin’s lymphoma, a circulating population of CD20+/CD27+ B cells with high levels of ALDH activity was found to be the source of Hodgkin and Reed-Sternberg cells32.

The goal of the present study was to characterize CSC populations in canine B and T cell lymphoma using a combination of surface marker expression and functional assays. We previously defined CSC populations in dogs with melanoma and osteosarcoma, but a similar analysis has not been done for dogs with lymphoma50. The current study leveraged many of the CSC properties defined in human lymphoma to evaluate canine lymphoma CSC. Our study was performed using flow cytometry and tumor aspirates from lymph nodes of 13 normal dogs, 44 dogs with untreated lymphoma, and 11 dogs with relapsed lymphoma following cytotoxic chemotherapy. The results of these studies indicated that CSC are present in relatively low numbers in dogs with BCL and TCL, and that CSC are enriched by cytotoxic chemotherapy. The presence of CSC subpopulations in canine lymphoma suggest that CSC could be considered an attractive target for selective targeted therapeutics.

Materials & Methods.

Study dogs.

Lymph node fine needle aspirates (FNA) and biopsies were obtained from dogs diagnosed with BCL or TCL by oncologists at the Veterinary Specialty & Emergency Hospital in Englewood, CO or the Flint Animal Cancer Center at the James Voss Veterinary Teaching Hospital at Colorado State University. Diagnosis of lymphoma was done by flow cytometry by the Clinical Immunology Laboratory at Colorado State University. Tumor relapsed dogs were defined as animals that initially experience complete remission following cytotoxic chemotherapy, and then later developed disease recurrence. All dogs were treated with either standard CHOP therapy protocols or with Tanovea (VetDC, Ft Collins, CO). Lymph node FNAs from healthy dogs were obtained from dogs owned by hospital staff, and the animals were deemed normal based on a normal physical exam and normal blood work conducted in the last year. All animal studies were approved by the Institutional Animal Care and Use Committee, and the Clinical Review Board, at Colorado State University.

The normal dog population included 8 females and 5 males. Ages for normal dogs ranged from 3 to 12 years, with the average age being 8 years. The BCL patient population included 20 untreated females, 3 relapsed females, 15 untreated males, and 6 relapsed males. The TCL patient population included 2 untreated females, 7 untreated males, 1 relapsed female, and 1 relapsed male. Ages for untreated lymphoma dogs ranged from 3 to 14 years with the average age being 9 years, and for relapsed lymphoma dogs from 5 to 13 years with the average age being 9 years. The mean age difference between normal, untreated lymphoma, and relapsed lymphoma was not statistically significant, Supplementary Figure 1.

Collection and processing of tumor samples.

Fine needle aspirates were obtained from the lymph nodes of 13 healthy dogs and from the affected lymph nodes of 35 dogs with untreated BCL, 9 dogs with untreated TCL, 9 dogs with relapsed BCL, and 2 dogs with relapsed TCL. Lymph node biopsies were obtained from 3 healthy dogs and from 3 dogs with untreated BCL. Samples were placed in tissue culture medium (described below) at 4C and were processed by analysis after overnight storage. Tissue sampling procedures were kept consistent for all samples and were obtained by the same hospital personnel.

Lymphoma cell lines.

The canine lymphoma cell lines CLBL1 (BCL51), 1771 (BCL52), and Oswald (TCL53) were maintained in MEM medium (Gibco, Grand Island, NY) supplemented with 10% FBS (Atlas Biologicals, Fort Collins, CO) and 5% CTM (10,000 ug/mL Pen/Strep, 200 mM L-glutamine, 10 mM essential amino acids without L-glutamine, 10 mM non-essential amino acids, and 7.5% bicarbonate solution (all from Gibco)54. These cell lines were validated as canine and screened to be genetically unique by PCR55.

Antibodies for flow cytometry and immunohistochemistry.

Antibodies used to quantitate expression of CSC markers included CD29 (clone Hmb1–1), CD34 (clone 1H6), CD90 (clone YK1X337.217), CD117 (clone ACK2), Sca1 (clone D7), CD133 (clone 13A4), and Oct3/4 (EM92) all obtained from eBioscience (San Diego, CA). Isotype matched, irrelevant antibodies were used at the same concentrations. To identify lymphocyte populations, B cell were identified as CD21+ (clone CA2.1D6) and T cells were identified as CD5+ (clone YKIX322.3) from Bio-Rad Laboratories (Hercules, CA), as described previously56.

Flow cytometry.

Samples were prepared for flow cytometry as described previously56. Briefly, fine needle aspirates were incubated with ammonium-chloride-potassium (ACK) lysis buffer (0.5% Phenol Red solution, 8% NH4Cl, 1% KHCO3, 0.037% Na2EDTA in distilled water) to lyse red blood cells prior to incubation with 10% normal dog serum (Jackson ImmunoResearch) to minimize non-specific binding. The samples were then immunostained with antibodies diluted in fluorescence-activated cell sorting (FACS) buffer (1% BSA in PBS with 0.05% sodium azide). To exclude dead cells from analysis, 7-AAD viability dye (eBioscience) was added prior to analysis, and 7-AAD+ cells were not analyzed. Cells were analyzed for fluorescence expression using a Beckman Coulter Gallios flow cytometer (Brea, CA). Data were analyzed using FlowJo Software (Ashland, OR).

Generation of chemotherapy-resistant canine cell lines.

Chemotherapy-resistant cell lines were generated by culturing tumor cells in the presence of a mixture of 3 of the 4 cytotoxic chemotherapy drugs (doxorubicin, vincristine, and dexamethasone) which comprise the CHOP cocktail, with the exception of cyclophosphamide8,9. The concentrations of each drug in the mixture was as follows: CLBL1 cells: 845 ng/ml dexamethasone, 3 ng/ml vincristine, and 80 ug/ml dexamethasone and Oswald cells: 30 ng/ml doxorubicin, 125 ng/ml vincristine, and 80 ug/ml dexamethasone). These concentrations were selected based on IC50 values for each drug against the individual lymphoma cell lines, as determined using in vitro cell survival assays. Selection of drug-resistant cell lines was done by treating cell lines with chemotherapy-selection drugs, with a minimum of 80% cell death achieved after the first chemo-selection treatment. Drug selection continued for 14 days until the cells were considered fully resistant.

Immunohistochemistry.

Lymph node biopsies were harvested and fixed in periodate-lysine-paraformaldehyde (PLP) for 24 hours before transferring to a 30% glucose solution for another 24 hours, all at 4C. Afterwards, tissues were embedded in OCT (Optimal Cutting Temperature compound), frozen at −80C, and cryosectioned to a thickness of 5 microns. The tissues were mounted on glass slides and blocked with 5% donkey serum for 10 minutes prior to staining. Tissues were stained with unlabeled CD117 antibody or its matched isotype control for 1 hour at room temperature, followed by donkey-anti-rat FITC for 1 hour at room temperature. The tissues were then permeabilized with 0.025% saponin for 1 hour at room temperature and stained with unlabeled Oct3/4 antibody or its matched isotype control overnight at 4C. This was followed by donkey anti-rat Cy3 for 1 hour at room temperature. Finally, the tissues were stained with DAPI to identify nucleated cells and coverslipped with Prolong Diamond mounting media (LifeTech, Carlsbad, CA). Images were acquired on a confocal microscope and exposure times were set according to each tissue’s matched isotype stain. Figures were then compiled using Adobe Photoshop.

Tumor sphere assay.

Matched numbers of untreated and chemotherapy-resistant Oswald cells were plated in low-adherence petri dishes (Falcon, Corning, NY) and maintained in cell culture medium alone or medium with chemotherapy. Images of the cells were then acquired with a light microscope after 3 days.

ALDH assay.

An Aldefluor kit (Stemcell Technologies, Vancouver, Canada) was used to measure ALDH activity in untreated and chemotherapy-resistant lymphoma cells. Verapamil (Sigma-Aldrich) was used to prevent export of the fluorescent Aldefluor reagent, and DEAB (Stemcell Technologies) was added immediately to the negative control wells to quench the ALDH activity. 7-AAD (eBioscience) was added prior to flow cytometric analysis for dead cell exclusion.

Doxorubicin efflux assay.

Efflux of doxorubicin by untreated and chemotherapy-resistant Oswald cells was measured by flow cytometry, as described previously57. Briefly, cells were re-suspended in medium alone, medium with 2 mg/ml Doxorubicin, or medium with 2 mg/ml Doxorubicin and 50 ug/ml Verapamil for a positive control of Doxorubicin inside the cells. After 1 hour at 37C, cells were washed with medium alone or medium with Verapamil, allowed to rest for 1.5 hours at 37C, washed a second time, then analyzed by flow cytometry.

Statistical analysis.

Statistical comparisons between those data sets with two sample groups were done using non-parametric t-tests (Mann-Whitney test). Comparisons between 3 or more groups were done using ANOVA, followed by Tukey multiple means post-test. Analyses were done using Prism7 software (GraphPad, La Jolla, CA) and statistical significance was determined for p < 0.05.

Results.

Expression of CSC molecules by healthy and malignant lymphocytes.

Malignant canine lymphocytes have been reported to exhibit a high forward scatter and medium side scatter properties58. These criteria were therefore used to identify malignant lymphocytes in dogs with BCL and TCL56. The malignant tumor cells were further classified as CD5+ T cells or CD21+ B cells (Figure 1A), using flow cytometry. Normal T and B cells were defined as CD5+ or CD21+ cells, respectively, when evaluating CSC populations in lymph nodes of healthy dogs. Tumor-infiltrating lymphocytes were defined as CD5+ T cells in tumors of dogs with B cell lymphoma. Expression of CSC markers by the gated populations of malignant B and T cells, as well as by normal B and T cells, was determined by analysis of flow cytometry data and was defined as percentage of CSC+ cells per overall cell population.

Figure 1. Cancer stem cell marker expression by normal and malignant canine lymph node B and T cells.

Figure 1.

Fine needle aspirates were obtained from lymph nodes of 13 normal dogs, 35 dogs with untreated BCL, and 9 dogs with untreated TCL for flow cytometric analysis. The cells were stained for CD21 and CD5 to distinguish between B and T cells, and positive staining for cancer stem cell markers (CD29, CD34, CD90, CD117, Sca1, CD133, and Oct3/4) was determined with respect to cells stained with irrelevant isotype antibody. The data were analyzed using FlowJo Software and the gating strategy is shown in (A). Data shown represents B) normal lymph node B cells, (C) malignant B cells from untreated BCL, D) normal lymph node T cells, and (E) malignant T cells from untreated TCL. Data are presented as mean ± SEM of the percentage of marker expression on lymphocytes.

When expression of CSC markers by malignant B cells in dogs with BCL was compared to expression by healthy B cells in normal lymph nodes, we found that amongst the population of malignant B cells, there was a significantly higher percentage of cells that expressed CSC markers CD34, CD90, CD117, and Oct3/4, compared to the percentages of healthy canine B cells expressing these stem cell markers (Figure 1B and 1C). The greatest percentages of CSC expression by malignant B cells occurred in the CD90, CD117, and Oct3/4 positive populations.

In dogs with TCL, malignant T cells had upregulated intracellular expression of Oct3/4 compared to normal T cells, whereas there was not an overall increase the in the percentages of malignant T cells expressing the other CSC molecules evaluated (Figure 1D and 1E). Interestingly, expression of the stem cell associated marker CD90 was downregulated on malignant T cells, compared to healthy T cells.

Thus, within both BCL and TCL, there existed cell subpopulations with phenotypic characteristics consistent with CSC. Notably, the CSC subpopulations found in BCL were very different from those present in TCL, with essentially no overlap. These findings suggest distinct pathways for the origin of CSC in these two types of lymphoma.

Immunohistochemical (IHC) evaluation of CSC populations in tumor tissues.

Lymph node biopsies from 3 normal dogs and from 3 dogs with BCL were evaluated using IHC for expression of CD117 (Figure 2A) and Oct3/4 (Figure 2B). In normal lymph nodes from 3 dogs, expression of neither marker could be detected using IHC. (It should be noted that IHC is generally less sensitive for detection of antibody binding than flow cytometry). However, in tumor tissues of one dog with BCL, CSC expressing CD117 and Oct3/4 could be detected, whereas in the other two tumor samples, expression was not detected using IHC.

Figure 2. Expression of CD117 and Oct3/4 by normal and malignant canine lymph nodes.

Figure 2.

Biopsies were obtained from the lymph nodes of 3 normal dogs and 3 dogs with BCL and prepared for immunofluorescent staining. The tissues were stained with antibodies against CD117 (A) and Oct3/4 (B), with positive staining determined with respect to isotype antibodystained tissues for each dog. Positive staining is shown in red, and the cells were counterstained with DAPI for nuclear detection as noted in Methods. Images were acquired with a confocal microscope, and images shown are representative images of one normal dog and one dog with BCL.

Effects of chemotherapy on in vitro CSC selection.

Previous studies have found that the percentages of CSC in certain cancers (breast, colorectal, and lung) is significantly increased following treatment with cytotoxic chemotherapy5961. This phenomenon is thought to reflect selection for chemotherapy resistant CSC, and the expanded CSC populations are associated with an overall decrease in responsiveness to subsequent chemotherapy62,63.

Studies were designed therefore to determine in vitro whether a similar response occurs in canine lymphoma following exposure to cytotoxic chemotherapy. To address this question, two canine lymphoma cell lines, one B cell (1771) and one T cell (Oswald) were exposed to high combined doses of cytotoxic chemotherapy drugs to select for chemotherapy resistant cells. The CSC phenotype of chemotherapy-resistant BCL and TCL cells was then compared to the phenotype of chemotherapy sensitive cells.

Following chemotherapy selection, there was a significant upregulation of expression of multiple important CSC associated molecules, including CD29, CD34, CD90, Oct3/4, Sca1, and CD133, by chemotherapy resistant BCL cells (1771), compared to unselected cells (Figure 3A–C). The most highly upregulated molecules were CD29 and CD34 in BCL cells. Similarly, upregulated expression of the CSC markers CD29, CD34, CD90, and Sca1 was noted in chemotherapy resistant TCL cells (OSW), with CD90 and CD34 exhibiting the highest degree of upregulation (Figure 3D).

Figure 3. Impact of chemotherapy resistance on CSC populations within lymphoma cell lines.

Figure 3.

Canine BCL cell lines CLBL1 and 1771 as well as the TCL cell line Oswald were selected for chemotherapy resistance using 4 times IC50 doses of doxorubicin, vincristine, and dexamethasone for 2 months. Untreated and chemotherapy-resistant cells were then immunostained for expression of cancer stem cell markers (CD29, CD34, CD90, CD117, Oct3/4, Sca1, and CD133). Cells were also immunostained with an isotype-matched irrelevant antibody. Representative histograms depict geometric mean fluorescence intensity (MFI) plots for 1771 cells (A). The percentage positive expression of noted CSC markers is shown for CLBL1 (B), 1771 (C), and Oswald cells (D). Each experiment was conducted with 3 separately selected chemotherapy-resistant cell lines, with similar results each time.

Next, chemotherapy-resistant and -sensitive cells were evaluated for functional changes associated with CSC properties. These functional characteristics included tumor sphere formation in non-adherent culture conditions, doxorubicin efflux activity, and aldehyde dehydrogenase activity. We observed that chemotherapy-resistant Oswald cells formed spheres much more readily in soft agar than chemotherapy-sensitive cells, suggesting a higher self-renewal capacity (Figure 4A)45. These chemotherapy resistant Oswald cells also had significantly increased ALDH activity (Figure 4B). The ALDH activity is important because ALDH enzyme is important for detoxifying and metabolizing chemotherapeutic drugs, and maintaining overall cell “stemness”4749. Chemotherapy selected Oswald cells also effluxed doxorubicin much more efficiently than chemotherapy-sensitive Oswald cells, consistent also with induction of efflux pump expression, another feature of CSC64,65(Figure 4C).

Figure 4. Cancer stem cell properties of a chemotherapy-resistant lymphoma cell line.

Figure 4.

The Oswald line was selected over 2 months for chemotherapy-resistant cells. A) Identical numbers of chemotherapy sensitive and resistant cells were plated and images were obtained after 3 days of initial culture to demonstrate colony formation: chemotherapy sensitive cells (left), chemotherapy-resistant cells (right). An aldefluor assay was used to measure ALDH activity by chemotherapy sensitive and resistant cells (B) and efflux of doxorubicin was measured in (C). Data are presented as mean ± SEM of the percentage of ALDH+ cells in (B) and fold change of fluorescence intensity in (C). Statistical analysis was performed using Student’s t-test and statistically significant differences were denoted as * = p < 0.05 and **** = p < 0.0001. These experiments were repeated 3 times, with similar results each time.

Upregulated ALDH activity and efflux of doxorubicin were also observed in chemotherapy-resistant BCL cells (1771) compared to chemotherapy-sensitive cells. However, unlike the case with Oswald cells, the chemotherapy resistant 1771 cells did not acquire the ability to form spheres in culture (data not shown).

Overall, these results indicate that not only do cytotoxic chemotherapeutics select for upregulated expression of CSC markers, but they also select for TCL and BCL cells with the functional properties of CSC. Thus, these results predict that relapsed lymphoma cells resistant to chemotherapy in dogs following relapse are likely to exhibit accentuated CSC characteristics.

Cancer stem cell marker expression by naïve and relapsed lymphoma.

Indeed, when CSC marker expression was evaluated in dogs with relapsed BCL, and compared to CSC marker expression with dogs with untreated BCL, there were increased numbers of CSC in relapsed dogs (Figure 5A). For example, 33% of tumor cells in relapsed dogs expressed the CSC marker Oct3/4, compared to 15% expression by tumor cells in dogs with naive BCL. These preliminary findings suggest that examination of larger numbers of animals with relapsed BCL would reveal expanded populations of CSC, relative to treatment naive tumors. Such a response might elucidate an important mechanism by which canine BCL become resistant to chemotherapy, including different classes of cytotoxic agents.

Figure 5. Cancer stem cell marker expression by normal lymphocytes, untreated lymphoma cells, and relapsed lymphoma cells.

Figure 5.

Fine needle aspirates were collected from lymph nodes of normal dogs and from lymph nodes of dogs with BCL or TCL (untreated and relapsed animals) for flow cytometric analysis of CSC subpopulations. Cells were initially immunostained to identify CD21+ B cells and CD5+ T cells, as shown in Figure 1, Expression of cancer stem cell markers CD29, CD34, CD90, CD117, Oct3/4, Sca1, and CD133 was determined with respect to the level of antibody binding in cells immunostained with irrelevant isotype-matched antibodies. Flow cytometry data were analyzed using FlowJo Software. The data depicted were generated from 12 healthy dogs, 35 dogs with untreated BCL, 9 dogs with untreated TCL, 9 dogs with relapsed BCL, and 2 dogs with relapsed TCL. (YOU SHOULD REPLOT GRAPH B WITHOUT THE RELAPSED TCL DOGS, SINCE YOU CANT PLOT SD WITH ONLY 2 SAMPLES) Expression of CSC markers by normal and malignant B cells is depicted in (A). In (B), CSC marker expression by normal and malignant T cells is depicted (B). Data are presented as mean ± SEM of the percentage of CSC marker expression on lymphocytes, and statistical analysis was performed using two-tailed ANOVA (excluding relapsed TCL patients), followed by Tukey’s multiple means comparison. Statistically significant differences were denoted as * = p < 0.05, ** = p < 0.005, and **** = p < 0.0001.

Discussion.

There are a number of mechanisms invoked to account for lymphoma relapse following chemotherapy, including intrinsic drug resistance, or induced upregulation of drug efflux pumps67,68, drug metabolizing enzymes69, and drug-induced DNA damage repair mechanisms70. Selective enrichment of CSC has also been invoked as an explanation for tumor relapse in lymphoma, based on evidence from experimental rodent models and human trials26,66,7174.

The studies reported here are the first to our knowledge to define the various subpopulations of CSC present in canine lymphoma and to compare the CSC subpopulations in BCL versus TCL in dogs. The key findings reported here were that in dogs with BCL, the primary CSC subpopulations identified were CD34+, CD90+, CD117+, and Oct3/4+ (Figure 1). In TCL, the most prominent CSC subpopulation was Oct3/4+ (Figure 1). Though there is some overlap between normal and malignant B and T cells with respect to CSC marker expression, in general the majority of CSC markers were upregulated by malignant cells.

Cytotoxic chemotherapy was found to strongly select for CSC enrichment by cultured lymphoma cell lines. For example, in chemotherapy resistant canine BLC tumor cell lines, there was strong selection for upregulated expression of multiple CSC markers, including CD29, CD34, CD90, CD117, Oct3/4, Sca1, and CD133. In the case of TCL lines, upregulated expression of the CSC markers CD29, CD34, CD90, and Sca1 was observed (Figure 3).

Moreover, in dogs with relapsed BCL that relapsed following chemotherapy, there was substantial upregulation of CSC populations, compared to treatment naive tumors (Figure 5). Thus, canine lymphoma appears to respond to chemotherapy in a manner similar to that of human lymphoma, with selective enrichment for CSC.

Previous studies have identified canine BCL cell lines cells with increased expression levels of ABC transporters and genes associated with the CSC phenotype, including ABCG2 and Bmi-175. In addition, function studies of these cell lines have demonstrated the presence of a side-population of cells characteristic of CSC, and have also demonstrated efflux of DNA specific dyes75. However, these previous studies have not compared the CSC phenotype of normal and malignant canine B cells and T cells. Nor have previous studies demonstrated how selection for chemotherapy resistance leads to enrichment of tumor cells with a CSC phenotype and CSC functional properties.

The functional properties of CSC not only contribute to longevity, but they also render these cells more resistant to radiation treatment and chemotherapy drugs, allowing them to evade the effects of cytotoxic chemotherapy76,77. Thus, it would be very desirable to identify drugs or other agents that could selectively target CSC for elimination. The ability to selectively enrich for lymphoma CSC using in vitro chemotherapy selection may provide one tool for identification of agents that more selectively target CSC. Furthermore, the ability to accurately phenotype CSC subpopulations in naive tumor samples using flow cytometry may provide a useful prognostic markers for predicting initial responses to chemotherapy as well as overall remission duration.

Supplementary Material

1

Acknowledgements.

These studies were supported by an award from the Shipley Foundation and by a grant from the NIH (grant P30-CA046934)

References.

  • 1.Ito D, Frantz AM & Modiano JF Canine lymphoma as a comparative model for human non-Hodgkin lymphoma: recent progress and applications. Veterinary immunology and immunopathology 159, 192–201, doi: 10.1016/j.vetimm.2014.02.016 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Seelig DM, Avery AC, Ehrhart EJ & Linden MA The Comparative Diagnostic Features of Canine and Human Lymphoma. Vet Sci 3, doi: 10.3390/vetsci3020011 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Williams MJ, Avery AC, Lana SE, Hillers KR, Bachand AM & Avery PR Canine lymphoproliferative disease characterized by lymphocytosis: immunophenotypic markers of prognosis. J Vet Intern Med 22, 596–601, doi: 10.1111/j.1939-1676.2008.0041.x (2008). [DOI] [PubMed] [Google Scholar]
  • 4.Simon D, Nolte I, Eberle N, Abbrederis N, Killich M & Hirschberger J Treatment of dogs with lymphoma using a 12-week, maintenance-free combination chemotherapy protocol. J Vet Intern Med 20, 948–954 (2006). [DOI] [PubMed] [Google Scholar]
  • 5.Garrett LD, Thamm DH, Chun R, Dudley R & Vail DM Evaluation of a 6-month chemotherapy protocol with no maintenance therapy for dogs with lymphoma. J Vet Intern Med 16, 704–709 (2002). [DOI] [PubMed] [Google Scholar]
  • 6.Marconato L, Gelain ME & Comazzi S The dog as a possible animal model for human non-Hodgkin lymphoma: a review. Hematol Oncol 31, 1–9, doi: 10.1002/hon.2017 (2013). [DOI] [PubMed] [Google Scholar]
  • 7.Jain T, Croswell C, Urday-Cornejo V, Awali R, Cutright J, Salimnia H, Reddy Banavasi HV, Liubakka A, Lephart P, Chopra T et al. Clostridium Difficile Colonization in Hematopoietic Stem Cell Transplant Recipients: A Prospective Study of the Epidemiology and Outcomes Involving Toxigenic and Nontoxigenic Strains. Biol Blood Marrow Transplant 22, 157–163, doi: 10.1016/j.bbmt.2015.07.020 (2016). [DOI] [PubMed] [Google Scholar]
  • 8.Johnston SA, Thamm DH & Legutki JB The immunosignature of canine lymphoma: characterization and diagnostic application. BMC Cancer 14, 657, doi: 10.1186/1471-2407-14-657 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Regan RC, Kaplan MS & Bailey DB Diagnostic evaluation and treatment recommendations for dogs with substage-a high-grade multicentric lymphoma: results of a survey of veterinarians. Veterinary and comparative oncology 11, 287–295, doi: 10.1111/j.1476-5829.2012.00318.x (2013). [DOI] [PubMed] [Google Scholar]
  • 10.Malanchi I, Santamaria-Martinez A, Susanto E, Peng H, Lehr HA, Delaloye JF & Huelsken J Interactions between cancer stem cells and their niche govern metastatic colonization. Nature 481, 85–89, doi: 10.1038/nature10694 (2012). [DOI] [PubMed] [Google Scholar]
  • 11.O’Hare T, Corbin AS & Druker BJ Targeted CML therapy: controlling drug resistance, seeking cure. Curr Opin Genet Dev 16, 92–99, doi: 10.1016/j.gde.2005.11.002 (2006). [DOI] [PubMed] [Google Scholar]
  • 12.Li X, Lewis MT, Huang J, Gutierrez C, Osborne CK, Wu MF, Hilsenbeck SG, Pavlick A, Zhang X, Chamness GC et al. Intrinsic resistance of tumorigenic breast cancer cells to chemotherapy. J Natl Cancer Inst 100, 672–679, doi: 10.1093/jnci/djn123 (2008). [DOI] [PubMed] [Google Scholar]
  • 13.Driessens G, Beck B, Caauwe A, Simons BD & Blanpain C Defining the mode of tumour growth by clonal analysis. Nature 488, 527–530, doi: 10.1038/nature11344 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Reya T, Morrison SJ, Clarke MF & Weissman IL Stem cells, cancer, and cancer stem cells. Nature 414, 105–111, doi: 10.1038/35102167 (2001). [DOI] [PubMed] [Google Scholar]
  • 15.Allan AL, Vantyghem SA, Tuck AB & Chambers AF Tumor dormancy and cancer stem cells: implications for the biology and treatment of breast cancer metastasis. Breast Dis 26, 87–98 (2006). [DOI] [PubMed] [Google Scholar]
  • 16.Li F, Tiede B, Massague J & Kang Y Beyond tumorigenesis: cancer stem cells in metastasis. Cell Res 17, 3–14, doi: 10.1038/sj.cr.7310118 (2007). [DOI] [PubMed] [Google Scholar]
  • 17.Shah A, Patel S, Pathak J, Swain N & Kumar S The evolving concepts of cancer stem cells in head and neck squamous cell carcinoma. ScientificWorldJournal 2014, 842491, doi: 10.1155/2014/842491 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Krishnamurthy S & Nor JE Head and neck cancer stem cells. J Dent Res 91, 334–340, doi: 10.1177/0022034511423393 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Rapp UR, Ceteci F & Schreck R Oncogene-induced plasticity and cancer stem cells. Cell Cycle 7, 45–51, doi: 10.4161/cc.7.1.5203 (2008). [DOI] [PubMed] [Google Scholar]
  • 20.Hermann PC, Huber SL, Herrler T, Aicher A, Ellwart JW, Guba M, Bruns CJ & Heeschen C Distinct populations of cancer stem cells determine tumor growth and metastatic activity in human pancreatic cancer. Cell stem cell 1, 313–323, doi: 10.1016/j.stem.2007.06.002 (2007). [DOI] [PubMed] [Google Scholar]
  • 21.Chen J, Li Y, Yu TS, McKay RM, Burns DK, Kernie SG & Parada LF A restricted cell population propagates glioblastoma growth after chemotherapy. Nature 488, 522–526, doi: 10.1038/nature11287 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Skubitz AP, Taras EP, Boylan KL, Waldron NN, Oh S, Panoskaltsis-Mortari A & Vallera DA Targeting CD133 in an in vivo ovarian cancer model reduces ovarian cancer progression. Gynecol Oncol 130, 579–587, doi: 10.1016/j.ygyno.2013.05.027 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Stratford EW, Bostad M, Castro R, Skarpen E, Berg K, Hogset A, Myklebost O & Selbo PK Photochemical internalization of CD133-targeting immunotoxins efficiently depletes sarcoma cells with stem-like properties and reduces tumorigenicity. Biochim Biophys Acta 1830, 4235–4243, doi: 10.1016/j.bbagen.2013.04.033 (2013). [DOI] [PubMed] [Google Scholar]
  • 24.Marangoni E, Lecomte N, Durand L, de Pinieux G, Decaudin D, Chomienne C, Smadja-Joffe F & Poupon MF CD44 targeting reduces tumour growth and prevents post-chemotherapy relapse of human breast cancers xenografts. British journal of cancer 100, 918–922, doi: 10.1038/sj.bjc.6604953 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Willingham SB, Volkmer JP, Gentles AJ, Sahoo D, Dalerba P, Mitra SS, Wang J, Contreras-Trujillo H, Martin R, Cohen JD et al. The CD47-signal regulatory protein alpha (SIRPa) interaction is a therapeutic target for human solid tumors. Proceedings of the National Academy of Sciences of the United States of America 109, 6662–6667, doi: 10.1073/pnas.1121623109 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Lathia JD & Liu H Overview of Cancer Stem Cells and Stemness for Community Oncologists. Target Oncol 12, 387–399, doi: 10.1007/s11523-017-0508-3 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Dawood S, Austin L & Cristofanilli M Cancer stem cells: implications for cancer therapy. Oncology (Williston Park) 28, 1101–1107, 1110 (2014). [PubMed] [Google Scholar]
  • 28.Lee CG, Das B, Lin TL, Grimes C, Zhang X, Lavezzi T, Huang L, Cole J, Yau L & Li L A rare fraction of drug-resistant follicular lymphoma cancer stem cells interacts with follicular dendritic cells to maintain tumourigenic potential. Br J Haematol 158, 79–90, doi: 10.1111/j.1365-2141.2012.09123.x (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Blair A & Sutherland HJ Primitive acute myeloid leukemia cells with long-term proliferative ability in vitro and in vivo lack surface expression of c-kit (CD117). Exp Hematol 28, 660–671 (2000). [DOI] [PubMed] [Google Scholar]
  • 30.Jordan CT, Upchurch D, Szilvassy SJ, Guzman ML, Howard DS, Pettigrew AL, Meyerrose T, Rossi R, Grimes B, Rizzieri DA et al. The interleukin-3 receptor alpha chain is a unique marker for human acute myelogenous leukemia stem cells. Leukemia 14, 1777–1784 (2000). [DOI] [PubMed] [Google Scholar]
  • 31.Brendel C, Mohr B, Schimmelpfennig C, Muller J, Bornhauser M, Schmidt M, Ritter M, Ehninger G & Neubauer A Detection of cytogenetic aberrations both in CD90 (Thy-1)-positive and (Thy-1)-negative stem cell (CD34) subfractions of patients with acute and chronic myeloid leukemias. Leukemia 13, 1770–1775 (1999). [DOI] [PubMed] [Google Scholar]
  • 32.Jones RJ, Gocke CD, Kasamon YL, Miller CB, Perkins B, Barber JP, Vala MS, Gerber JM, Gellert LL, Siedner M et al. Circulating clonotypic B cells in classic Hodgkin lymphoma. Blood 113, 5920–5926, doi: 10.1182/blood-2008-11-189688 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Jung HJ, Chen Z & McCarty N Stem-like tumor cells confer drug resistant properties to mantle cell lymphoma. Leuk Lymphoma 52, 1066–1079, doi: 10.3109/10428194.2011.562570 (2011). [DOI] [PubMed] [Google Scholar]
  • 34.Lapidot T, Sirard C, Vormoor J, Murdoch B, Hoang T, Caceres-Cortes J, Minden M, Paterson B, Caligiuri MA & Dick JE A cell initiating human acute myeloid leukaemia after transplantation into SCID mice. Nature 367, 645–648, doi: 10.1038/367645a0 (1994). [DOI] [PubMed] [Google Scholar]
  • 35.Blair A, Hogge DE, Ailles LE, Lansdorp PM & Sutherland HJ Lack of expression of Thy-1 (CD90) on acute myeloid leukemia cells with long-term proliferative ability in vitro and in vivo. Blood 89, 3104–3112 (1997). [PubMed] [Google Scholar]
  • 36.Buccisano F, Rossi FM, Venditti A, Del Poeta G, Cox MC, Abbruzzese E, Rupolo M, Berretta M, Degan M, Russo S et al. CD90/Thy-1 is preferentially expressed on blast cells of high risk acute myeloid leukaemias. Br J Haematol 125, 203–212, doi: 10.1111/j.1365-2141.2004.04883.x (2004). [DOI] [PubMed] [Google Scholar]
  • 37.Yang ZF, Ho DW, Ng MN, Lau CK, Yu WC, Ngai P, Chu PW, Lam CT, Poon RT & Fan ST Significance of CD90+ cancer stem cells in human liver cancer. Cancer cell 13, 153–166, doi: 10.1016/j.ccr.2008.01.013 (2008). [DOI] [PubMed] [Google Scholar]
  • 38.Tang KH, Dai YD, Tong M, Chan YP, Kwan PS, Fu L, Qin YR, Tsao SW, Lung HL, Lung ML et al. A CD90(+) tumor-initiating cell population with an aggressive signature and metastatic capacity in esophageal cancer. Cancer research 73, 2322–2332, doi: 10.1158/0008-5472.CAN-12-2991 (2013). [DOI] [PubMed] [Google Scholar]
  • 39.Hosen N, Park CY, Tatsumi N, Oji Y, Sugiyama H, Gramatzki M, Krensky AM & Weissman IL CD96 is a leukemic stem cell-specific marker in human acute myeloid leukemia. Proceedings of the National Academy of Sciences of the United States of America 104, 11008–11013, doi: 10.1073/pnas.0704271104 (2007). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Du W, Li XE, Sipple J & Pang Q Overexpression of IL-3Ralpha on CD34+CD38-stem cells defines leukemia-initiating cells in Fanconi anemia AML. Blood 117, 4243–4252, doi: 10.1182/blood-2010-09-309179 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Jaiswal S, Jamieson CH, Pang WW, Park CY, Chao MP, Majeti R, Traver D, van Rooijen N & Weissman IL CD47 is upregulated on circulating hematopoietic stem cells and leukemia cells to avoid phagocytosis. Cell 138, 271–285, doi: 10.1016/j.cell.2009.05.046 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Liesveld JL, Dipersio JF & Abboud CN Integrins and adhesive receptors in normal and leukemic CD34+ progenitor cells: potential regulatory checkpoints for cellular traffic. Leuk Lymphoma 14, 19–28, doi: 10.3109/10428199409049647 (1994). [DOI] [PubMed] [Google Scholar]
  • 43.Moshaver B, van Rhenen A, Kelder A, van der Pol M, Terwijn M, Bachas C, Westra AH, Ossenkoppele GJ, Zweegman S & Schuurhuis GJ Identification of a small subpopulation of candidate leukemia-initiating cells in the side population of patients with acute myeloid leukemia. Stem cells 26, 3059–3067, doi: 10.1634/stemcells.2007-0861 (2008). [DOI] [PubMed] [Google Scholar]
  • 44.Ponti D, Costa A, Zaffaroni N, Pratesi G, Petrangolini G, Coradini D, Pilotti S, Pierotti MA & Daidone MG Isolation and in vitro propagation of tumorigenic breast cancer cells with stem/progenitor cell properties. Cancer research 65, 5506–5511, doi: 10.1158/0008-5472.CAN-05-0626 (2005). [DOI] [PubMed] [Google Scholar]
  • 45.Lee CH, Yu CC, Wang BY & Chang WW Tumorsphere as an effective in vitro platform for screening anti-cancer stem cell drugs. Oncotarget 7, 1215–1226, doi: 10.18632/oncotarget.6261 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Lohberger B, Rinner B, Stuendl N, Absenger M, Liegl-Atzwanger B, Walzer SM, Windhager R & Leithner A Aldehyde dehydrogenase 1, a potential marker for cancer stem cells in human sarcoma. PloS one 7, e43664, doi: 10.1371/journal.pone.0043664 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Magni M, Shammah S, Schiro R, Mellado W, Dalla-Favera R & Gianni AM Induction of cyclophosphamide-resistance by aldehyde-dehydrogenase gene transfer. Blood 87, 1097–1103 (1996). [PubMed] [Google Scholar]
  • 48.Marchitti SA, Brocker C, Stagos D & Vasiliou V Non-P450 aldehyde oxidizing enzymes: the aldehyde dehydrogenase superfamily. Expert Opin Drug Metab Toxicol 4, 697–720, doi: 10.1517/17425255.4.6.697 (2008). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Ginestier C, Wicinski J, Cervera N, Monville F, Finetti P, Bertucci F, Wicha MS, Birnbaum D & Charafe-Jauffret E Retinoid signaling regulates breast cancer stem cell differentiation. Cell Cycle 8, 3297–3302, doi: 10.4161/cc.8.20.9761 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Guth AM, Deogracias M & Dow SW Comparison of cancer stem cell antigen expression by tumor cell lines and by tumor biopsies from dogs with melanoma and osteosarcoma. Veterinary immunology and immunopathology 161, 132–140, doi: 10.1016/j.vetimm.2014.07.006 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Rutgen BC, Hammer SE, Gerner W, Christian M, de Arespacochaga AG, Willmann M, Kleiter M, Schwendenwein I & Saalmuller A Establishment and characterization of a novel canine B-cell line derived from a spontaneously occurring diffuse large cell lymphoma. Leuk Res 34, 932–938, doi: 10.1016/j.leukres.2010.01.021 (2010). [DOI] [PubMed] [Google Scholar]
  • 52.Steplewski Z, Jeglum KA, Rosales C & Weintraub N Canine lymphoma-associated antigens defined by murine monoclonal antibodies. Cancer immunology, immunotherapy : CII 24, 197–201 (1987). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Kisseberth WC, Nadella MV, Breen M, Thomas R, Duke SE, Murahari S, Kosarek CE, Vernau W, Avery AC, Burkhard MJ et al. A novel canine lymphoma cell line: a translational and comparative model for lymphoma research. Leuk Res 31, 1709–1720, doi: 10.1016/j.leukres.2007.04.003 (2007). [DOI] [PubMed] [Google Scholar]
  • 54.Fowles JS, Dailey DD, Gustafson DL, Thamm DH & Duval DL The Flint Animal Cancer Center (FACC) Canine Tumour Cell Line Panel: a resource for veterinary drug discovery, comparative oncology and translational medicine. Veterinary and comparative oncology, doi: 10.1111/vco.12192 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.O’Donoghue LE, Rivest JP & Duval DL Polymerase chain reaction-based species verification and microsatellite analysis for canine cell line validation. Journal of veterinary diagnostic investigation : official publication of the American Association of Veterinary Laboratory Diagnosticians, Inc 23, 780–785, doi: 10.1177/1040638711408064 (2011). [DOI] [PubMed] [Google Scholar]
  • 56.Hartley G, Elmslie R, Dow S & Guth A Checkpoint molecule expression by B and T cell lymphomas in dogs. Veterinary and comparative oncology, doi: 10.1111/vco.12386 (2018). [DOI] [PubMed] [Google Scholar]
  • 57.Hu C, Li H, Li J, Zhu Z, Yin S, Hao X, Yao M, Zheng S & Gu J Analysis of ABCG2 expression and side population identifies intrinsic drug efflux in the HCC cell line MHCC-97L and its modulation by Akt signaling. Carcinogenesis 29, 2289–2297, doi: 10.1093/carcin/bgn223 (2008). [DOI] [PubMed] [Google Scholar]
  • 58.Lana S, Plaza S, Hampe K, Burnett R & Avery AC Diagnosis of mediastinal masses in dogs by flow cytometry. J Vet Intern Med 20, 1161–1165 (2006). [DOI] [PubMed] [Google Scholar]
  • 59.Dylla SJ, Beviglia L, Park IK, Chartier C, Raval J, Ngan L, Pickell K, Aguilar J, Lazetic S, Smith-Berdan S et al. Colorectal cancer stem cells are enriched in xenogeneic tumors following chemotherapy. PloS one 3, e2428, doi: 10.1371/journal.pone.0002428 (2008). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Creighton CJ, Li X, Landis M, Dixon JM, Neumeister VM, Sjolund A, Rimm DL, Wong H, Rodriguez A, Herschkowitz JI et al. Residual breast cancers after conventional therapy display mesenchymal as well as tumor-initiating features. Proceedings of the National Academy of Sciences of the United States of America 106, 13820–13825, doi: 10.1073/pnas.0905718106 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Freitas DP, Teixeira CA, Santos-Silva F, Vasconcelos MH & Almeida GM Therapy-induced enrichment of putative lung cancer stem-like cells. Int J Cancer 134, 1270–1278, doi: 10.1002/ijc.28478 (2014). [DOI] [PubMed] [Google Scholar]
  • 62.Tehranchi R, Woll PS, Anderson K, Buza-Vidas N, Mizukami T, Mead AJ, Astrand-Grundstrom I, Strombeck B, Horvat A, Ferry H et al. Persistent malignant stem cells in del(5q) myelodysplasia in remission. The New England journal of medicine 363, 1025–1037, doi: 10.1056/NEJMoa0912228 (2010). [DOI] [PubMed] [Google Scholar]
  • 63.Saito Y, Uchida N, Tanaka S, Suzuki N, Tomizawa-Murasawa M, Sone A, Najima Y, Takagi S, Aoki Y, Wake A et al. Induction of cell cycle entry eliminates human leukemia stem cells in a mouse model of AML. Nat Biotechnol 28, 275–280, doi: 10.1038/nbt.1607 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Moitra K, Lou H & Dean M Multidrug efflux pumps and cancer stem cells: insights into multidrug resistance and therapeutic development. Clin Pharmacol Ther 89, 491–502, doi: 10.1038/clpt.2011.14 (2011). [DOI] [PubMed] [Google Scholar]
  • 65.Hirschmann-Jax C, Foster AE, Wulf GG, Nuchtern JG, Jax TW, Gobel U, Goodell MA & Brenner MK A distinct “side population” of cells with high drug efflux capacity in human tumor cells. Proceedings of the National Academy of Sciences of the United States of America 101, 14228–14233, doi: 10.1073/pnas.0400067101 (2004). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Visvader JE & Lindeman GJ Cancer stem cells: current status and evolving complexities. Cell stem cell 10, 717–728, doi: 10.1016/j.stem.2012.05.007 (2012). [DOI] [PubMed] [Google Scholar]
  • 67.Yoshimori M, Takada H, Imadome K, Kurata M, Yamamoto K, Koyama T, Shimizu N, Fujiwara S, Miura O & Arai A P-glycoprotein is expressed and causes resistance to chemotherapy in EBV-positive T-cell lymphoproliferative diseases. Cancer Med 4, 1494–1504, doi: 10.1002/cam4.494 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Izquierdo MA, Neefjes JJ, Mathari AE, Flens MJ, Scheffer GL & Scheper RJ Overexpression of the ABC transporter TAP in multidrug-resistant human cancer cell lines. British journal of cancer 74, 1961–1967 (1996). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Michael M & Doherty MM Drug metabolism by tumours: its nature, relevance and therapeutic implications. Expert Opin Drug Metab Toxicol 3, 783–803, doi: 10.1517/17425255.3.6.783 (2007). [DOI] [PubMed] [Google Scholar]
  • 70.Slupphaug G, Kavli B & Krokan HE The interacting pathways for prevention and repair of oxidative DNA damage. Mutat Res 531, 231–251 (2003). [DOI] [PubMed] [Google Scholar]
  • 71.Majeti R, Chao MP, Alizadeh AA, Pang WW, Jaiswal S, Gibbs KD Jr., van Rooijen N & Weissman IL CD47 is an adverse prognostic factor and therapeutic antibody target on human acute myeloid leukemia stem cells. Cell 138, 286–299, doi: 10.1016/j.cell.2009.05.045 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Dierks C, Beigi R, Guo GR, Zirlik K, Stegert MR, Manley P, Trussell C, Schmitt-Graeff A, Landwerlin K, Veelken H et al. Expansion of Bcr-Abl-positive leukemic stem cells is dependent on Hedgehog pathway activation. Cancer cell 14, 238–249, doi: 10.1016/j.ccr.2008.08.003 (2008). [DOI] [PubMed] [Google Scholar]
  • 73.Zhao C, Chen A, Jamieson CH, Fereshteh M, Abrahamsson A, Blum J, Kwon HY, Kim J, Chute JP, Rizzieri D et al. Hedgehog signalling is essential for maintenance of cancer stem cells in myeloid leukaemia. Nature 458, 776–779, doi: 10.1038/nature07737 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Chao MP, Alizadeh AA, Tang C, Jan M, Weissman-Tsukamoto R, Zhao F, Park CY, Weissman IL & Majeti R Therapeutic antibody targeting of CD47 eliminates human acute lymphoblastic leukemia. Cancer research 71, 1374–1384, doi: 10.1158/0008-5472.CAN-10-2238 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Kim MC, D’Costa S, Suter S & Kim Y Evaluation of a side population of canine lymphoma cells using Hoechst 33342 dye. J Vet Sci 14, 481–486 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Dembinski JL & Krauss S Characterization and functional analysis of a slow cycling stem cell-like subpopulation in pancreas adenocarcinoma. Clinical & experimental metastasis 26, 611–623, doi: 10.1007/s10585-009-9260-0 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Kim SH, Joshi K, Ezhilarasan R, Myers TR, Siu J, Gu C, Nakano-Okuno M, Taylor D, Minata M, Sulman EP et al. EZH2 protects glioma stem cells from radiation-induced cell death in a MELK/FOXM1-dependent manner. Stem cell reports 4, 226–238, doi: 10.1016/j.stemcr.2014.12.006 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

1

RESOURCES