The present invention relates to a method for the treatment and mitigation of the progression of cancer, more particularly to a method for using Interleukin-1 to limit the response of mesenchymal stem cells to breast cancer cells.
Breast cancer survivors can develop metastasis after more than ten years of remission. The bone marrow is an organ for which breast cancer shows preference. Interestingly, in many cases, the bone marrow has been attributed as the source of breast cancer cells during breast cancer resurgence. It has been unclear if a particular subset of breast cancer cells survives as dormant cells in the bone marrow, and if so, how they can be targeted. Metastasis of breast cancer can occur without history of a primary tumor. Avoiding tertiary metastasis requires a thorough understanding of how breast cancer cells are protected towards dormancy, such as protection by cells in bone marrow.
Breast cancer cells can adopt quiescence at regions close to the endosteum. Changes in cytokine production and gap junctional intercellular communications (GJIC) have been implicated in breast cancer cell quiescence. Mesenchymal stem cells (MSCs) can contribute to breast cancer dormancy by virtue of its ability to interact with breast cancer cells through CXCR4 and the ligand, stromal cell-derived factor 1α (CXCL12). In addition, MSCs can protect breast cancer cells from immune response, thereby providing the breast cancer cells with an advantage to attain dormancy. CXCR4 is a seven-transmembrane, G-protein coupled receptor that facilitates chemoattaction of breast cancer cells to organs with high CXCL12 levels.
Upon entering bone marrow, MSCs can be among the first cells to interact with breast cancer cells since they are at the interface, around the main blood vessel. Here the immune suppressive effects of MSCs can provide an immediate advantage by protecting the breast cancer cells from immune clearance. This will give the breast cancer cells an advantage to survive in bone marrow and integrate in regions close to the endosteum as dormant cells.
MSCs are important for blood vessel integrity, thereby making them unlikely drug discovery targets. Similarly, direct targeting of GJIC between breast cancer cells and stroma could be toxic since gap junctions between stromal cells are important for hematopoietic support. Thus, there is a current need to identify and understand the role of MSCs in the behavior of breast cancer cells at metastatic sites and also at the primary region in order to provide novel methods of therapeutic intervention.
The present invention relates to a method of treating breast cancer which comprises dissociating quiescent breast cancer cells from their environment in the bone marrow and treating the disassociated breast cancer cells with standard chemotherapeutic agents.
More particularly, the invention relates to a method which comprises using a CXCR4 antagonist, such as, for example AMD3100 to break a BCC-MSC association, thus dissociating the quiescent breast cancer cells and inducing breast cancer cell proliferation and susceptibility to chemotherapy.
Thus the present invention relates to a novel immune therapy for cancer using Interleukin-1 to limit the response of mesenchymal stem cells to breast cancer cells. It has now been shown that MSCs support the growth of breast cancer cells via the production of IL-1 and make the breast cancer cells susceptible to carboplatin treatment. Thus the present invention provides a novel treatment which features immune therapy in the context of the functions of MSCs as a novel approach to breast cancer treatment. By identifying and elucidating the role of MSCs in the behavior of breast cancer cells at metastatic sites and also at the primary region, this invention provides novel approaches for therapeutic intervention. More particularly, in the presence of MSCs a CXCR4 antagonist transitioned BCCs into cycling cells and conferred susceptibility to a chemotherapeutic agent. The proliferation of BCCs depended on the release of IL-1α and IL-1β from MSCs, but only if the CXCR4 antagonist uncoupled BCCs from MSCs.
MSCs can pose a significant clinical dilemma for breast cancer treatment, due to MSCs acting as suppressor of breast cancer cells proliferation. In the absence of proliferation, the breast cancer cells will show a seemingly dormant phenotype and could be refractory to most anti-cancer agents. New therapies are required to reverse the dormant breast cancer cells into proliferation cells for targeting. In the studies which yielded the present invention, we showed CXCL12 and its receptor, CXCR4, as mediators in the interactions between MSCs and breast cancer cells, leading to growth arrest (
MDA-MB-231 seems to be more responsive to AMD3100 than T47D (
Interestingly, AMD3100 alone reduced the growth of the breast cancer cells cultured alone (
The studies are significant to breast cancer dormancy in any organ, but specifically for bone marrow. In addition to acting as an antagonist to CXCR4, AMD3100, also known as Plerixafor, is a macrocyclic compound which is an allosteric agonist of CXCR7. Our studies do not indicate that AMD3100 activates CXCR7 for the proliferation of breast cancer cells since alone, breast cancer cells show suppressed growth with AMD3100 (
Indeed, an interesting and unexpected observation is the need for continued presence of AMD3100 during treatment with carboplatin. This suggests that the interaction between breast cancer cells and MSCs is rapid. Pre-treatment with the antagonist for 48 hours prior to chemotherapy did not induce significant cell death (
We determined that for the highly aggressive MDA-MD-231 breast cancer cells, treatment of co-cultures with AMD3100 promoted the subsequent release of IL-1α and IL-1β from the MSCs (
AMD3100 can mobilize hematopoietic stem cells (HSCs). This poses some concerns if this agent is used in patients to target bone marrow-resident breast cancer cells. If AMD3100 were to be used in combination with chemotherapy, then one potential side effect would be mobilization of HSCs. Additional antagonists for CXCR4 should be studied for those that can facilitate targeting of breast cancer cells but minimize toxicity to resident HSCs.
The findings on CXCL12/CXCR4 axis suggest that this is a druggable target to combat breast cancer metastasis to bone marrow, and other organs. It is possible that CXCR4 antagonists can induce re-cycling of breast cancer cells that interact with MSCs within the bone marrow, as well as within the tumor (
Clinical evidence indicates that breast cancer cells can remain quiescent without clinical evidence, and in bone marrow, without affecting hematopoiesis. The dormant breast cancer cells could resurge more than ten years later, with the re-animated breast cancer cells metastasizing to tertiary sites. Breast cancer cells can interact with (MSCs) through membrane-bound CXCL12 (SDF-1α) and its receptor, CXCR4. MSCs can also protect breast cancer cells from immune clearance through suppression of NK and CTL activity, as well as expansion of T-regulator cells (Tregs). However, since MSCs can also support the growth of breast cancer cells, the present invention is based upon the novel hypothesis that MSC-derived cytokine production could explain these differences. In the presence of MSCs, the CXCR4 antagonist, AMD3100, transitioned breast cancer cells into cycling cells and conferred susceptibility to carboplatin. The proliferation of breast cancer cells depended on the release of IL-1α and IL-1β from MSCs, but only if AMD3100 uncoupled breast cancer cells from MSCs. The findings which led to the present invention were validated in nude BALB/c where AMD3100 and carboplatin showed significant reduction in tumor volume, despite the presence of MSCs. However, when the activity of IL-1 was blocked with a receptor antagonist, the cancer cells resisted carboplatin. This verified a critical role for IL-1 for MSCs to transition from protective to supporting the growth of breast cancer cells. These findings are essential to the present invention, a novel, efficient immune therapy, in light of MSCs having a central role not only in breast cancer, but also in other solid tumors.
As part of the present invention, the CXCL12/CXCR4 interaction in breast cancer cell quiescence was elucidated. Breast cancer cells and MSCs interact through membrane-bound CXCL12 and CXCR4. The proliferation of breast cancer cells, in the presence or absence of MSCs, was compared and then the extent of involvement by the CXCL12/CXCR4 interaction was determined. The breast cancer cells and MSCs were studied as untransfected and CXCL12 or CXCR4 knockout (
Similar studies were also conducted with MSCs and breast cancer cells, stably knockdown for CXCL12 and CXCR4. The knockdown of CXCL12 and/or CXCR4 in both breast cancer cells and MSCs (
Pharmacological disruption of CXCL12/CXCR4 in breast cancer cell proliferation was necessarily involved with the conception of the present invention. The study of the molecular disruption of CXCL12-CXCR4 with a CXCR4 antagonist, AMD3100 was expanded. We first examined the effects of AMD3100 on the proliferation of MDA-MB-231 and T47D, in co-culture with MSCs (
Breast cancer cells alone showed a decrease in cell proliferation with the antagonist (open circles). In contrast to MDA-MD-231, T47D proliferation was inversely related to AMD3100 concentration (
Some studies also looked at AMD3100 in long-term breast cancer cell/MSC co-cultures. In this set of studies we determined the effect of AMD3100 when breast cancer cells were cultured with MSCs for over 48 hours since this will recapitulate in situ when breast cancer cells are expected to contact MSCs for long periods. Breast cancer cells were co-cultured with MSCs for 12 days (D12), with optimal AMD3100 (
It was also necessary to understand the role of AMD3100 in breast cancer cell cell cycle re-entry. We studied cell cycle phase of 12-day co-cultured breast cancer cells, with or without AMD3100, as described for
Soluble factors in re-entry of breast cancer cell into cycling were also examined. AMD3100 promotes proliferation (
IL-1α in AMD3100-mediated breast cancer cell cycling is very important to the present invention. To assess the factor(s) responsible for breast cancer cell proliferation (
In pursuit of the present invention, we next identified whether MSCs and/or breast cancer cells are the sources of IL-1α and IL-1β, by repeating the transwell assay. Instead, we added “naïve” breast cancer cells, knockdown for IL-1α and IL-1β, or wild-type (
As part of developing novel treatment approaches, AMD3100-mediated susceptibility of breast cancer cells to carboplatin was integral. The final set of studies determined the clinical relevance of AMD3100 as a breast cancer therapeutic by examining its ability to promote susceptibility to the chemotherapeutic, carboplatin, despite the presence of MSCs. Breast cancer cells co-cultured with MSCs (
To validate our in vitro studies, female nude BALB/c mice were injected subreast cancerutaneously with 106 matrigel-resuspended MDA-MB-231 alone or in combination with 106 MSCs, as per the experimental design described in
The chemotherapeutic agents and the CXCR4 antagonists can be administered by means known in the art, such as intravenously or by infusion in pharmaceutically acceptable carriers as are known in the art.
All tissue culture media were purchased from Gibreast cancero (Grand Island, N.Y.), fetal calf serum (FCS) from Hyclone Laboratories (Logan, Utah), IL-1α/β siRNA, Ficoll-Hypaque and AMD3100 from Sigma (St. Louis, Mo.), propidium iodide from BD Biosciences (San Jose, Calif.) and carboplatin from Teva Parenteral Medicines (Irvine, Calif.).
Goat anti-SDF-1α and anti-IL-1RI from R&D Systems (Minneapolis, Minn.), rabbit anti-CXCR4 from Affinity Bioreagents (Golden, Colo.), rabbit anti-caspase-3 from BD Pharmingen (San Diego, Calif.), mouse anti-cytokeratin, -β-actin mAb, horseradish-peroxidase (HRP)-conjugated anti-rabbit, -goat and -mouse IgG were purchased from Sigma. CellTiter-Blue cell viability assay was purchased from Promega (Madison, Wis.) and CyQUANT Cell Proliferation kit from Invitrogen (Carlsbad, Calif.).
T47D and MDA-MB-231 were purchased from American Type Culture Collection (ATCC; Manassas, Va.) and cultured in accordance with manufacturer's instructions.
MSCs were cultured from bone marrow aspirates. The use of human bone marrow aspirates followed a protocol approved by the Institutional Review Board of The University of Medicine and Dentistry of New Jersey-Newark Campus. Unfractionated bone marrow aspirates were cultured in DMEM with 10% FCS (D10 media) in Falcon 3003 dishes. After 3 days, red blood cells and granulocytes were removed with Ficoll Hypaque. After four cell passages, the adherent cells were asymmetric, CD14−, CD29+, CD44+, CD34−, CD45−, SH2+, prolyl-4-hydroxylase.
The shRNA vector, pPMSKH1-SDF-1/KC (wild-type and mutant), for CXCL12 was previously described in the art. pSUPER-CXCR4 (wild-type and mutant) shRNA vector for knockdown of CXCR4 was kindly provided by Dr. Si-Yi Chen (Baylor University). Breast cancer cells or MSCs were co-transfected with pTK-Hyg and pPMSKH1-SDF-1/KC or pSUPER-CXCR4 (both either wild-type or mutant) and then selected with hygromycin or G418. All knockdown cultures were maintained in media containing hygromycin. Levels of CXCL12 and CXCR4 protein expression were determined by western blot to validate knockdown.
Whole cell extracts from breast cancer cells and MSCs were prepared in accordance with accepted methods and 20 μg were analyzed by western blots using 4-20% SDS-PAGE (Bio-Rad, Hercules, Calif.). The proteins were transferred onto polyvinylidene difluoride membranes (Perkin Elmer Life Sciences, Boston, Mass.). Membranes were incubated overnight with primary antibodies and then detected the following day by 2 hour incubation with HRP-conjugated IgG. All primary and secondary antibodies were used at final dilutions of 1/1000 and 1/2000, respectively. HRP was developed with chemiluminscence detection reagent (Perkin Elmer Life Sciences). The membranes were stripped with Restore Stripping Buffer (Pierce, Rockford, Ill.) for reprobing with other antibodies.
Cultures of breast cancer cells and MSCs, alone or in co-culture, from each experimental setup were assayed for cellular proliferation and viability using the CyQuant Cell Proliferation Assay Kit (Molecular Probes; Eugene, Oreg.) and CellTiter-Blue Cell Viability Assay (Promega), respectively, according to manufacturer's specific instructions.
For determination of proliferation, cells grown in 96-well plates were frozen overnight at −70° C. The next day, thawed cells were incubated in CyQuant GR dye/cell-lysis buffer for 5 minutes at room temperature, and examined using a fluorescence microplate reader at 480 nm excitation/520 nm emission. Proliferation was calculated from a standard curve of known numbers of breast cancer cells and MSCs. To accurately assess the proliferation of breast cancer cells, but not MSC proliferation in co-culture, proliferation of MSCs grown alone were subtracted from the total cellular proliferation recorded.
For assessment of viability, CellTiter-Blue reagent was added to cells grown in 96-well plates, and then incubated for 4 hours at 37° C. Following incubation, wells were read using a fluorescence microplate reader at 560 nm excitation/590 nm emission. Percent viability was calculated from a reference using untreated healthy cells, which were considered 100% viable, and cell-free wells containing reagent alone, which were considered 0% viable.
Breast cancer cells and MSC co-cultures were incubated with anti-cytokeratin primary and FITC-anti IgG secondary antibodies to label the breast cancer cell fraction. Cells were then treated with RNase A (1 mg/ml) and fixed with cold 70% ethanol. Cells were stained with 20 μg/ml propidium iodide (PI) solution and transferred to round bottom tubes for DNA analysis by BD FACScan. Double positive cells identified only the desired cellular fraction, consisting of breast cancer cells that incorporated the DNA dye. Cultures incubated with FITC anti-IgG alone were used as isotype controls. All analyses were performed using BD CellQuest software and percent statistics were given.
Breast cancer cells (5×104) were added to the outer well of 24-well transwell cultures, 0.4 μM insert (BD Falcon). In parallel, breast cancer cells were separated from co-cultures with MSCs by positive selection with anti-cytokeratin-conjugated; pan anti-mouse IgG Dynabeads (Invitrogen). MSCs from the negative fraction were then added to the inner wells of the transwell chamber in order to determine the effects of MSC-derived soluble factors on breast cancer cell proliferation. For neutralization of soluble IL-1α and IL-1β, anti-IL-1RI was titrated into the 24-well plates. Neutralization of breast cancer cell proliferation was observed at a concentration of 1 μg/μl.
Cytokine production by MSCs in the transwell assay was assessed using the Human Cytokine Antibody Array 5 (RayBiotech; Norcross, Ga.). Briefly, after 48 hours of culture, the transwell was removed and the breast cancer cell growth media collected for cytokine determination. Background levels obtained with media alone were subtracted. The densities of spots were quantitated with UN-SCAN-IT densitometry software (Silk Scientific; Orem, Utah). Cytokines demonstrating differential expression were normalized to internal positive controls and presented as fold change relative to an internal control, arbitrarily assigned a value of 1.
Transient Transfection of IL-1α/β siRNA
IL-1α and IL-1β siRNA duplexes (Sigma) were used to knockdown IL-1 production in breast cancer cells, prior to culture with MSC transwell inserts. MDA-MB-231 (5×104) were seeded in 24-well plates, and after 24 hours, 100 nM IL-1α and IL-1β siRNA was delivered via DharmaFECT Transfection reagent (Dharmacon; Lafayette, Colo.). siRNA sequences were as follows: IL-1α|5′-guc auc aaa gga uga ugc u-3′|; IL-1β|5′-gau guc ugg ucc aua uga a-3′|. Knockdown was confirmed by PCR.
Female nude BALB/c mice (4 weeks) were obtained from Harlan Laboratories (Indianapolis, Ind.) and housed in a laminar flow hood at an AALAC-accredited facility. The use of mice was approved by the Institutional Animal Care and Use Committee, New Jersey Medical School (Newark, N.J.). MDA-MB-231 (106), alone or in combination with MSCs (106), in 0.1 mL and equal volume of BD Matrigel (BD Biosciences, Bedford, Mass.) were injected by subreast cancerutaneous route in the nude mice (Day 0, D0). After 4 days, the tumors were injected with AMD3100 alone or in combination with IL-1ra or vehicle (hereafter termed ‘treatment’). Mice were given a second ‘treatment’ at D8 along with a first dose of chemotherapy delivered intra-peritoneally. A second dose of chemotherapy was given at D10 and the mice euthanized at D12.
Statistical data analyses were performed with analysis of variance and Tukey-Kramer multiple comparisons test. p<0.05 was considered significant.
The effect on cycling of breast cancer cells by AMD3100 is shown in
In
Similarly, in
In
1. Braun, S., and B. Naume. 2005. Circulating and Disseminated Tumor Cells. J. Clin. Oncol. 23: 1623-1626.
2. Gluck, S. 1995. Autologous transplantation for patients with advanced breast cancer with emphasis on bony metastasis. Can. J. Oncol. 5 Suppl 1: 58-62.
3. Habeck, M. 2000. Bone-marrow analysis predicts breast-cancer recurrence. Mol. Med. Today 6: 256-257.
4. Mansi, J. L., U. Berger, T. McDonnell, A. Pople, Z. Rayter, J. C. Gazet, and R. C. Coombes. 1989. The fate of bone marrow micrometastases in patients with primary breast cancer. J. Clin. Oncol. 7: 445-449.
5. Patel, S. A., A. C. Heinrich, B. Y. Reddy, B. Srinivas, N. Heidaran, and P. Rameshwar. 2008. Breast cancer biology: the multifaceted roles of mesenchymal stem cells. J. Oncol. 2008: 425895.
6. Naume, B., X. Zhao, M. Synnestvedt, E. Borgen, H. G. Russnes, O. C. Lingjaerde, M. Stromberg, G. Wiedswang, G. Kvalheim, R. Karesen, J. M. Nesland, A. L. Borresen-Dale, and T. Sorlie. 2007. Presence of bone marrow micrometastasis is associated with different recurrence risk within molecular subtypes of breast cancer. Mol. Oncol. 1: 160-171.
7. Katz, D., and D. Aharoni. 2004. Lytic Lesions in Breast Cancer. N Engl J Med 351: 2850.
8. Dai, H., L. van't Veer, J. Lamb, Y. D. He, M. Mao, B. M. Fine, R. Bernards, M. van de Vijver, P. Deutsch, A. Sachs, R. Stoughton, and S. Friend. 2005. A Cell Proliferation Signature Is a Marker of Extremely Poor Outcome in a Subpopulation of Breast Cancer Patients. Cancer Res. 65: 4059-4066.
9. Corcoran, K. E., N. Patel, and P. Rameshwar. 2007. Stromal Derived Growth Factor-1{alpha}: Another Mediator in Neural-Emerging Immune System through Tac1 Expression in Bone Marrow Stromal Cells. J. Immunol. 178: 2075-2082.
10. Moharita, A. L., M. Taborga, K. E. Corcoran, M. Bryan, P. S. Patel, and P. Rameshwar. 2006. SDF-1{alpha} regulation in breast cancer cells contacting bone marrow stroma is critical for normal hematopoiesis. Blood 108: 3245-3252.
11. Oh, H. S., A. Moharita, J. G. Potian, I. P. Whitehead, J. C. Livingston, T. A. Castro, P. S. Patel, and P. Rameshwar. 2004. Bone Marrow Stroma Influences Transforming Growth Factor-{beta} Production in Breast Cancer Cells to Regulate c-myc Activation of the Preprotachykinin-I Gene in Breast Cancer Cells. Cancer Res. 64: 6327-6336.
12. Corcoran, K. E., K. A. Trzaska, H. Fernandes, M. Bryan, M. Taborga, V. Srinivas, K. Packman, P. S. Patel, and P. Rameshwar. 2008. Mesenchymal Stem Cells in Early Entry of Breast Cancer into Bone Marrow. PLoS ONE 3: e2563.
13. Patel, S. A., J. R. Meyer, S. J. Greco, K. E. Corcoran, M. Bryan, and P. Rameshwar. 2010. Mesenchymal Stem Cells Protect Breast Cancer Cells through Regulatory T Cells: Role of Mesenchymal Stem Cell-Derived TGF-{beta}. J. Immunol. 184: 5885-5894.
14. Dennis, J. E., and P. Charbord. 2002. Origin and differentiation of human and murine stroma. Stem Cells 20: 205-214.
15. Helbig, G., W. Kent, P. Bhat-Nakshatri, S. Kumar, H. Kishimoto, K. D. Miller, H. E. Broxmeyer, and H. Nakshatri. 2003. NF-kB Promotes Breast Cancer Cell Migration and
Metastasis by Inducing the Expression of the Chemokine Receptor CXCR4. J. Biol. Chem. 278: 21631-21638.
16. Muller-Sieburg, C. E., and E. Deryugina. 1995. The stromal cells' guide to the stem cell universe. Stem Cells 13: 477-486.
17. Orimo, A., P. B. Gupta, D. C. Sgroi, F. Arenzana-Seisdedos, T. Delaunay, R. Naeem, V. J. Carey, A. L. Richardson, and R. A. Weinberg. 2005. Stromal fibroblasts present in invasive human breast carcinomas promote tumor growth and angiogenesis through elevated SDF-1/CXCL12 secretion. Cell 121: 335-348.
18. Rao, G., P. S. Patel, S. P. Idler, P. Maloof, P. Gascon, J. A. Potian, and P. Rameshwar. 2004. Facilitating Role of Preprotachykinin-I Gene in the Integration of Breast Cancer Cells within the Stromal Compartment of the Bone Marrow: A Model of Early Cancer Progression. Cancer Res. 64: 2874-2881.
19. Deans, R. J., and A. B. Moseley. 2000. Mesenchymal stem cells: biology and potential clinical uses. Exp. Hematol. 28: 875-884.
20. von Tell, D., A. Armulik, and C. Betsholtz. 2006. Pericytes and vascular stability. Exp. Cell Res. 312: 623-629.
21. Potian, J. A., H. Aviv, N. M. Ponzio, J. S. Harrison, and P. Rameshwar. 2003. Veto-Like Activity of Mesenchymal Stem Cells: Functional Discrimination Between Cellular Responses to Alloantigens and Recall Antigens. J. Immunol. 171: 3426-3434.
22. Foss, B., T. Hervig, and O. Bruserud. 2009. Connexins are active participants of hematopoietic stem cell regulation. Stem Cells Dev. 18: 807-812.
23. Greco, S. J., C. Zhou, J. H. Ye, and P. Rameshwar. 2007. An Interdisciplinary Approach and Characterization of Neuronal Cells Transdifferentiated from Human Mesenchymal Stem Cells. Stem Cells and Dev. 16: 811-826.
24. Lapteva, N., A. G. Yang, D. E. Sanders, R. W. Strube, and S. Y. Chen. 2004. CXCR4 knockdown by small interfering RNA abrogates breast tumor growth in vivo. Cancer Gene Ther. 12: 84-89.
25. 2007. Plerixafor: AMD 3100, AMD3100, JM 3100, SDZ SID 791. Drugs R. D. 8: 113-119.
26. Mirisola, V., A. Zuccarino, B. E. Bachmeier, M. P. Sormani, J. Falter, A. Nerlich, and U. Pfeffer. 2009. CXCL12/SDF1 expression by breast cancers is an independent prognostic marker of disease-free and overall survival. Eur. J. Cancer 45: 2579-2587.
27. Smith, M. C. P., K. E. Luker, J. R. Garbow, J. L. Prior, E. Jackson, D. Piwnica-Worms, and G. D. Luker. 2004. CXCR4 Regulates Growth of Both Primary and Metastatic Breast Cancer. Cancer Res. 64: 8604-8612.
28. Teicher, B. A., and S. P. Fricker. 2010. CXCL12 (SDF-1)/CXCR4 Pathway in Cancer. Clin. Cancer Res. 16: 2927-2931.
29. Kalatskaya, I., Y. A. Berchiche, S. p. Gravel, B. J. Limberg, J. S. Rosenbaum, and N. Heveker. 2009. AMD3100 Is a CXCR7 Ligand with Allosteric Agonist Properties. Mol. Pharmacol. 75: 1240-1247.
30. Chan, J. L., K. C. Tang, A. P. Patel, L. M. Bonilla, N. Pierobon, N. M. Ponzio, and P. Rameshwar. 2006. Antigen-presenting property of mesenchymal stem cells occurs during a narrow window at low levels of interferon-{gamma}. Blood 107: 4817-4824.
31. Kang, H. S., M. Habib, J. Chan, C. Abavana, J. A. Potian, N. M. Ponzio, and P. Rameshwar. 2005. A paradoxical role for IFN-gamma in the immune properties of mesenchymal stem cells during viral challenge. Exp. Hematol. 33: 796-803.
32. Tang, K. C., K. A. Trzaska, S. V. Smirnov, S. V. Kotenko, S. K. Schwander, J. J. Ellner, and P. Rameshwar. 2008. Down-Regulation of MHC II in Mesenchymal Stem Cells at High IFN-{gamma} Can Be Partly Explained by Cytoplasmic Retention of CIITA. J Immunol. 180: 1826-1833.
33. Sakaguchi, Y., I. Sekiya, K. Yagishita, S. Ichinose, K. Shinomiya, and T. Muneta. 2004. Suspended cells from trabecular bone by collagenase digestion become virtually identical to mesenchymal stem cells obtained from marrow aspirates. Blood 104: 2728-2735.
This application claims the benefit of U.S. Provisional application No. 61/289,561, filed Dec. 23, 2009, which is incorporated herein by reference in its entirety.
The research disclosed in this application was funded in part by Department of Defense Grants W81XWH-0810561 and W81XWH-0610689. Accordingly, the US government has certain rights in the invention.
Number | Date | Country | |
---|---|---|---|
61289561 | Dec 2009 | US |