Multiple myeloma (MM), characterized by an accumulation of malignant plasma cells in the bone marrow (BM), is the most common bone malignancy in the United States. Although chemotherapy is the most effective treatment, the majority of patients experience relapse and die of the disease. The major cause of treatment failure is the development of multidrug resistance. The BM microenvironment confers MM chemoresistance. Deducing how the BM creates a microenvironment friendly to MM cells and confers resistance is thus the key to overcoming drug resistance and greatly improving patient survival. Recently we discovered that human MM- derived MIF (macrophage migration inhibitory factor) regulates the homing or affinity of MM cells for BM and, as a result, their sensitivity to chemotherapy. MIF is highly expressed by human MM cells and the expression levels positively correlate with advanced disease and poor survival in patients. Surprisingly, knocking down MIF in MM cells impaired their adhesion to BM stromal cells (BMSCs) in vitro and led to formation of extramedullary tumors in SCID mice. More importantly, MIF-knockdown human MM cells were more sensitive, compared with control cells, to chemotherapy in SCID mice because chemotherapy effectively eradicated extramedullary but not intramedullary tumors in the host. Inhibiting MIF activity in MM cells (cell lines and primary MM cells from patients) by the MIF inhibitor (4-IPP) or neutralizing mAbs also resulted in impaired adhesion to BMSCs in vitro and formation of extramedullary tumors in SCID and SCID-hu mice without affecting tumor burdens. Furthermore, MM-(transwell)-conditioned human BMSCs mediated stronger adhesion to MM cells, provided greater protection to MM cells against chemotherapy-induced apoptosis, and attracted more monocytes than MIF-knockdown MM-conditioned BMSCs. Based on these novel findings, we hypothesize that high MIF in MM cells contributes to poor patient survival by enhancing the affinity of MM cells for BM and by conditioning BM to become a MM-friendly microenvironment, leading to enhanced MM growth and survival and induction of drug resistance.
Aim 1 will elucidate the mechanisms of MM-derived MIF in regulating MM homing to and affinity for BM.
Aim 2 will determine the importance and mechanisms of MM- derived MIF in conditioning BM to become a MM-friendly microenvironment, and Aim 3 will determine and validate the role of MM-expressing MIF in patients with MM. Accomplishing these aims will provide the justification and tools for developing novel and effective strategies to target MIF to improve the therapeutic efficacy of chemotherapy. The proposed studies will also lead to a better understanding of the fundamental mechanisms underlying MM homing or metastasis to the bone and MM conditioning the microenvironment, and could pave the way to the first substantial improvements in current MM treatment by mobilizing MM cells away from the protective BM microenvironment.

Public Health Relevance

In this project we will determine the roles of a special molecule, called macrophage migration inhibitory factor (MIF), in regulating tumor cell affinity for the bone marrow of patients with multiple myeloma. We hypothesize that MIF is crucial to myeloma cell retention in the bone marrow and to myeloma drug resistance, and targeting MIF will significantly improve chemotherapy efficacy in patients. In this project, we will examine how MIF induces drug resistance in myeloma cells and how to target this molecule to enhance myeloma cell sensitivity to chemotherapy in patients.

Agency
National Institute of Health (NIH)
Institute
National Cancer Institute (NCI)
Type
Research Project (R01)
Project #
5R01CA211073-05
Application #
10078263
Study Section
Tumor Microenvironment Study Section (TME)
Program Officer
Howcroft, Thomas K
Project Start
2017-01-01
Project End
2021-12-31
Budget Start
2021-01-01
Budget End
2021-12-31
Support Year
5
Fiscal Year
2021
Total Cost
Indirect Cost
Name
Methodist Hospital Research Institute
Department
Type
DUNS #
185641052
City
Houston
State
TX
Country
United States
Zip Code
77030
Wang, Q; Lu, Y; Li, R et al. (2018) Therapeutic effects of CSF1R-blocking antibodies in multiple myeloma. Leukemia 32:176-183
Lu, Yong; Wang, Qiang; Xue, Gang et al. (2018) Th9 Cells Represent a Unique Subset of CD4+ T Cells Endowed with the Ability to Eradicate Advanced Tumors. Cancer Cell 33:1048-1060.e7
Ma, Xingzhe; Bi, Enguang; Huang, Chunjian et al. (2018) Cholesterol negatively regulates IL-9-producing CD8+ T cell differentiation and antitumor activity. J Exp Med 215:1555-1569
Bi, Enguang; Ma, Xingzhe; Lu, Yong et al. (2017) Foxo1 and Foxp1 play opposing roles in regulating the differentiation and antitumor activity of TH9 cells programmed by IL-7. Sci Signal 10:
Yang, Jing; Liu, Zhiqiang; Liu, Huan et al. (2017) C-reactive protein promotes bone destruction in human myeloma through the CD32-p38 MAPK-Twist axis. Sci Signal 10: