Affiliations of authors: Department of Blood and Marrow Transplantation, Section of Molecular Hematology and Therapy (MS, FCM, JLD CZ, MCH, REC, MA) and Department of Biostatistics and Applied Mathematics (BNB), The University of Texas M. D. Anderson Cancer Center, Houston, TX
Correspondence to: Michael Andreeff, MD, PhD, Department of Blood and Marrow Transplantation, The University of Texas M. D. Anderson Cancer Center, 1515 Holcombe Blvd., Unit 448, Houston, TX 77030 (e-mail: mandreef{at}mdanderson.org)
![]() |
ABSTRACT |
---|
![]() ![]() ![]() ![]() ![]() ![]() ![]() ![]() |
---|
![]() |
INTRODUCTION |
---|
![]() ![]() ![]() ![]() ![]() ![]() ![]() ![]() |
---|
We have developed a therapeutic strategy that uses mesenchymal stem cells (MSC) as cellular vehicles for the targeted delivery and local production of biologic agents in tumors (16). MSC are bone marrowderived non-hematopoietic precursor cells (17,18) that contribute to the maintenance and regeneration of connective tissues through engraftment. MSC can be obtained from bone marrow aspirates, expanded in vitro, and genetically modified for therapeutic strategies in vivo. However, it has become evident that in vivo engraftment is not only an intrinsic function of MSC but also depends on appropriate external signals produced by the tissue microenvironment (16,19,20). These signals are related to the hierarchic organization of tissues with regard to their proliferative and differentiation potentials (21). Tissues that have a high spontaneous turnover, such as skin or gut, continuously replace terminally differentiated epithelial cells from compartments of undifferentiated precursor cells and stem cells. This process involves the proliferation of undifferentiated precursor cells present in situ or possibly their migration from other sites in the organism (2123). By contrast, the turnover of connective tissues is low, and their proliferative potential becomes apparent only when the demand for new functional connective cells increases, such as during wound healing or tissue regeneration after injury. These conditions are characterized by an increased turnover of connective tissues that can possibly also mediate engraftment of bone marrowderived MSC. Indeed, MSC have been shown to contribute to tissue regeneration and to the formation of fibrous scars at the sites of injury (19,23).
Tumors are composed of malignant tumor cells and nonmalignant benign cells. The "benign" tumor compartment includes blood vessels, infiltrating inflammatory cells, and stromal fibroblasts. Stromal fibroblasts provide structural support for malignant cells and influence the behavior and aggressiveness of cancers (24). The formation of tumor stroma closely resembles wound healing and scar formation (25). Malignant cells induce de novo formation of connective tissue in order to provide enough stroma to support cancer growth (26,27). We proposed that signals that mediate increased turnover and proliferation of connective stromal cells in tumors may also mediate the engraftment and proliferation of MSC in tumors (16). MSC engrafted in tumors could potentially serve as delivery vehicles to target anticancer agents to malignant cells. We tested this hypothesis by examining the effects of MSC transduced with adenovirus expressing human IFN- (MSC-IFN-
) against metastatic MDA 231 breast carcinomas and A375SM melanomas in a mouse xenograft model. We wanted to investigate the direct inhibitory effects of IFN-
on tumor cells in vivo, without interference by its immunomodulatory and anti-angiogenic properties. To this end, we tested the effects of human IFN-
, which is species-specific and therefore would not be expected to influence any residual severe combined immunodeficiency (SCID) mouse immune cells or tumor endothelial cells of murine origin (28).
![]() |
MATERIALS AND METHODS |
---|
![]() ![]() ![]() ![]() ![]() ![]() ![]() ![]() |
---|
MSC were isolated from the bone marrow of 10 healthy donors who were undergoing bone marrow harvest for use in allogeneic bone marrow transplantation. All bone marrow donors provided written informed consent, and this study was conducted according to institutional guidelines under an approved protocol. Bone marrow was subjected to centrifugation (700g for 15 minutes at 4 °C), as described (18) over a FicollHypaque gradient (Sigma, St. Louis, MO) to separate mononuclear cells, which were resuspended in alpha-minimal essential medium (-MEM) containing 20% fetal bovine serum (Gibco BRL, Rockville, MD), L-glutamine, and penicillinstreptomycin (Flow Laboratories, Rockville, MD) and plated at an initial density of 1 x 106 cells/cm2. Three days later, the cultures were washed with phosphate-buffered saline (PBS) to remove nonadherent cells, and the remaining monolayers of adherent cells were cultured in fresh medium until they reached confluence. The cells were harvested by trypsinization (0.25% trypsin with 0.1% EDTA), subcultured at densities of 50006000 cells/cm2, and used during the third or fourth passages for experiments. The resulting cells reacted positively with SH2, SH3, and SH4 antibodies, which detect CD105 (Endoglyn) and CD 73 (18), two antigens co-expressed on MSC (a generous gift from Dr. Robert Dean, Osiris, Baltimore, MD), when analyzed by FACScan flow cytometry (Becton-Dickinson, San Jose, CA) (data not shown). The cells also differentiated into adipocytes, chondroblasts, and osteoblasts in commercially available differentiation assays (BioWhittaker, Walkersville, MD) (data not shown). These assays confirmed that we obtained MSC.
Human melanoma A375SM and breast cancer MDA 231 cells were a gift from Dr. I. Fidler (Department of Cancer Biology, M. D. Anderson Cancer Center, Houston, TX). The cells were maintained in -MEM containing 10% fetal calf serum (FCS), sodium pyruvate, nonessential amino acids, L-glutamine, vitamin solution (Life Technologies, Grand Island, NY), and penicillinstreptomycin. Human kidney 293 cells were obtained from American Type Culture Collection (ATCC, Manassas, VA) and cultured in Dulbecco's modified Eagle Medium (DMEM) supplemented with 10% FCS and penicillinstreptomycin.
Adenoviral Vectors and MSC Transduction
We used the AdEasy Adenoviral Vector System (Qbiogene, Carlsbad, CA) to create adenoviral vectors that expressed either Escherichia coli -galactosidase (
-gal) or human IFN-
. Briefly, we subcloned the gene for
-gal into the NotI and HindIII sites of an adenovirus shuttle vector that contains a cytomegalovirus (CMV) promoter (pShuttle CMV) to create pShuttleCMV(
-gal). A plasmid containing the gene for human IFN-
(hIFN-
; obtained from InvivoGen, San Diego, CA) was digested with ClaI, and the 3' overhangs were filled in using DNA polymerase to achieve blunt ends. This product was further digested with BglII to release a 570-base pair fragment containing the human IFN-
gene, which was then subcloned into the BglII and EcoRV sites of pShuttleCMV to create pShuttleCMV(hIFN-
). Both plasmids were sequenced to verify correct reading frames and DNA sequences and then digested with PmeI, dephosphorylated using calf-alkaline phosphatase, extracted twice with buffer-saturated phenolchloroform (1 : 1, vol/vol), and admixed with PacI-digested pAdEasy-1 (Qbiogene), an adenoviral backbone plasmid bearing a gene for kanamycin resistance. These two plasmid combinations (pShuttleCMV(
-gal)/pAdEasy-1 and pShuttleCMV(hIFN-
)/pAdEasy-1) were electroporated into E. coli BJ5183 cells, according to the manufacturer's protocol. The bacteria were plated on kanamycin-containing agar plates, kanamycin-resistant clones were picked, plasmid DNA was extracted, and clones containing the gene product for
-gal or IFN-
were identified by restriction enzyme digestion. Four clones containing each recombinant adenoviral plasmid plus gene product (
-gal or IFN-
) were identified, and theses clones were expanded in a 3-mL bacterial culture. We isolated plasmid DNAs from these cultures and used Fugene6 transfection reagent (Roche, Indianapolis, IA) to transfect them into human kidney 293 cells, according to the manufacturer's instructions. The transfected cells were incubated for 1820 days to allow homologous recombination to occur and recombinant adenoviral plaques to form; recombinant plaques were then picked and eluted in 5 mM TrisHCl (pH 7.8). These supernatants were used to infect 293 cells grown in 24-well culture plates, and the resulting recombinant virus was rescued from the cultures. We performed two rounds of viral amplification; recombinant adenoviruses that expressed human IFN-
(identified with the use of an enzyme-linked immunosorbent assay; Fujirebio, Tokyo, Japan) or
-gal (as detected by staining with 5-bromo-4-chloro-3-indolyl-
-D-galactopyranoside [X-gal]) were then used to transduce MSC. MSC were incubated with each adenovirus at a multiplicity of infection of 3000 for 2 hours. MSC transduced with adenovirus expressing human IFN-
(i.e., MSC-IFN-
) produced 3 x 104 to 4 x 104 IU of IFN-
per 106 MSC during the first 24 hours after infection, and MSC transduced with adenovirus expressing
-gal (i.e., MSC-
-gal) produced X-galpositive staining in greater than 90% of the MSC during the first 24 hours after infection.
Cell Proliferation Assay
Monolayers of MDA 231 or A375SM cells were washed with PBS and the cells were harvested by trypsinization, resuspended in RPMI-1640 medium containing 10% FCS at a concentration of 1.5 x 104 cells/mL, plated into 96-well plates at 3000 cells (200 µL) per well, and incubated overnight at 37 °C to allow the cells to adhere to the plates. We then added fresh medium containing 010 000 IU/mL recombinant IFN- (Avonex, Biogen, Cambridge, MA). Eight wells were used for each dilution. Nine wells from one 96-well plate were subjected to the MTS assay, which measures the number of viable cells (Promega, Madison, WI), at the time of initial addition of IFN-
. Absorbance (at 490 nm) at that time point was denoted as ODDay0. Media with IFN-
were changed daily in the other plates, and the assay was read again after 5 days (ODDay5). In this assay, absorbance at 490 nm is proportional to the number of viable cells in the well. Results were expressed as the percentage of control cell growth (ODDay5 ODDay0)/(ODcontrol ODDay0) x 100, where ODcontrol corresponded to absorbance measured for wells that received medium without IFN-
on day 5.
Flow Cytometry Analysis of MDA 231 Cell or A375SM Cell Co-Cultures With MSC In Vitro
A375SM cells (5 x 104 cells per well) and MDA 231 cells (105 cells per well) were plated in six-well plates alone or mixed with MSC or MSC-IFN- cells, respectively, at a ratio of 10 A375SM cells or MDA 231 cells to 1 MSC-Gal or MSC-IFN-
cell. One plate was used for each experimental condition. After 5 days, the cells were trypsinized, counted, and fixed with 70% ethanol. The cells were then labeled with phycoerythrin (Sigma), and the DNA content of the cells was analyzed with a FACScan flow cytometer (16). The relative numbers of MSC or MSC-IFN-
(diploid cells) and A375SM or MDA 231 tumor cells (aneuploid cells) were determined by using ModFit software, version 2.0 (Verity Software House, Topsham, ME). Results were expressed as the percentage of control cell growth: (the number of tumor cells co-cultured with MSC or with MSC-IFN-
on day 5 the number of tumor cells co-cultured on day 0)/(the number of tumor cells cultured alone on day 5 number of tumor cells cultured alone on day 0) x 100.
Determination of A375SM Cell Death
A375SM melanoma cells were plated in 75-cm2 flasks at a density of 300 000 cells per flask and incubated for 24 hours to allow them to adhere to the flask. Recombinant IFN- was then added to each flask at a concentration of 1000 IU/mL. Medium was changed daily, and fresh IFN-
was added daily for 3 days. Cells were then harvested by trypsinization, counted, and stained with propidium iodide (PI). Dead cells stained PI-positive because of loss of cell membrane integrity. Percentages of dead and living cells were analyzed on a FACScan flow cytometer.
Mouse Xenograft Model
Female C.B-17 SCID mice (6 weeks old) were obtained from Harlan (Indianapolis, IN). Mice were used according to approved institutional protocols. Mice were injected intravenously in the lateral tail vein with 2 x 106 MDA 231 or A375SM tumor cells suspended in 200 µL of PBS. In preliminary experiments, we determined that all mice injected with 2 x 106 MDA 231 or 2 x 106 A375SM cells developed macroscopic tumor nodules in their lungs at 8 days after tumor cell injection (data not shown).
Determination of Effect of MSC-IFN-, MSC-Gal, and Recombinant IFN-
on MDA 231 Tumor Weight in Mouse Lung
Eight days after MDA 231 tumor cell injection (as above), the mice started treatment with recombinant IFN- (100 000 IU injected every other day by subcutaneous injections for the whole duration of the experiment [n = 4]), MSC-IFN-
(three doses of 106 cells given weekly by intravenous injections [n = 4]), or MSC-Gal (three doses of 106 cells given weekly by intravenous injection [n = 4]). Mice injected with MDA 231 tumor cells alone (n = 4) and healthy mice with no tumor cell injection (n = 4) served as controls. Mice were killed by asphyxiation with CO2 30 days after tumor cell injection. We measured the weight of whole lungs in all groups of mice and used whole lung weight as a surrogate endpoint of MDA 231 tumor burden in the lung and to assess the effect of MSC-IFN-
, MSC-Gal, and recombinant IFN-
on tumor growth. Lungs and other organs from mice injected with MSC-Gal were also used for histochemistry studies described below.
Survival Analysis
Eight days after MDA 231 cell injection (see above), the mice started treatment with recombinant IFN- (100 000 IU injected subcutaneously every other for the duration of the experiment [n = 10]), MSC-IFN-
(three doses of 106 cells given weekly by intravenous injections [n = 8]), or MSC-Gal (three doses of 106 cells given weekly by intravenous injections [n = 5]). Mice injected with MDA 231 tumor cells only, with no further treatment, served as controls (n = 10).
Eight days after A375SM tumor cell injection (as above), the mice started treatment with recombinant IFN- (40 000 IU injected subcutaneously daily for the duration of the experiment [n = 10]), MSC-IFN-
IV (three doses of 106 MSC-IFN-
cells given weekly by intravenous injections [n = 8]), or MSC-IFN-
SC (three doses of 106 cells given weekly by subcutaneous injections [n = 5]). Mice injected with A575SM tumor cells only with no further treatment served as controls (n = 10). All mice were followed daily until death. None of the mice had to be sacrificed because of excessive bleeding, open wound infection, moribund status, or prostration with weight loss of more than 25% of initial body weight, as specified in our protocols.
Tissue Processing and Imaging Studies
Eight days after MDA 231 tumor cell injection, the mice (n = 5) received MSC-Gal (three doses of 106 cells given weekly by intravenous injection). The mice were killed by asphyxiation with CO2 at 30 days after tumor cell injection. Healthy mice (n = 5) with no tumors received MSC-Gal (three doses of 106 cells given weekly by intravenous injection) and were used as controls. Lungs and other organs from both groups of mice were embedded in Tissue TEK OTC compound (Miles, Elkhart, IN), snap-frozen in liquid nitrogen, and stored at 80 °C. Frozen tissue was sectioned (6- to 8-µm-thick sections), mounted onto slides, and stained with hematoxylineosin or for -gal as described below. Images were captured with the use of an Axioplan2 microscope (Carl Zeiss, Thornwood, NY) equipped with a charge-coupled device camera (Hamamatsu, Bridgewater, NJ) and processed using Adobe Photoshop software, version 5.0 (Adobe Systems, San Jose, CA).
Histochemistry
Whole lungs were incubated with 2% X-gal (Sigma) in 1 M MgCl2, 30 mM potassium ferricyanide, and 30 mM potassium ferrocyanide overnight, and refixed in 10% neutral-buffered formalin. The tissues were then dehydrated with ethanol and, after minimal exposure to xylene, embedded in paraffin, sectioned (5 µm thick), and mounted on slides. The sections were de-paraffinized through minimal exposure to xylene and decreasing concentrations of ethanol and counterstained with eosin or Nuclear Fast red (Arcturus, Mountain View, CA), according to the manufacturer's instructions. Alternatively, slides with sections of frozen tissues were fixed with cold acetone : ethanol (1 : 1 vol/vol) for 20 minutes and stained with X-gal.
Measurement of IFN- Concentration in Mouse Plasma
Mice with established MDA 231 lung metastases were injected intravenously (n = 5) or subcutaneously (n = 5) with 106 MSC-IFN- cells or subcutaneously with 40 000 IU (n = 5) or 100 000 IU (n = 5) of recombinant IFN-
. Beginning 1 day after MSC-IFN-
injection or 1 hour after recombinant IFN-
injection and at appropriate intervals thereafter, we collected 200 µL of blood from each mouse into heparinized capillary tubes through tail vein incisions. Blood was immediately centrifuged (700g for 5 minutes at 4 °C) to remove cells, and plasma was collected and stored at 80 °C. We used an enzyme-linked immunosorbent assay (Fujirebio, Tokyo, Japan) and a National Institutes of Health (NIH) standard for IFN-
1a provided by the manufacturer to determined plasma concentrations of IFN-
. The NIH standard of IFN-
1a allowed us to compare serum levels (in IU/mL) from our study with the data published in the literature.
Statistical Methods
We initially used the KruskalWallis test to assess the statistical significance of overall differences in lung weights between all treatment groups at day 30. Because the results of the KruskalWallis test showed that the difference between MSC-IFN-treated and control mice was statistically significant (P = .004), we used the Wilcoxon rank sum test to perform pairwise comparisons of treatment effect on lung weight between all groups. Survival was measured from the day of MDA 231 cell or A375SM cell injection until the day of death. For the survival data, the log-rank test was used to assess differences in survival among the four treatment groups. Because this overall test showed that the difference between MSC-IFN-
treated and control mice was statistically significant for both tumor models (P<.001), pairwise log-rank tests were performed. All statistical tests were two-sided; a P value of less than .05 was considered statistically significant. Statistical analyses were performed by using either SAS, version 8.2 (SAS, Cary, NC) or Statistica, version 7.0, software (StatSoft, Tulsa, OK).
![]() |
RESULTS |
---|
![]() ![]() ![]() ![]() ![]() ![]() ![]() ![]() |
---|
We examined the effect of recombinant IFN- on the growth of MDA 231 breast carcinoma cells and A375SM melanoma cells by using the MTS assay, which measures cell viability as a percentage of the growth of control cells. Recombinant IFN-
inhibited the growth of both cell lines in a concentration-dependent fashion (Fig. 1, A and B). High concentrations of recombinant IFN-
(i.e.,
200 IU/mL) led to negative values for A375SM cells and therefore suggested that higher concentrations of IFN-
induced death in these tumor cells. We tested whether this was the case and found that A375SM cells incubated with 1000 IU/mL of IFN-
had more propidium iodidepositive (i.e., apoptotic) cells than A375SM cells incubated in the absence of IFN-
(Fig. 1, C). Next, we investigated whether co-culturing the tumor cells with MSC that were transduced with adenovirus expressing IFN-
(i.e., MSC-IFN-
cells) would inhibit tumor cell growth. MDA 231 and A375SM cells were each co-cultured with uninfected MSC or MSC-IFN-
cells at a 10 : 1 ratio. Compared with MDA 231 cells cultured alone, MDA 231 cells co-cultured with MSC-IFN-
cells displayed statistically significantly reduced growth (difference in mean percentage of control cell growth = 94%, 95% CI = 81.2% to 106.8%; P<.001), whereas MDA 231 cells co-cultured with uninfected MSC were not growth inhibited (difference in mean percentage of control cell growth = 35.7%, 95% CI = 23.7% to 47.7%;P = .063). Growth of A375SM cells co-cultured with MSC-IFN-
cells was also statistically significantly inhibited compared with the growth of A375SM cells alone (difference in mean percentage of control cell growth = 104.8%, 95% CI = 82.1% to 127.5%; P<.001). By contrast, the growth of A375SM cells co-cultured with uninfected MSC was enhanced compared with the growth of A375SM cells alone (difference in mean percentage of control cell growth = 60.1%, 95% CI = 28.6% to 91.6%; P = .031) (Fig. 1, D and E). These data suggest that MSC-IFN-
cells inhibited the proliferation of these tumor cells and, because these effects were observed in vitro, this inhibition did not require additional components of the host immune system.
|
We next examined the in vivo antitumor activity of MSC-IFN- cells by using an SCID mouse xenograft model. We injected MDA 231 cells intravenously into the tail veins of the mice to establish pulmonary metastases. Eight days later, we began treating the mice with intravenous injections of 106 MSC-IFN-
cells at weekly intervals for 3 weeks (n = 4). Control mice received either no treatment (n = 4) or intravenous injections of 106 MSC-Gal cells at weekly intervals for 3 weeks (n = 4). An additional group of mice (n = 4) was treated with subcutaneous injections of 100 000 IU of recombinant IFN-
every other day from day 8 after tumor cell injection until the end of the experiment. A group of healthy mice (n = 4) that received no cell injections served as the referent for measurement of normal lung weight. Thirty days after tumor cell injection, we measured the weight of whole lungs.
The mean lung weight of mice injected with MDA 231 tumor cells was statistically significantly greater than the mean lung weight of healthy mice (mean lung weights: 0.977 g versus 0.413 g; difference = 0.564 g, 95% CI = 0.426 g to 0.702 g, P = .021) (Fig. 2, B; Table 1). Much of this weight difference was due to the tumor tissue occupying substantial portions of the lungs of the mice injected with the tumor cells (Fig. 2, A). Therefore, we used whole lung weight as a surrogate endpoint of tumor burden in lungs and to assess efficacy of treatment (Fig. 2, B; Table 1). Mice injected with tumor cells and treated intravenously with MSC-IFN- cells had statistically significantly smaller lungs than control untreated mice injected with tumor cells only (mean lung weights: 0.408 g versus 0.977 g; difference: 0.569 g, 95% CI= 0.446 g to 0.692 g; P = .021). By contrast, the mean weight of lungs from mice treated with recombinant IFN-
was not statistically significantly different from that of untreated control mice injected with tumor cells only (1.09 g versus 0.977 g;difference = 0.113 g, 95% CI = 0.014 to 0.246 g; P = .083). The mean lung weight for mice treated with MSC-Gal cells was also not statistically significantly different from that of untreated mice with tumors (1.125 g versus 0.977 g; difference = 0.148 g, 95% CI = 0.019 to 0.315 g; P = .081). These findings suggest that intravenous administration of MSC-IFN-
cells inhibits the growth of tumors in lungs, whereas systemically administered recombinant IFN-
(at the dose used) or intravenously injected MSC-Gal cells does not (Table 1).
|
|
We examined whether the various treatments improved the survival of mice with preestablished pulmonary metastases derived from MDA 231 cells or A375SM cells (Fig. 3, A; Table 2). Mice were treated with recombinant IFN-, MSC-IFN-
cells, or MSC-Gal cells, as described above and followed until death due to lung tumors. Among mice bearing MDA 231 cellderived pulmonary metastases, those treated with MSC-IFN-
cells lived statistically significantly longer than untreated mice (median survival: 60 days versus 37 days, difference = 23 days, 95% CI = 14.5 to 34 days; P<.001), whereas mice treated with MSC-Gal cells (median survival: 36 days) or with systemically administered IFN-
(median survival: 41 days) did not (difference in median survival: 1 day; P = .509 and 4 days; P = .308, respectively) (Fig. 3, A; Table 2).
|
|
|
MSC Engraftment In Vivo
To confirm that injected MSC engrafted in tumors, we intravenously injected mice that did or did not carry MDA 231 xenograft tumors with three weekly doses of 106 MSC-Gal cells and traced the progeny of those cells by histochemical staining of mouse lungs and other organs for -gal. Histochemical staining was performed 14 days after the last doses of MSC-Gal cells were administered; representative images are shown in Fig. 4. We observed numerous X-galpositive cells in MDA 231 pulmonary tumors (Fig. 4, B). These cells had formed colonies (average of 4 colonies per section, 95% CI = 2 to 6 colonies) and were incorporated into the tumor architecture (Fig. 4, B), suggesting that MSC can reach the extravascular space and contribute to the development of tumor connective stroma. We suspect that each X-galpositive colony originated from a single (or very few) MSC that proliferated in situ, presumably under the influence of signals from the surrounding microenvironment (16).
|
Plasma Levels of IFN- After Administration of Recombinant IFN-
or MSC-IFN-
Cells to Mice
The activity of human IFN- is species-specific; thus, human IFN-
does not substantially affect murine cells (28). Accordingly, the clinically relevant dose of human IFN-
in mouse xenograft models cannot be determined on the basis of drug tolerance in mice. However, because IFN-
toxicity in patients is associated with high IFN-
levels in serum (11,12), it is possible that serum concentrations of IFN-
in mice could serve as a surrogate marker of the toxicity that might be expected in humans. We therefore examined plasma levels of IFN-
in mice with MDA 231 cellderived pulmonary metastases at various times after the mice had received subcutaneous injections of recombinant human IFN-
. We found that mice injected with 40 000 IU of recombinant human IFN-
had mean serum IFN-
levels of 156 IU/mL (95% CI = 128 to184 IU/mL) after 2 hours, 29 IU/mL (95% CI = 11.5 to 46.5 IU/mL) after 6 hours, and 0 IU/mL (95% CI = 0 to 0 IU/mL) after 24 hours. Mice injected with 100 000 IU of recombinant human IFN-
had mean serum IFN-
levels of 192 IU/mL (95% CI = 152 to 232 IU/mL) after 2 hours, 61 IU/mL (95% CI = 0 to128 IU/mL) after 6 hours, and 16 IU/mL (95% CI = 0 to 40 IU/mL) after 24 hours (Table 4). We also examined serum levels of human IFN-
in mice after a single subcutaneous or intravenous injection of 106 MSC-IFN-
cells. Subcutaneously injected MSC-IFN-
resulted in mean serum IFN-
levels of 47.2 IU/mL (95% CI = 21.7 to 72.7 IU/mL) after 1 day, 7.7 IU/mL (95% CI = 1.0 to 14.3 IU/mL) after 3 days, and 1.4 IU/mL (95% CI = 0.6 to 2.1 IU/mL) after 6 days. Intravenous injection of MSC-IFN-
resulted in mean serum IFN-
levels of 14 IU/mL (95% CI = 4.8 to 24.3 IU/mL) after 1 day, 0.48 IU/mL (95% CI = 0.5 to 1.4 IU/mL) after 3 days, and 1.0 IU/mL (95% CI = 0.1 to 2.0 IU/mL) after 6 days (Table 4). Mean serum IFN-
levels in mice injected subcutaneously with MSC-IFN-
were not statistically significantly different from those in mice injected intravenously with MSC-IFN-
at 1 day (difference = 32.6 IU/mL, 95% CI = 5.32 to 59.9 IU/mL; P = .07), at 3 days (difference = 7.2 IU/mL, 95% CI = 0.5 to 13.9 IU/mL; P = .1), or at 6 days (difference = 0.33 IU/mL, 95% CI = 0.84 to 1.5 IU/mL; P = .81) after cell administration. However, given our finding that intravenously injected but not subcutaneously injected MSC-IFN-
inhibited A375SM tumor growth in lung (Fig. 3, B; Table 3), we conclude that the observed antitumor effect of intravenously injected MSC-IFN-
is not associated with an increase of IFN-
levels in serum. Overall, these results provide further evidence that intravenously injected MSC-IFN-
cells inhibit the growth of malignant cells by local production of IFN-
in the tumor microenvironment and that this effect occurs even though systemic plasma concentrations of IFN-
are low.
|
![]() |
DISCUSSION |
---|
![]() ![]() ![]() ![]() ![]() ![]() ![]() ![]() |
---|
Our results suggest that the antitumor activity of MSC-IFN- cells is associated not with the serum concentration of IFN-
but rather with the engraftment of MSC in tumors and the local effects of the IFN-
they produce on malignant cells. Therefore, selective engraftment of MSC in tumors is an important issue for further clinical development of this method. We did not observe engraftment of intravenously administered MSC in any of the healthy organs we examined (i.e., lung, liver, spleen, kidney, and muscle). These findings are consistent with earlier reports that systemically administered MSC poorly engraft in healthy tissues (32). Jiang et al. (33) have reported extensive engraftment of multipotent adult progenitor cells (MAPC), which are similar to and can co-purify with MSC. However, MAPC differ from the MSC used in our study. MAPC can differentiate into a wide range of epithelial cell types and can engraft in lung, skin, blood, gut, and liver (33). By contrast, the differentiation potential of MSC is restricted to cells with mesodermal or connective tissue phenotypes: osteoblasts, chondroblasts, adipocytes, myocytes, and fibroblasts (17,18). This limited differentiation potential of MSC likely prevents them from engrafting in epithelial tissues, which have high spontaneous cell turnover. Future clinical trials of this method should consider the possibility that MSC may engraft at sites of tissue injury and thus exclude patients who have undergone surgery or invasive procedures or have had infections (23). In addition, the selectivity of MSC for other cancer metastases will require careful investigation.
Even if MSC-IFN- cells engraft selectively in tumors, the IFN-
they produce could still be released into the circulation and contribute to toxicity by affecting other organs. In this regard, we demonstrated that intravenous administration of MSC-IFN-
cells results in a peak mean IFN-
serum level of 14.0 IU/mL. This value is similar to the mean (± standard deviation) maximal IFN-
serum concentration of 12.3 ± 3.9 IU/mL reported in humans after subcutaneous administration of IFN-
at the maximally tolerated dose of 18 x 106 IU (11,12). Therefore, intravenous administration of MSC-IFN-
at a dose that inhibits malignant cell growth may also be tolerated in a clinical setting. These findings are again consistent with the known rapid metabolism and short half-life of IFN-
in circulation and support its physiologic role as a paracrine regulator of cell fate that acts locally in tissues.
It has been postulated that IFN- is an important inhibitor of cell growth during development (31,34) and oncogenesis (35). IFN-
can reverse the oncogenic potential of transformed tumorigenic cells in experimental systems (36,37), and recent data have also suggested that IFN-
has a role in the regulation of apoptosis in malignant cells (35). Although the molecular mechanisms that are responsible for growth inhibition and elimination of tumor cells by IFN-
await clarification, results of numerous studies have implicated a variety of IFN-regulated genes, such as members of the interferon regulatory factory gene family, PKR, and tumor necrosis factor-related apoptosis-inducing ligand (TRAIL)/Apo2L, in these processes (35,38). Several reports have suggested that abnormally low expression of IFN-
in the tissues surrounding certain tumors contributes to unregulated cell growth and malignant potential in vivo (39,40). Therefore, therapy with MSC-IFN-
cells could be used to increase IFN-
expression in tumors and surrounding tissues and to control the growth of malignant cells. In fact, the potential clinical application of MSC-IFN-
cells could be broad, because many types of malignancies are sensitive to the antiproliferative or proapoptotic effects of IFN-
in vitro (13,5). Moreover, MSC may be effective carriers for other biologic molecules, such as the tumor necrosis factor family of proteins (41) and anti-angiogenic molecules (42). These agents have shown potential promise as cancer therapies in many experimental systems, but their clinical application has suffered from pharmacologic limitations similar to those seen for IFN (41,42).
Overall, we have demonstrated that MSC engrafted in tumors may act as precursors for stromal cells and can serve as cellular vehicles for the delivery and local production of biologic agents. This approach may overcome the extensive metabolism and toxicity associated with some biologic agents and could serve as a versatile tool for manipulating the extracellular milieu of malignant cells.
![]() |
NOTES |
---|
![]() ![]() ![]() ![]() ![]() ![]() ![]() ![]() |
---|
![]() |
REFERENCES |
---|
![]() ![]() ![]() ![]() ![]() ![]() ![]() ![]() |
---|
1 Wong VL, Rieman DJ, Aronson L, Dalton BJ, Greig R, Anzano MA. Growth-inhibitory activity of interferon-beta against human colorectal carcinoma cell lines. Int J Cancer 1989;43:52630.[ISI][Medline]
2 Johns TG, Mackay IR, Callister KA, Hertzog PJ, Devenish RJ, Linnane AW. Antiproliferative potencies of interferons on melanoma cell lines and xenografts: higher efficacy of interferon beta. J Natl Cancer Inst 1992;84:118590.[Abstract]
3 Chawla-Sarkar M, Leaman DW, Borden EC. Preferential induction of apoptosis by interferon (IFN)-beta compared with IFN-alpha2: correlation with TRAIL/Apo2L induction in melanoma cell lines. Clin Cancer Res 2001;7:182131.
4 Lokshin A, Mayotte JE, Levitt ML. Mechanism of interferon beta-induced squamous differentiation and programmed cell death in human non-small-cell lung cancer cell lines. J Natl Cancer Inst 1995;87:20612.[Abstract]
5 Zhang H, Koty PP, Mayotte J, Levitt ML. Induction of multiple programmed cell death pathways by IFN-beta in human non-small-cell lung cancer cell lines. Exp Cell Res 1999;247:13341.[CrossRef][ISI][Medline]
6 Bradley JD, Scott CB, Paris KJ Demas WF, Machtay M, Komaki R, et al. A phase III comparison of radiation therapy with or without recombinant beta-interferon for poor-risk patients with locally advanced non-small-cell lung cancer (RTOG 93-04). Int J Radiat Oncol Biol Phys 2002;52:11739.[CrossRef][ISI][Medline]
7 Kowalzick L, Weyer U, Lange P, Breitbart EW. Systemic therapy of advanced metastatic malignant melanoma with a combination of fibroblast interferon-beta and recombinant interferon-gamma. Dermatologica 1990;181:298303.[ISI][Medline]
8 Schiller JH, Storer B, Bittner G, Willson JK, Borden EC. Phase II trial of a combination of interferon-beta ser and interferon-gamma in patients with advanced malignant melanoma. J Interferon Res 1988;8:5819.[ISI][Medline]
9 Tester WJ, Kim KM, Krigel RL, Bonomi PD, Glick JH, Asbury RF, et al. A randomized phase II study of interleukin-2 with and without beta-interferon for patients with advanced non-small cell lung cancer: an Eastern Cooperative Oncology Group study (PZ586). Lung Cancer 1999;5:199206.
10 Von Hoff DD, Metch B, Lucas JG, Balcerzak SP, Grunberg SM, Rivkin SE. Phase II evaluation of recombinant interferon-beta (IFN-beta ser) in patients with diffuse mesothelioma: a Southwest Oncology Group study. J Interferon Res 1990;10:5314.[ISI][Medline]
11 Buchwalder PA, Buclin T, Trinchard I, Munafo A, Biollaz J. Pharmacokinetics and pharmacodynamics of IFN-beta 1a in healthy volunteers. J Interferon Cytokine Res 2000;20:85766.[CrossRef][ISI][Medline]
12 Salmon P, Le Cotonnec JY, Galazka A, Abdul-Ahad A, Darragh A. Pharmacokinetics and pharmacodynamics of recombinant human interferon-beta in healthy male volunteers. J Interferon Cytokine Res 1996;16:75964.[ISI][Medline]
13 Einhorn S, Grander D. Why do so many cancer patients fail to respond to interferon therapy? J Interferon Cytokine Res 1996;16:27581.[ISI][Medline]
14 Bocci V, Carraro F, Naldini A, Borrelli E, Biagi G, Gotti G, et al. Interferon levels in human pulmonary tumors are lower than plasma levels. J Biol Regul Homeost Agents 1990;4:1536.[ISI][Medline]
15 Johns TG, Kerry JA, Veitch BA, Mackay IR, Tutton PJ, Tymms M, et al. Pharmacokinetics, tissue distribution, and cell localization of [35S]methionine-labeled recombinant human and murine alpha interferons in mice. Cancer Res 1990;50:471823.[Abstract]
16 Studeny M, Marini FC, Champlin RE, Zompetta C, Fidler IJ, Andreeff M. Bone marrow-derived mesenchymal stem cells as vehicles for interferon-beta delivery into tumors. Cancer Res 2002;62:36038.
17 Prockop DJ. Marrow stromal cells as stem cells for nonhematopoietic tissues. Science 1997;276:714.
18 Pittenger MF, Mackay AM, Beck SC Jaiswal RK, Douglas R, Mosca JD, et al. Multilineage potential of adult human mesenchymal stem cells. Science 1999:284:1437.
19 Horwitz EM, Prockop DJ, Fitzpatrick LA, Koo WW, Gordon PL, Neel M, et al. Transplantability and therapeutic effects of bone marrow-derived mesenchymal cells in children with osteogenesis imperfecta. Nat Med 1999;5:30913.[CrossRef][ISI][Medline]
20 Liechty KW, Mackenzie TC, Shaaban AF, Radu A, Moseley AM, Deans R, et al. Human mesenchymal stem cells engraft and demonstrate site-specific differentiation after in utero transplantation in sheep. Nat Med 2000;6:12826.[CrossRef][ISI][Medline]
21 Weissman IL. Stem cells: units of development, units of regeneration, and units in evolution. Cell 2000:100:15768.[ISI][Medline]
22 Reyes M, Dudek A, Jahagirdar B, Koodie L, Marker PH, Verfaillie CM. Origin of endothelial progenitors in human postnatal bone marrow. J Clin Invest 2002:109:33746.
23 Wu GD, Nolta JA, Jin YS, Baar ML, Yu H, Starnes VA, et al. Migration of mesenchymal stem cells to heart allografts during chronic rejection. Transplantation 2003;75:67985.[ISI][Medline]
24 Hanahan D, Weinberg RA. The hallmarks of cancer. Cell 2000;100:5770.[ISI][Medline]
25 Dvorak HF. Tumors: wounds that do not heal. Similarities between tumor stroma generation and wound healing. N Engl J Med 1986;315:16509.[ISI][Medline]
26 Hasebe T, Sasaki S, Sugitoh M, Ono M, Saitoh N, Ochiai A. Highly proliferative intratumoral fibroblasts and a high proliferative microvessel index are significant predictors of tumor metastasis in T3 ulcerative-type colorectal cancer. Hum Pathol 2001;32:4019.[CrossRef][ISI][Medline]
27 Kuniyasu H, Abbruzzese JL, Cleary KR, Fidler IJ. Induction of ductal and stromal hyperplasia by basic fibroblast growth factor produced by human pancreatic carcinoma. Int J Oncol 2001;19:6815.[ISI][Medline]
28 Qin XQ, Beckham C, Brown JL, Lukashev M, Barsoum J. Human and mouse IFN-beta gene therapy exhibits different anti-tumor mechanisms in mouse models. Mol Ther 2001;4:35664.[CrossRef][ISI][Medline]
29 Lee K, Majumdar MK, Buyaner D, Hendricks JK, Pittenger MF, Mosca JD. Human mesenchymal stem cells maintain transgene expression during expansion and differentiation. Mol Ther 2001;3:85766.[CrossRef][ISI][Medline]
30 Bielenberg DR, McCarty MF, Bucana CD, Yuspa SH, Morgan D, Arbeit JM, et al. Expression of interferon-beta is associated with growth arrest of murine and human epidermal cells. J Invest Dermatol 1999;112:8029.
31 Hertzog PJ, Hwang SY, Kola I. Role of interferons in the regulation of cell proliferation, differentiation, and development. Mol Reprod Dev 1994;39:22632.[ISI][Medline]
32 Koc ON, Peters C, Aubourg P, Raghavan S, Dyhouse S, DeGasperi R, et al. Bone marrow-derived mesenchymal stem cells remain host-derived despite successful hematopoietic engraftment after allogeneic transplantation in patients with lysosomal and peroxisomal storage diseases. Exp Hematol 1999;27:167581.[CrossRef][ISI][Medline]
33 Jiang Y, Jahagirdar BN, Reinhardt RL, Schwartz RE, Keene CD, Ortiz-Gonzales XR, et al. Pluripotency of mesenchymal stem cells derived from adult marrow. Nature 2002;418:419.[CrossRef][ISI][Medline]
34 Clemens MJ, McNurlan MA. Regulation of cell proliferation and differentiation by interferons. Biochem J 1985:226:34560.[ISI][Medline]
35 Clifford JL, Walch E, Yang X, Xu X, Alberts DS, Clayman GL, et al. Suppression of type I interferon signaling proteins is an early event in squamous skin carcinogenesis. Clin Cancer Res 2002;8:206772.
36 Gerfaux J, Sergiescu D, Fournier JG, Pochart F, Joret AM, Chany C. Presence of a constitutive paracrine beta-interferon in v-mos-bearing nonmalignant reverted cells. Cancer Res 1989;49:12416.[Abstract]
37 Sergiescu D, Gerfaux J, Joret AM, Chany C. Persistent expression of v-mos oncogene in transformed cells that revert to nonmalignancy after prolonged treatment with interferon. Proc Natl Acad Sci U S A 1986;83:57648.[Abstract]
38 Barber GN. The interferons and cell death: guardians of the cell or accomplices of apoptosis? Semin Cancer Biol 2000;10:10311.[CrossRef][ISI][Medline]
39 Bielenberg DR, Bucana CD, Sanchez R, Mulliken JB, Folkman J, Fidler IJ. Progressive growth of infantile cutaneous hemangiomas is directly correlated with hyperplasia and angiogenesis of adjacent epidermis and inversely correlated with expression of the endogenous angiogenesis inhibitor, IFN-beta. Int J Oncol 1999;14:4018.[ISI][Medline]
40 Kuniyasu H, Yasui W, Shinohara H, Yano S, Ellis LM, Wilson MR, et al. Induction of angiogenesis by hyperplastic colonic mucosa adjacent to colon cancer. Am J Pathol 2000;157:152335.
41 Hohlbaum AM, Moe S, Marshak-Rothstein A. Opposing effects of transmembrane and soluble Fas ligand expression on inflammation and tumor cell survival. J Exp Med 2000:191:120920.
42 Eder JP Jr, Supko JG, Clark JW, Puchalski TA, Garcia-Carbonero R, Ryan DP, et al. Phase I clinical trial of recombinant human endostatin administered as a short intravenous infusion repeated daily. J Clin Oncol 2002:20:377284.
Manuscript received January 23, 2004; revised August 25, 2004; accepted September 13, 2004.
This article has been cited by other articles in HighWire Press-hosted journals:
Correspondence about this Article
![]() |
||||
|
Oxford University Press Privacy Policy and Legal Statement |