Abstract
Myeloid-derived suppressor cells (MDSCs) are a heterogeneous, immature myeloid cell population with the ability to suppress innate and adaptive immune responses that promote tumor growth. MDSCs are increased in patients with multiple myeloma (MM) and have bidirectional interaction with tumors within the MM microenvironment. MM-MDSCs promote MM tumor growth and induce immune suppression, while conversely, MM cells induce MDSC development and survival. Although the role of MDSCs in infections, inflammatory diseases and solid tumors has been extensively characterized, their tumor-promoting and immune-suppressive role in MM and the MM microenvironment is only beginning to emerge. The presence and activation of MDSCs in MM patients has been well documented, however direct actions and functional consequences of MDSCs on cancer cells is poorly defined. Immunosuppressive MDSCs play an important role in tumor-progression primarily because of their capability to promote immune-escape, angiogenesis, drug resistance and metastasis. However, their role in the bone marrow (BM), the primary MM site, is poorly understood. MM remains an incurable malignancy, and it is likely that the BM microenvironment protects MM against chemotherapy agents and the host immune system. A growing body of evidence suggests that host immune cells with a suppressive phenotype contribute to a myeloma immunosuppressive network. Among the known suppressor cells, MDSCs and T regulatory cells (Tregs) have been found to be significantly increased in myeloma patients and their levels correlate with disease stage and clinical outcome. Furthermore, it has been shown that MDSC can mediate suppression of myeloma-specific T-cell responses through the induction of T-cell anergy and Treg development in the MM microenvironment. Here, we review clinical correlations and the preclinical proof-of-principle data on the role of MDSCs in myeloma immunotolerance and highlight the mechanistically relevant MDSC-targeted compounds and their potential utility in a new approach for anti-myeloma therapy.
Keywords: Multiple myeloma, Myeloid-derived suppressor cells, Immunotherapy, Pre-clinical models
1. Introduction
Despite the advent of novel agents and doubling of survival rates, multiple myeloma (MM) is still considered an incurable malignancy 1–3. MM is characterized by generalized immune suppression that contributes to susceptibility to infection as well as tumor progression 4–6 and the discovery that anti-MM novel agents (i.e., bortezomib and lenalidomide) retain immunomodulatory properties underlies the role of the deregulated immune effector cells in this disease 7–10. T-lymphocyte and natural killer mediated immunotherapy have been evaluated or are currently under investigation as potential new avenues to overcome the myeloma immunosuppressive network and boost a specific anti-MM immune response 11–13.
A well-recognized feature of MM is the bidirectional interaction between malignant plasma cells and the bone marrow microenvironment, which provides a protective niche from the patient’s immune system and chemotherapy agents. Importantly, inadequate prediction of myeloma progression based on gene-expression profiling of isolated malignant plasma cells underscores the likely essential role for non-plasma cells components in MM disease progression and survival 14. While MM is a more widespread disease compared to smoldering multiple myeloma (SMM) and monogammopathy of unknown significance (MGUS), it harbors the same genetic defects as the other two subtypes of plasma cell dyscrasias 15,16 suggesting that genetic mutations are necessary but not enough for developing symptomatic myeloma. Transformation of MGUS to MM seems to be caused by a developing permissive myeloma microenvironment which leads to “immune escape” and advancement toward full-blown myeloma 12,17. Also, the myeloma microenvironment has a substantial role in chemotherapy resistance and thereby the persistence of residual disease, which is the source of frequent relapses leading to poor clinical outcomes 18–21.
The MM microenvironment includes osteoclast, osteoblasts, endothelial and immune cells with the structural support of an extracellular matrix, adhesion molecules and cytokines 21. Increased immune suppressor cells have been reported in the bone marrow of myeloma patients, which correlates with clinical outcomes, emphasizing the important role of these cells in providing the “immune escape” that favors myeloma progression. Myeloid-derived suppressor cells (MDSCs) are a heterogeneous population of immature myeloid cells that accumulate in different cancer types, including MM. Besides immune regulation, MDSCs promote tumor angiogenesis and tumor growth through the secretion of cytokines and growth factors. Recently, the role of MDSCs in tumor-induced immunosuppression has been established in a variety of malignancies. MDSCs are a heterogeneous mixture of myeloid cells in different maturation stages with the antigen-presenting ability that contributes to immune evasion of cancer cells 22–25. They are comprised of immature granulocytes and precursors of macrophages and dendritic cells that promote tumor growth by suppressive adaptive immunity, leading to suppression of CD4 and CD8 cell-mediated immunity 22,26,27. These cells secrete arginase, which is able to deplete the microenvironment of arginine, an essential amino acid for T-cell activity. Moreover, MDSCs inhibit T-cell receptors by nitrosylation and reactive oxygen species (ROS) release 28. MDSCs are activated by a key transcription factor, signal transducer and activator of transcription 3 (STAT3) 29. This review presents a summary of preclinical data and clinical correlations and highlights the MDSCs as an important target for therapeutics development for patients with MM.
2. MDSC evolution and phenotype
In mice, MDSCs are classified according to presence of Ly-6C or Ly-6G on their membrane, respectively. In humans, they are characterized as CD33+ cells, common myeloid marker, and CD11b+ with no marker for mature lymphoid or myeloid on their membrane including HLA-DR. They can be divided in two main groups based on CD14 positivity; granulocyte MDSCs (G-MDSCs) are CD11b+ CD14− CD33+ CD15+ HLA-DRlow/− and monocytic-MDSC (M-MDSCs) that are CD11b+ CD14+ CD33+ HLA-DRlow/− 30,31. Detailed information about the different subsets of MDSCs can be found elsewhere 32.
MDSC evolution from hematopoietic progenitor cells (HPC) includes a primarily MDSC expansion phase and a second phase of MDSC activation. The first phase is mediated by a variety of cytokines, including GM-CSF, M-CSF, G-CSF, IL-6 and vascular endothelial growth factor (VEGF), produced by tumors or bone marrow stromal cells, and activation of STAT3 and STAT5. These signals promote proliferation of immature myeloid cells from hematopoietic progenitor cells and prevent their differentiation to more mature myeloid cells. The second phase involves MDSC activation through STAT1 and NF-kB pathway activation which involves a different set of cytokines such as IL-13, toll-like receptor (TLR) ligands and leads to upregulation of arginase and nitric oxide (NO). Importantly, to mount a full MDSC effect during tumorigenesis or chronic inflammation the simultaneous activation of two phases is required. In myeloma the G-MDSC forms the predominant MDSC population in bone marrow as well as peripheral blood samples as opposed to M-MDSC 33. MDSCs are phenotypically different in humans and mice.
3. Clinical correlations
Higher Peripheral blood MDSC levels correlates with higher tumor stage and volume at diagnosis in a variety of solid tumors as well as malignant hematology neoplasms and confers chemoresistance34,35. On multiple reports, MDSCs were functionally and phenotypically different in MM patients compared to patients with MGUS and normal individuals 33,36,37. Also, CD14+HLA-DR−/low M-MDSCs increase in newly diagnosed and relapsed MM compared to a myeloma patient who is in remission. More importantly the MDSC burden correlates with MM stages and poor clinical outcome with proteasome inhibitors38. A balanced amalgam of different immune system compartments including NK cells, Tregs, MDSCs, Th17 and T helper cells correlates with long-term remission after initial anti-myeloma therapy 39,40. Details regarding the potential role of MDSCs as a population influencing the GVHD as well as Donor Lymphocyte Infusion (DLI) outcome after stem cell transplant is also emerging (section 5.2).
4. Role of MDSCs in the tumor-induced immunosuppressive network
While MDSCs are rare or absent in healthy individuals, they play a prominent role in some physiologic process like fetal allotolerance during pregnancy as well as in pathologic conditions such as limiting the inflammatory response due to infections 41, and the development of immunotolerance to different types of malignancies42,43. During the process of metastasis, the shift of myeloid-progenitor cells to MDSCs is a transition observed in the microenvironment of organs such as lung or liver now viewed to be a critical step in enabling establishment of new metastases 44.
The myelomagenesis process, from MGUS to symptomatic myeloma, creates a suppressed immunome through a complicated cross-talk of the immune effecter cells and cytokines. The main deregulated elements of MM immunome are: Tregs, dendritic cells (DC), MDSCs and T-helper 17 (TH17) 45,46. However, MDSCs are emerging as the main regulatory population, playing an essential role in disease progression through the orchestration of immune suppression (Figure 1) 47.
Figure 1.
Myeloid derived suppressor cells (MDSCs) interaction with myeloma cells and multiple mechanisms used by these cells to dampen anti-myeloma immunity. AMPK: AMP-activated protein kinase; MCP-1: Monocyte chemoattractant protein-1; MCL-1: Myeloid Cell Leukemia 1; TGF: Transforming growth factor.
4.1. Suppressive effect of MDSC on T cell function
MDSCs suppress T-cell function through three main mechanisms: 1) arginine depletion 2) production of high levels of nitric oxide (NO) and 3) production of reactive oxygen species (ROS). MDSCs contain high levels of argeninase which depletes the microenvironment of L-arginine, leading to impaired production of the CD3ζ chain, an integral part of T-cell receptors (TCR). Also, L-arginine depletion causes T-cell proliferation arrest by affecting cell cycle regulators 48,49. NO can increase T-cell apoptosis 50,51, and can also inhibit the IL-2 induced inflammatory cascade by inhibiting phosphorylation of downstream IL-2 pathways such as Stat5 or Jak3 52. Also, accumulation of ROS can suppress T-cell functions as peroxynitrite is known to lead to receptor modification including TCR/MHC modification. Lastly, MDSCs upregulate the immunoregulatory molecules, Cox-2 and prostaglandin E2 (PGE2) 53.
4.2. Stimulatory effect of MDSC on regulatory T cells
MDSCs can promote regulatory T-cells (Tregs) and interfere with lymph node homing of naïve T-cells 54,55. MDSCs also promote the expansion of induced Treg (iTreg) cells from naïve Tregs through TGF-β-dependent and -independent mechanisms which involve surface receptor interaction of CD40-CD40L or IL-10 and IFN-gamma secretion, respectively 56. The latter may be important in the MDSC suppression of NK cell as well as natural killer T-cell (NKT cell) mediated tumor responses in the myeloma microenvironment 57.
4.2.1. The MDSC role in NK cell anergy
Cancer-induced MDSCs can induce anergy of NK cells via membrane-bound TGF-β1 58. Also, prostaglandins are involved in MDSC-mediated NK cell suppression 59. There is also an essential role for transforming growth factor β (TGF-β) in the biology of MDSCs and NK cell suppression. TGF-β1 is a cytokine with potent immunosuppressive effects which is overexpressed in many tumors. Inhibitors of TGF-β1 suppress development and function of MDSCs and enhance the efficacy of anti-tumor vaccination 60. More importantly, MDSCs have been shown to induce NK anergy through membrane-bound TGF-β1, while TGF-β1 inhibition leads to more tumor-toxic NK cells 58. Gr-1+CD11b+ cells contribute to TGFβ-mediated metastasis through enhancing tumor cell invasion and metastasis 61. Tumor exosomal TGF-β1 leads to MDSCs accumulation and tumor growth promotion 62. NK surveillance and cytotoxicity against MM decreases as disease progresses 63–65.
4.3. MDSCs enhance the tumor-promoting effect of T helper 17
CD4+ T cells, upon activation and expansion, develop into different T helper cell subsets with different cytokine profiles. A subset of IL-17-producing effector T helper cells, called Th17 cells, has now been characterized which favors angiogenesis and tumor growth in a variety of malignancies including MM 66,67. MDSCs recruit Th17 cells and increases IL-17 production 68,69. NLRP3 inflammasome activation has been suggested as the mechanism of MDSC-mediated Th17 activation 70,71.
4.4. MDSC expansion as a counter-regulatory mechanism after anti-myeloma therapy
The alkylating agents melphalan and cyclophosphamide induce a robust initial antitumor immune response, which however is also associated with M-MDSC expansion, and an attenuated antitumor CD4+ T cell response through activation of PD1-PD-L1 axis 72. This may provide a conceptual platform to test the potential synergistic effect of alkylating agents and MDSC-directed therapies to enhance the anti-myeloma immune response.
IMiDs modify the myeloma immunome in a specific way. Busch et al. provided a comprehensive analysis of the immune system in 68 myeloma patients who were treated with these agents73. These investigators showed increased numbers of Tregs and MDSCs, consistent with observations reported by others 74,75. The observations include a demonstrated increase in both the number and activity of effector T and NK cells along with elevated HLA-DR expression, consistent with other studies 76–78. These findings suggest that MDSC expansion exists as a counter-regulatory mechanism elicited by certain anti-myeloma agents, and suggests concomitant MDSC suppression could be an important strategy to prevent relapse after autologous stem cell transplant and chemoimmunotherapy 79. Interestingly, dexamethasone potentiates MDSC immunosuppressive effects 80,81 which is considered a mechanism of action for glucocorticoids in the allograft setting 82.
5. Role of MDSC in Myeloma Immunotherapy
5.1. MDSC impairs the efficacy of Myeloma vaccine and check point inhibitors
The premise of implementing different modalities of immunotherapy in MM is based upon the observation that myeloma evolution that correspond by progressive loss of T cell repertoire surveillance, suppression of antigen-presenting function and increased number and function of inhibitory accessory cells. A variety of strategies to reverse the myeloma-induced immunosuppression has been developed and are in clinical trials.
Dendritic cell (DC) has a critical role in the antigen presenting and developing anti-myeloma immune response. DC vaccination using different tumor antigens can render an active and potentially long lasting immunization against the tumor. In this method DCs are loaded with tumor antigen exhibit a potent T-cell stimulatory activity currently is under study. However, the vaccine trials have had mixed result. Studies suggest MDSC impair the immune response from DC vaccines 83. This is important because the current method of DC ex vivo growth may collect MDSC. The most common approach to generate DC vaccine is DC ex vivo expansion before load them with tumor antigen. In this method most protocols uses CD14+ cells to generate their DC produce from primary patient samples. Although this method can generate active DCs from normal individuals but has limitation to generate matures DC population from cancer patients that correlate with abundance of monocytes with altered surface marker expression (CD14+HLA-DRlo/neg) which shows similarity with increased MDSC population in patient with cancer84. This underlines the importance of MDSC-suppressive strategies to augment the DC vaccination effect.
MDSCs from peripheral blood and bone marrow of myeloma patient contains more PD-L1 in compare to normal individuals 85. Indeed, PD-L1 expression on MDSC is higher than antigen-presenting cells including dendritic cells and PD-1/PD-L1 blockade limits myeloma-promoting effect of MDSC 86. In another study, Disruption of MDSC trafficking abrogate the response to PD1 inhibition 87, providing the framework for evaluation of check point inhibitor combination with MDSC-targeted agents.
5.2. MDSC role in GVHD and stem cell transplant outcomes
Allogeneic hematopoietic stem cell transplant and donor lymphocyte infusion (DLI) confers a prolonged remission in a subgroup of myeloma patients. This is due to “graft versus myeloma” effect as an effective immunologic therapy. However, the application of this modality of therapy is limited by high transplant-related mortality and GVHD. Interestingly, MDSCs are collected with G-CSF-mobilized stem cell collection from healthy donors 88, and the fraction of MDSC in the collected peripheral blood CD34+ cells inversely correlates with occurrence of acute Graft vs. Host Disease (GVHD) 89 89. Messmann et al. demonstrated in-vitro-generated MDSCs prevent murine GVHD by tipping the TH2/TH1 balance in favor of TH2 cells without disabling antitumor cytotoxicity of the graft. This effect is independent of MHC class I and may be exploited to treat GVHD without affecting graft-versus-tumor (GVT) effect by expansion and infusion of autologous MDSCs from an individual patient 90. In another study the number of circulating peripheral blood MDSCs negatively correlates with donor lymphocyte infusion (DLI) efficacy in the post allogeneic stem cell transplant setting 91. The intensity of the conditioning regimen affects the expansion rate of MDSC populations post-transplant inversely, which may contribute to the higher GVHD incidence by myeloablative compared to a non-myeloablative conditioning regimens 92. Moreover, MDSCs populate the bone marrow and increase Treg activity, leading to delayed T-cell immunity 93.
5.3. MDSCs role in future anti-myeloma CAR-T cells therapies
Genetically engineered T-cells with chimeric antigen receptors (CAR-T) endow T cell populations with high antigen specificity and remarkable anti-tumor activity in acute leukemia94,95. Currently, there are two ongoing clinical trials using CAR-T targeting the kappa light chain and CD138 testing the effectiveness of this modality in MM. There are more anti-myeloma CARs in pre-clinical development using the B cell maturation antigen (BCMA) and cell surface glycoprotein CS1 antigens 96. This modality of therapy is emerging as an effective therapy with a curative potential of allogeneic stem cell transplant without morbidities related to graft versus host disease (GVHD). However, there have been challenges to implement this modality. Although, CAR-T cells are designed based on a restrictive antigen on tumor cells and are invariably able to lyse tumor cells in-vitro, they most often fail to expand and mount an anti-tumor effect in-vivo 97. Mounting evidence suggests the suppressive role of MDSCs against in-vivo expansion of these engineered T-cells 98. It has been observed that MDSCs are able to inhibit CAR-T cell expansion in vitro and the extent of T-cell proliferation correlates inversely with circulating MDSC counts in a pivotal trial of CAR-T therapy in acute leukemia 95. Importantly, suppression of MDSCs by all-trans retinoic acid (ATRA) followed by infusion of antigen specific CART enhanced the anti-tumor effect of CAR-T therapy 87.
6. Myeloma preclinical models and therapeutic targeting of MDSCs
6.1. Immunocompetent myeloma mouse models
Most preclinical studies of the development and function of immune suppressor cells in MM have been done using an immunocompetent mouse model, C57BL/KaLwRij, and two murine myeloma cell lines originated from the mice, 5T2MM and 5T33MM. These models resemble the human myeloma disease as they exhibit widespread systemic disease with bony lesions. 5T33MM cells develop a more aggressive myeloma that grows in 3 weeks, while 5T2MM cells grow in 12 weeks with a slower paced disease.
Valckenborgh and colleagues, using the immunocompetent mouse and murine myeloma cells, demonstrated that immunosuppressive MDSCs subsets (CD11bhighLy6Glow) are present in MM and their immunosuppressive capacity is induced by myeloma cells 99. In another study, the Dana-Farber group demonstrated a bidirectional interaction between myeloma cells and MDSCs, influencing the cell mediated immune response. They demonstrated that myeloma cells promote development of MDSCs from healthy donor peripheral blood mononuclear cells and conversely, that MDSCs favor myeloma growth by suppressing T-cell mediated immune responses 30. MDSC-mediated T-cell suppression is partially dependent on arginine and NO synthase. Interestingly, they showed that bortezomib and melphalan abrogate the effect of MDSC suppressor factors (IL-6, IL-10, ARG1, iNOS and ROS).
5.2. MDSC cellular interactions in the myeloma microenvironment
The S100A9 knock-out (KO) murine model lacks S100A9, a STAT3-inducible myeloid-related protein crucial in MDSC production and accumulation in a tumor-bearing mouse, and thus provides a good model to study the effect of MDSC depletion in MM progression 33. Ramachandran and colleagues showed that MDSCs play an essential role at early stages of MM as well as disease progression using this murine myeloma model 33. These experiments were done in mice on a mixed FVB/N × C57BL/6 background, which develop spontaneous MM and subsequent widespread myeloma measurable by serum electrophoresis. Th1 CD4+ cell activation and antigen-mediated T-cell expansion in the BM is accelerated in the S100A9KO model leading to slower myeloma progression in this model. These data highlight the important role of MDSC to create a quiescent immunological microenvironment for myeloma cells. Apart from immunosuppressive effects, MDSCs also exert a direct tumor-promoting effect on myeloma cells. AMP-activated protein kinase (AMPK), a key regulator of energy homeostasis in MM, increases after co-culture of MDSCs with myeloma cells with increased protection against apoptosis induced by bortezomib and melphalan 100. Furthermore, the intrinsic plasticity of MDSCs to differentiate to osteoclasts, an essential element of the myeloma niche, may contribute to the dynamics of the myeloma microenvironment, favoring disease progression101,102. It appears that the osteoclast differentiation of MDSCs is mediated through a variety of osteoclastogenic growth factors, including RANTES and MCP-1 secreted by tumor cells. Zoledronic acid, a potent bisphophonate, is able to reduce accumulation of MDSCs within the bone marrow and a concomitant decrease in osteoclastogenesis 101.
5.3. MDSC distribution through myeloma progression
Veirman et al. studied MDSC distribution throughout the myeloma progression course. They showed that BM MDSCs increase early in MM progression in the 5TMM mouse model, and that they appear in the peripheral blood at later stages. Also, the immunosuppressive capacity of MDSC subsets is increased early in the myelomagenesis process. Furthermore, co-culture with MDSCs promotes MM cell survival by upregulation of AMPK, the anti-apoptotic factors MCL-1 and BCL2, and contributes to resistance to bortezomib and melphalan 100. Also, they demonstrated that MDSC-suppressive therapies by anti-GR1 and 5-fluorouracil can augment the anti-myeloma effects of bortezomib.
MDSCs have been shown to exert T-cell suppression and to prolong survival of MM cells in vitro 103. Also, MDSC targeting by anti-GR1 antibody resulted in a significant anti-tumor effect in vivo. MCL-1 up-regulation showed a major survival mechanism for MM-induced MDSC. STAT3 inhibition abolishes the suppressive effect of MDSCs on T cells 104 and inhibition of MDSC STAT3 mediated by sunitinib, a multitargeted tyrosine kinase inhibitor, blocks MDSCs proliferation and restores T-cell function 105,106.
5.4. MDSC-targeted strategies
The increased understanding of the underlying mechanisms responsible for MDSC development, trafficking and function has contributed to the exploration of a variety of strategies that may effectively target the cancer immunosuppression network. For example, ATRA has been used to promote MDSC differentiation into more mature cells 107. STAT3 inhibitors lead to S100A9 protein depletion, which affects MDSC accumulation and their activation in cancer sites 29. There are several promising STAT3 inhibitor compounds which are in phase II and III clinical trials, mainly for malignancies other than MM, and some of their anti-tumor efficacy may be attributed to MDSC-suppressive effects 108. The combination of these compounds with standard anti-myeloma agents should be explored in early phase clinical trials. The downregulatory effect of phosphodiesterase inhibitors on arginase 1, nitric oxide synthase-2 expression and subsequent MDSC inaction and depletion 28 was translated into MDSC-directed clinical trials for myeloma patients; Gosh and colleges reported a combination therapy study using tadalafil, a phosphodiesterase 5 inhibitor, and lenalidomide 109. Thirteen lenalidomide-refractory patients were recruited; one patient achieved minimal response and 4 patients, stable disease. Although tolerable, this approach lacked efficacy. MDSCs were measured by flow cytometry using CD14+, CD33+, HLADRlow, IL4Rα+ or CD15+, CD33+, HLADRlow on pre and post-treatment samples. Importantly, MDSCs were not detected in the pre-treatment blood or bone marrow samples of these heavily treated patients. Therefore, it was speculated that the poor response from MDSC-suppressive therapy was due to the lack of sufficient target cells (MDSCs) in these patient populations.
Immunosuppressive effects of MDSCs can also be inhibited by the class of small molecules known as triterpenoids 110. In mouse models, RTA 408 depletes tumor nitrotyrosine burden and inhibits MDSC activity with subsequent enhancement of T-cell antitumor activity. This finding was utilized to augment the effect of checkpoint inhibitors in melanoma. Currently, the next generation of synthetic triterpenoids RTA 408 I are in a phase 1b/2 clinical trial in melanoma patients to test their ability to augment checkpoint inhibitors such as ipilimumab and PD1 inhibitors. A summary of the different MDSC-targeting agents based on the mechanism of action is presented in Table 1.
Table 1.
Potential therapeutic agents targeting MDSC classified according to mechanism.
Mechanism of Action | Compounds |
---|---|
Differentiation Promotion | Triterponoids111 |
HDAC inhibitor112 | |
All-trans-retinoic-acid (ATRA)113,114 | |
Vit D3115 | |
Maturation blockage | Sunitinib116 |
STAT3 inhibitors (anti-apoptotic genes suppression and reduced ROC)29 | |
MMP9 inhibitors117 | |
Bevacizumab118 | |
Anti-BV8 mAb119 | |
Axitinib120 | |
Amino-Bisphoonates (Zoldronate) 117 | |
MDSC Trafficking | Gemcitabine121 |
5-FU122 | |
Doxorubicin123 | |
CXCR2 and CXCR6 antagonist124 | |
Function | Phosphodiestrase-5 inhibitor125 |
Triterponoids111 | |
TGF-β inhibitors60 | |
ROS inhibitor (Nitraspirin and N-Acetyl Cysteine)126 | |
COX2 inhibitors (decrease in ARG and NOS expression)127 |
7. Summary and future directions
During the last decade, a growing effort has been devoted to understanding the role of immune regulatory cells in tumor progression. MDSCs appear to play a critical role in the generation of an immune dysfunctional microenvironment in patients with multiple myeloma. Identification of molecular pathways involved in MDSC evolution, circulation and function could potentially lead to new targets for therapeutics development. Novel agents targeting intermediates in these pathways will have the capacity to disrupt the myeloma immunosuppressive network, and will be incorporated in unique strategies that could improve the efficacy of current therapies as well as form the basis for cancer chemoprevention throughout the myelomagenesis process. Inhibition of the tumor-promoting and immune-suppressive functions of MDSCs in MM represents a needed and promising novel immune-based therapeutic approach in this disease.
Practice Points.
Myeloma-induced immunosuppression is result of a complex cross-talk between immunoeffector and immunosuppressive cells.
MDSCs are increased in myeloma patients in compare to normal individuals and their presence correlate to response to therapy.
There is bidirectional talk between myeloma cells and MDSC; Myeloma cells promote MDSC differentiation and function and MDSC contributes to chemotherapy resistance.
MDSCs play a central role to orchestrate the myeloma immunosuppression network by affecting NK, effector T cells, Tregs and dendritic cells.
Therapeutics to overcome MDSCs can augment the effect of upcoming immunotherapy for MM.
Research Agenda.
Investigate MDSC trafficking further to elucidate the underlying mechanisms of cell-cell interactions between MDSC and other component of immune system.
Study MDSC-targeting agents in conjunction with current and future anti-myeloma agents, specifically immunotherapy modalities.
Acknowledgments
This work has been supported by National Cancer Institute training grant K12 CA076917, Clinical Oncology Research Career Development Program (CORP).
Footnotes
Conflict of interest
The authors declare no commercial or financial relationships that could be construed as a potential conflict of interest.
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
References
- 1.Niesvizky R. Immunomodulatory agents changing the landscape of multiple myeloma treatment. Crit Rev Oncol Hematol. 2013;88:S1–S4. doi: 10.1016/j.critrevonc.2012.12.011. [DOI] [PubMed] [Google Scholar]
- 2.Bird J, Russell N, Samson D. Minimal residual disease after bone marrow transplantation for multiple myeloma: evidence for cure in long-term survivors. Bone Marrow Transplant. 1993;12:651–654. [PubMed] [Google Scholar]
- 3.Mahindra A, Laubach J, Raje N, Munshi N, Richardson PG, Anderson K. Latest advances and current challenges in the treatment of multiple myeloma. Nat Rev Clin Oncol. 2012;9:135–143. doi: 10.1038/nrclinonc.2012.15. [DOI] [PubMed] [Google Scholar]
- 4.Rutella S, Locatelli F. Targeting multiple-myeloma-induced immune dysfunction to improve immunotherapy outcomes. Clin Dev Immunol. 2012;2012:196063. doi: 10.1155/2012/196063. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Pratt G, Goodyear O, Moss P. Immunodeficiency and immunotherapy in multiple myeloma. Br J Haematol. 2007;138:563–579. doi: 10.1111/j.1365-2141.2007.06705.x. [DOI] [PubMed] [Google Scholar]
- 6.Nucci M, Anaissie E. Infections in patients with multiple myeloma in the era of high-dose therapy and novel agents. Clin Infect Dis. 2009;49:1211–1225. doi: 10.1086/605664. [DOI] [PubMed] [Google Scholar]
- 7.Nencioni A, Schwarzenberg K, Brauer KM, Schmidt SM, Ballestrero A, Grünebach F, et al. Proteasome inhibitor bortezomib modulates TLR4-induced dendritic cell activation. Blood. 2006;108:551–558. doi: 10.1182/blood-2005-08-3494. [DOI] [PubMed] [Google Scholar]
- 8.Spisek R, Charalambous A, Mazumder A, Vesole DH, Jagannath S, Dhodapkar MV. Bortezomib enhances dendritic cell (DC)–mediated induction of immunity to human myeloma via exposure of cell surface heat shock protein 90 on dying tumor cells: therapeutic implications. Blood. 2007;109:4839–4845. doi: 10.1182/blood-2006-10-054221. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Feng X, Yan J, Wang Y, Zierath JR, Nordenskjöld M, Henter JI, et al. The proteasome inhibitor bortezomib disrupts tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) expression and natural killer (NK) cell killing of TRAIL receptor-positive multiple myeloma cells. Mol Immunol. 2010;47:2388–2396. doi: 10.1016/j.molimm.2010.05.003. [DOI] [PubMed] [Google Scholar]
- 10.Shi J, Tricot GJ, Garg TK, Malaviarachchi PA, Szmania SM, Kellum RE, et al. Bortezomib down-regulates the cell-surface expression of HLA class I and enhances natural killer cell–mediated lysis of myeloma. Blood. 2008;111:1309–1317. doi: 10.1182/blood-2007-03-078535. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Hallett WH, Jing W, Drobyski WR, Johnson BD. Immunosuppressive effects of multiple myeloma are overcome by PD-L1 blockade. Biol Blood Marrow Transplant. 2011;17:1133–1145. doi: 10.1016/j.bbmt.2011.03.011. [DOI] [PubMed] [Google Scholar]
- 12.Rossi M, Botta C, Correale P, Tassone P, Tagliaferri P. Immunologic microenvironment and personalized treatment in multiple myeloma. Expert Opin Biol Ther. 2013;13:S83–S93. doi: 10.1517/14712598.2013.799130. [DOI] [PubMed] [Google Scholar]
- 13.Benson DM, Jr, Bakan CE, Mishra A, Hofmeister CC, Efebera Y, Becknell B, et al. The PD-1/PD-L1 axis modulates the natural killer cell versus multiple myeloma effect: a therapeutic target for CT-011, a novel monoclonal anti–PD-1 antibody. Blood. 2010;116:2286–2294. doi: 10.1182/blood-2010-02-271874. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Amin S, Yip W, Minvielle S, Broyl A, Li Y, Hanlon B, et al. Gene expression profile alone is inadequate in predicting complete response in multiple myeloma. Leukemia. 2014;28:2229–2234. doi: 10.1038/leu.2014.140. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Brousseau M, Leleu X, Gerard J, Gastinne T, Godon A, Genevieve F, et al. Hyperdiploidy is a common finding in monoclonal gammopathy of undetermined significance and monosomy 13 is restricted to these hyperdiploid patients. Clin Cancer Res. 2007;13:6026–6031. doi: 10.1158/1078-0432.CCR-07-0031. [DOI] [PubMed] [Google Scholar]
- 16.Chiecchio L, Dagrada GP, Ibrahim AH, Cabanas ED, Protheroe RK, Stockley DM, et al. Timing of acquisition of deletion 13 in plasma cell dyscrasias is dependent on genetic context. Haematologica. 2009;94:1708–1713. doi: 10.3324/haematol.2009.011064. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Bianchi G, Munshi NC. Pathogenesis beyond the cancer clone (s) in multiple myeloma. Blood. 2015;125:3049–3058. doi: 10.1182/blood-2014-11-568881. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Steiner N, Kern J, Untergasser G. Angiogenesis inhibition, hypoxia, and targeting the bone marrow microenvironment in multiple myeloma: new strategies and targets. Memo. 2014;7:202–205. [Google Scholar]
- 19.Manier S, Sacco A, Leleu X, Ghobrial IM, Roccaro AM. Bone marrow microenvironment in multiple myeloma progression. J Biomed Biotechnol. 2012;2012:157496. doi: 10.1155/2012/157496. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Kawano Y, Moschetta M, Manier S, Glavey S, Görgün GT, Roccaro AM, et al. Targeting the bone marrow microenvironment in multiple myeloma. Immunol Rev. 2015;263:160–172. doi: 10.1111/imr.12233. [DOI] [PubMed] [Google Scholar]
- 21.Andrews S, Kabrah S, May J, Donaldson C, Morse H. Multiple myeloma: the bone marrow microenvironment and its relation to treatment. Br J Biomed Sci. 2013;70:110–120. doi: 10.1080/09674845.2013.11669945. [DOI] [PubMed] [Google Scholar]
- 22.Goh C, Narayanan S, Hahn YS. Myeloid-derived suppressor cells: the dark knight or the joker in viral infections? Immunol Rev. 2013;255:210–221. doi: 10.1111/imr.12084. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Khaled YS, Ammori BJ, Elkord E. Myeloid-derived suppressor cells in cancer: recent progress and prospects. Immunol Cell Biol. 2013;91:493–502. doi: 10.1038/icb.2013.29. [DOI] [PubMed] [Google Scholar]
- 24.Rodríguez PC, Ochoa AC. Arginine regulation by myeloid derived suppressor cells and tolerance in cancer: mechanisms and therapeutic perspectives. Immunol Rev. 2008;222:180–191. doi: 10.1111/j.1600-065X.2008.00608.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Ostrand-Rosenberg S. Myeloid-derived suppressor cells: more mechanisms for inhibiting antitumor immunity. Cancer Immunol Immunother. 2010;59:1593–1600. doi: 10.1007/s00262-010-0855-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Srivastava MK, Zhu L, Harris-White M, Kar U, Huang M, Johnson MF, et al. Myeloid suppressor cell depletion augments antitumor activity in lung cancer. PLoS One. 2012;7:e40677. doi: 10.1371/journal.pone.0040677. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Mougiakakos D, Jitschin R, von Bahr L, Poschke I, Gary R, Sundberg B, et al. Immunosuppressive CD14+HLA-DRlow/neg IDO+ myeloid cells in patients following allogeneic hematopoietic stem cell transplantation. Leukemia. 2013;27:377–388. doi: 10.1038/leu.2012.215. [DOI] [PubMed] [Google Scholar]
- 28.Serafini P, Borrello I, Bronte V. Myeloid suppressor cells in cancer: Recruitment, phenotype, properties, and mechanisms of immune suppression. Semin Cancer Biol. 2006;16:53–65. doi: 10.1016/j.semcancer.2005.07.005. [DOI] [PubMed] [Google Scholar]
- 29.Kujawski M, Kortylewski M, Lee H, Herrmann A, Kay H, Yu H. Stat3 mediates myeloid cell–dependent tumor angiogenesis in mice. The Journal of clinical investigation. 2008;118:3367. doi: 10.1172/JCI35213. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Görgün GT, Whitehill G, Anderson JL, Hideshima T, Maguire C, Laubach J, et al. Tumor-promoting immune-suppressive myeloid-derived suppressor cells in the multiple myeloma microenvironment in humans. Blood. 2013;121:2975–2987. doi: 10.1182/blood-2012-08-448548. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Diaz-Montero CM, Finke J, Montero AJ. Myeloid-derived suppressor cells in cancer: therapeutic, predictive, and prognostic implications. Semin Oncol. 2014;41:174–84. doi: 10.1053/j.seminoncol.2014.02.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Peranzoni E, Zilio S, Marigo I, Dolcetti L, Zanovello P, Mandruzzato S, et al. Myeloid-derived suppressor cell heterogeneity and subset definition. Curr Opin Immunol. 2010;22:238–244. doi: 10.1016/j.coi.2010.01.021. [DOI] [PubMed] [Google Scholar]
- 33.Ramachandran IR, Martner A, Pisklakova A, Condamine T, Chase T, Vogl T, et al. ZoledronicMyeloid-derived suppressor cells regulate growth of multiple myeloma by inhibiting T cells in bone marrow. J Immunol. 2013;190:3815–3823. doi: 10.4049/jimmunol.1203373. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Diaz-Montero CM, Salem ML, Nishimura MI, Garrett-Mayer E, Cole DJ, Montero AJ. Increased circulating myeloid-derived suppressor cells correlate with clinical cancer stage, metastatic tumor burden, and doxorubicin–cyclophosphamide chemotherapy. Cancer Immunol Immunother. 2009;58:49–59. doi: 10.1007/s00262-008-0523-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Christiansson L, Söderlund S, Svensson E, Mustjoki S, Bengtsson M, Simonsson B, et al. Increased level of myeloid-derived suppressor cells, programmed death receptor ligand 1/programmed death receptor 1, and soluble CD25 in Sokal high risk chronic myeloid leukemia. PLoS One. 2013;8:e55818. doi: 10.1371/journal.pone.0055818. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Favaloro J, Liyadipitiya T, Brown R, Yang S, Suen H, Woodland N, et al. Myeloid derived suppressor cells are numerically, functionally and phenotypically different in patients with multiple myeloma. Leuk Lymphoma. 2014;55:2893–2900. doi: 10.3109/10428194.2014.904511. [DOI] [PubMed] [Google Scholar]
- 37.Brimnes MK, Vangsted AJ, Knudsen L, Gimsing P, Gang A, Johnsen HE, et al. Increased Level of both CD4+ FOXP3+ Regulatory T Cells and CD14+ HLA-DR−/low Myeloid-Derived Suppressor Cells and Decreased Level of Dendritic Cells in Patients with Multiple Myeloma. Scand J Immunol. 2010;72:540–547. doi: 10.1111/j.1365-3083.2010.02463.x. [DOI] [PubMed] [Google Scholar]
- 38.Wang Z, Zhang L, Wang H, Xiong S, Li Y, Tao Q, et al. Tumor-induced CD14+ HLA-DR−/low myeloid-derived suppressor cells correlate with tumor progression and outcome of therapy in multiple myeloma patients. Cancer Immunol Immunother. 2015;64:389–399. doi: 10.1007/s00262-014-1646-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.de Magalhães RJP, Vidriales MB, Paiva B, Fernandez-Gimenez C, García-Sanz R, Mateos MV, et al. Analysis of the immune system of multiple myeloma patients achieving long-term disease control by multidimensional flow cytometry. Haematologica. 2013;98:79–86. doi: 10.3324/haematol.2012.067272. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Bryant C, Suen H, Brown R, Yang S, Favaloro J, Aklilu E, et al. Long-term survival in multiple myeloma is associated with a distinct immunological profile, which includes proliferative cytotoxic T-cell clones and a favourable Treg/Th17 balance. Blood Cancer J. 2013;3:e148. doi: 10.1038/bcj.2013.34. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Tebartz C, Horst SA, Sparwasser T, Huehn J, Beineke A, Peters G, et al. A Major Role for Myeloid-Derived Suppressor Cells and a Minor Role for Regulatory T Cells in Immunosuppression during Staphylococcus aureus Infection. J Immunol. 2015;194:1100–1111. doi: 10.4049/jimmunol.1400196. [DOI] [PubMed] [Google Scholar]
- 42.Sinha P, Carter D, Ostrand-Rosenberg S. Myeloid-derived suppressor cells contribute to maintaining allogeneic pregnancies (IRC4P.461) J Immunol. 2015;194(1 Supplement):57.14. [Google Scholar]
- 43.Vollbrecht T, Stirner R, Tufman A, Roider J, Huber RM, Bogner JR, et al. Chronic progressive HIV-1 infection is associated with elevated levels of myeloid-derived suppressor cells. AIDS. 2012;26:F31–F37. doi: 10.1097/QAD.0b013e328354b43f. [DOI] [PubMed] [Google Scholar]
- 44.Giles A, Vicioso Y, Persenaire C, Kasai M, Highfill S, Mendoza A, et al. Abstract LB-284: Bone marrow-derived progenitor cells develop into myeloid-derived suppressor cells at metastatic sites. Cancer Res. 2013;73:LB-284–LB-284. [Google Scholar]
- 45.Mahnke K, Ring S, Johnson TS, Schallenberg S, Schönfeld K, Storn V, et al. Induction of immunosuppressive functions of dendritic cells in vivo by CD4+ CD25+ regulatory T cells: Role of B7-H3 expression and antigen presentation. Eur J Immunol. 2007;37:2117–2126. doi: 10.1002/eji.200636841. [DOI] [PubMed] [Google Scholar]
- 46.Nair JR, Rozanski CH, Lee KP. Under one roof: The bone marrow survival niche for multiple myeloma and normal plasma cells. Oncoimmunology. 2012;1:388–389. doi: 10.4161/onci.18746. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Condamine T, Gabrilovich DI. Molecular mechanisms regulating myeloid-derived suppressor cell differentiation and function. Trends Immunol. 2011;32:19–25. doi: 10.1016/j.it.2010.10.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Rodriguez PC, Quiceno DG, Zabaleta J, Ortiz B, Zea AH, Piazuelo MB, et al. Arginase I production in the tumor microenvironment by mature myeloid cells inhibits T-cell receptor expression and antigen-specific T-cell responses. Cancer Res. 2004;64:5839–5849. doi: 10.1158/0008-5472.CAN-04-0465. [DOI] [PubMed] [Google Scholar]
- 49.Rodriguez PC, Quiceno DG, Ochoa AC. L-arginine availability regulates T-lymphocyte cell-cycle progression. Blood. 2007;109:1568–1573. doi: 10.1182/blood-2006-06-031856. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Mannick JB, Hausladen A, Liu L, Hess DT, Zeng M, Miao QX, et al. Fas-induced caspase denitrosylation. Science. 1999;284:651–654. doi: 10.1126/science.284.5414.651. [DOI] [PubMed] [Google Scholar]
- 51.Bronte V, Serafini P, De Santo C, Marigo I, Tosello V, Mazzoni A, et al. IL-4-induced arginase 1 suppresses alloreactive T cells in tumor-bearing mice. J Immunol. 2003;170:270–278. doi: 10.4049/jimmunol.170.1.270. [DOI] [PubMed] [Google Scholar]
- 52.Bingisser RM, Tilbrook PA, Holt PG, Kees UR. Macrophage-derived nitric oxide regulates T cell activation via reversible disruption of the Jak3/STAT5 signaling pathway. J Immunol. 1998;160:5729–5734. [PubMed] [Google Scholar]
- 53.Rodriguez PC, Hernandez CP, Quiceno D, Dubinett SM, Zabaleta J, Ochoa JB, et al. Arginase I in myeloid suppressor cells is induced by COX-2 in lung carcinoma. J Exp Med. 2005;202:931–939. doi: 10.1084/jem.20050715. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Huang B, Pan PY, Li Q, Sato AI, Levy DE, Bromberg J, et al. Gr-1+ CD115+ immature myeloid suppressor cells mediate the development of tumor-induced T regulatory cells and T-cell anergy in tumor-bearing host. Cancer Res. 2006;66:1123–1131. doi: 10.1158/0008-5472.CAN-05-1299. [DOI] [PubMed] [Google Scholar]
- 55.Hoechst B, Ormandy LA, Ballmaier M, Lehner F, Krüger C, Manns MP, et al. A new population of myeloid-derived suppressor cells in hepatocellular carcinoma patients induces CD4+ CD25+ Foxp3+ T cells. Gastroenterology. 2008;135:234–243. doi: 10.1053/j.gastro.2008.03.020. [DOI] [PubMed] [Google Scholar]
- 56.Serafini P, Mgebroff S, Noonan K, Borrello I. Myeloid-derived suppressor cells promote cross-tolerance in B-cell lymphoma by expanding regulatory T cells. Cancer Res. 2008;68:5439–5449. doi: 10.1158/0008-5472.CAN-07-6621. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Youn JI, Gabrilovich DI. The biology of myeloid-derived suppressor cells: The blessing and the curse of morphological and functional heterogeneity. Eur J Immunol. 2010;40:2969–2975. doi: 10.1002/eji.201040895. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Li H, Han Y, Guo Q, Zhang M, Cao X. Cancer-expanded myeloid-derived suppressor cells induce anergy of NK cells through membrane-bound TGF-β1. J Immunol. 2009;182:240–249. doi: 10.4049/jimmunol.182.1.240. [DOI] [PubMed] [Google Scholar]
- 59.Mao Y, Sarhan D, Steven A, Seliger B, Kiessling R, Lundqvist A. Inhibition of tumor-derived prostaglandin-E2 blocks the induction of myeloid-derived suppressor cells and recovers natural killer cell activity. Clin Cancer Res. 2014;20:4096–4106. doi: 10.1158/1078-0432.CCR-14-0635. [DOI] [PubMed] [Google Scholar]
- 60.Llopiz D, Dotor J, Casares N, Bezunartea J, Díaz-Valdés N, Ruiz M, et al. Peptide inhibitors of transforming growth factor-β enhance the efficacy of antitumor immunotherapy. Int J Cancer. 2009;125:2614–2623. doi: 10.1002/ijc.24656. [DOI] [PubMed] [Google Scholar]
- 61.Yang L, Huang J, Ren X, Gorska AE, Chytil A, Aakre M, et al. Abrogation of TGFβ signaling in mammary carcinomas recruits Gr-1+ CD11b+ myeloid cells that promote metastasis. Cancer Cell. 2008;13:23–35. doi: 10.1016/j.ccr.2007.12.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Xiang X, Poliakov A, Liu C, Liu Y, Deng Zb, Wang J, et al. Induction of myeloid-derived suppressor cells by tumor exosomes. Int J Cancer. 2009;124:2621–2633. doi: 10.1002/ijc.24249. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.García-Sanz R, Gonzalez M, Orfao A, Moro M, Hernandez J, Borrego D, et al. Analysis of natural killer-associated antigens in peripheral blood and bone marrow of multiple myeloma patients and prognostic implications. Br J Haematol. 1996;93:81–88. doi: 10.1046/j.1365-2141.1996.4651006.x. [DOI] [PubMed] [Google Scholar]
- 64.King MA, Radicchi-Mastroianni MA. Natural killer cells and CD56+ T cells in the blood of multiple myeloma patients: Analysis by 4-colour flow cytometry. Cytometry. 1996;26:121–124. doi: 10.1002/(SICI)1097-0320(19960615)26:2<121::AID-CYTO4>3.0.CO;2-J. [DOI] [PubMed] [Google Scholar]
- 65.Jurisic V, Srdic T, Konjevic G, Markovic O, Colovic M. Clinical stage-depending decrease of NK cell activity in multiple myeloma patients. Med Oncol. 2007;24:312–317. doi: 10.1007/s12032-007-0007-y. [DOI] [PubMed] [Google Scholar]
- 66.Prabhala RH, Pelluru D, Fulciniti M, Prabhala HK, Nanjappa P, Song W, et al. Elevated IL-17 produced by TH17 cells promotes myeloma cell growth and inhibits immune function in multiple myeloma. Blood. 2010;115:5385–5392. doi: 10.1182/blood-2009-10-246660. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Noonan K, Marchionni L, Anderson J, Pardoll D, Roodman GD, Borrello I. A novel role of IL-17–producing lymphocytes in mediating lytic bone disease in multiple myeloma. Blood. 2010;116:3554–3563. doi: 10.1182/blood-2010-05-283895. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Ortiz ML, Kumar V, Martner A, Mony S, Donthireddy L, Condamine T, et al. Immature myeloid cells directly contribute to skin tumor development by recruiting IL-17–producing CD4+ T cells. J Exp Med. 2015;212:351–367. doi: 10.1084/jem.20140835. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.He D, Li H, Yusuf N, Elmets CA, Li J, Mountz JD, et al. IL-17 promotes tumor development through the induction of tumor promoting microenvironments at tumor sites and myeloid-derived suppressor cells. J Immunol. 2010;184:2281–2288. doi: 10.4049/jimmunol.0902574. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Bruchard M, Mignot G, Derangère V, Chalmin F, Chevriaux A, Végran F, et al. Chemotherapy-triggered cathepsin B release in myeloid-derived suppressor cells activates the Nlrp3 inflammasome and promotes tumor growth. Nat Med. 2013;19:57–64. doi: 10.1038/nm.2999. [DOI] [PubMed] [Google Scholar]
- 71.Condamine T, Mastio J, Gabrilovich DI. Transcriptional regulation of myeloid-derived suppressor cells. J Leukoc Biol. 2015;98:913–22. doi: 10.1189/jlb.4RI0515-204R. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Ding ZC, Lu X, Yu M, Lemos H, Huang L, Chandler P, et al. Immunosuppressive myeloid cells induced by chemotherapy attenuate antitumor CD4+ T-cell responses through the PD-1–PD-L1 axis. Cancer Res. 2014;74:3441–3453. doi: 10.1158/0008-5472.CAN-13-3596. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Busch A, Zeh D, Janzen V, Mugge LO, Wolf D, Fingerhut L, et al. Treatment with lenalidomide induces immunoactivating and counter-regulatory immunosuppressive changes in myeloma patients. Clin Exp Immunol. 2014;177:439–453. doi: 10.1111/cei.12343. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Minnema M, Van der Veer M, Aarts T, Emmelot M, Mutis T, Lokhorst H. Lenalidomide alone or in combination with dexamethasone is highly effective in patients with relapsed multiple myeloma following allogeneic stem cell transplantation and increases the frequency of CD4+ Foxp3+ T cells. Leukemia. 2009;23:605. doi: 10.1038/leu.2008.247. [DOI] [PubMed] [Google Scholar]
- 75.Neuber B, Herth I, Tolliver C, Schoenland S, Hegenbart U, Hose D, et al. Lenalidomide enhances antigen-specific activity and decreases CD45RA expression of T cells from patients with multiple myeloma. J Immunol. 2011;187:1047–1056. doi: 10.4049/jimmunol.1002460. [DOI] [PubMed] [Google Scholar]
- 76.Wu L, Adams M, Carter T, Chen R, Muller G, Stirling D, et al. Lenalidomide enhances natural killer cell and monocyte-mediated antibody-dependent cellular cytotoxicity of rituximab-treated CD20+ tumor cells. Clin Cancer Res. 2008;14:4650–4657. doi: 10.1158/1078-0432.CCR-07-4405. [DOI] [PubMed] [Google Scholar]
- 77.Jungkunz-Stier I, Zekl M, Stühmer T, Einsele H, Seggewiss-Bernhardt R. Modulation of natural killer cell effector functions through lenalidomide/dasatinib and their combined effects against multiple myeloma cells. Leuk Lymphoma. 2014;55:168–176. doi: 10.3109/10428194.2013.794270. [DOI] [PubMed] [Google Scholar]
- 78.Lagrue K, Carisey A, Morgan DJ, Chopra R, Davis DM. Lenalidomide augments actin remodelling and lowers NK cell activation thresholds. Blood. 2015;126:50–60. doi: 10.1182/blood-2015-01-625004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Ochando J, Conde P, Bronte V. Monocyte-Derived Suppressor Cells in Transplantation. Curr Transplant Rep. 2015;2:176–183. doi: 10.1007/s40472-015-0054-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Ochando J, Conde P. Editorial: Dexamethasone and MDSC in transplantation: yes to NO. J Leukoc Biol. 2014;96:669–671. doi: 10.1189/jlb.3CE0514-272R. [DOI] [PubMed] [Google Scholar]
- 81.Varga G, Ehrchen J, Tsianakas A, Tenbrock K, Rattenholl A, Seeliger S, et al. Glucocorticoids induce an activated, anti-inflammatory monocyte subset in mice that resembles myeloid-derived suppressor cells. J Leukoc Biol. 2008;84:644–650. doi: 10.1189/jlb.1107768. [DOI] [PubMed] [Google Scholar]
- 82.Liao J, Wang X, Bi Y, Shen B, Shao K, Yang H, et al. Dexamethasone potentiates myeloid-derived suppressor cell function in prolonging allograft survival through nitric oxide. J Leukoc Biol. 2014;96:675–684. doi: 10.1189/jlb.2HI1113-611RR. [DOI] [PubMed] [Google Scholar]
- 83.Poschke I, Mao Y, Adamson L, Salazar-Onfray F, Masucci G, Kiessling R. Myeloid-derived suppressor cells impair the quality of dendritic cell vaccines. Cancer Immunol Immunother. 2012;61:827–838. doi: 10.1007/s00262-011-1143-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Laborde RR, Lin Y, Gustafson MP, Bulur PA, Dietz AB. Cancer vaccines in the world of immune suppressive monocytes (CD14 (+) HLA-DR (lo/neg) Cells): the gateway to improved responses. Front Immunol. 2014;5:3389. doi: 10.3389/fimmu.2014.00147. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Castella B, Foglietta M, Sciancalepore P, Rigoni M, Coscia M, Griggio V, et al. Anergic bone marrow Vγ9Vδ2 T cells as early and long-lasting markers of PD-1-targetable microenvironment-induced immune suppression in human myeloma. OncoImmunology. 2015;4:e1047580. doi: 10.1080/2162402X.2015.1047580. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Gorgun G, Samur MK, Cowens KB, Paula S, Bianchi G, Anderson JE, et al. Lenalidomide Enhances Immune Checkpoint Blockade Induced Immune Response in Multiple Myeloma. Clin Cancer Res. 2015;21:4607–18. doi: 10.1158/1078-0432.CCR-15-0200. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Highfill S, Smith J, Long A, Cui Y, Orentas R, Mackall C. Neutralization of murine myeloid suppressor cells enhances the efficacy of GD2-specific CAR T cells directed against human sarcoma in a xenograft model (VAC11P. 1010) J Immunol. 2014;192(1 Supplement):205.11. [Google Scholar]
- 88.Luyckx A, Schouppe E, Rutgeerts O, Lenaerts C, Fevery S, Devos T, et al. G-CSF stem cell mobilization in human donors induces polymorphonuclear and mononuclear myeloid-derived suppressor cells. Clin Immunol. 2012;143:83–87. doi: 10.1016/j.clim.2012.01.011. [DOI] [PubMed] [Google Scholar]
- 89.D’Aveni M, Rossignol J, Coman T, Sivakumaran S, Henderson S, Manzo T, et al. G-CSF mobilizes CD34+ regulatory monocytes that inhibit graft-versus-host disease. Sci Transl Med. 2015;7:281ra42. doi: 10.1126/scitranslmed.3010435. [DOI] [PubMed] [Google Scholar]
- 90.Messmann JJ, Reisser T, Leithäuser F, Lutz MB, Debatin KM, Strauss G. In vitro-generated MDSCs prevent murine GVHD by inducing type 2 T cells without disabling antitumor cytotoxicity. Blood. 2015;126:1138–1148. doi: 10.1182/blood-2015-01-624163. [DOI] [PubMed] [Google Scholar]
- 91.Franssen LE, van de Donk NW, Emmelot ME, Roeven MW, Schaap N, Dolstra H, et al. The impact of circulating suppressor cells in multiple myeloma patients on clinical outcome of DLIs. Bone Marrow Transplant. 2015;50:822–828. doi: 10.1038/bmt.2015.48. [DOI] [PubMed] [Google Scholar]
- 92.Guan Q, Blankstein AR, Anjos K, Synova O, Tulloch M, Giftakis A, et al. Functional Myeloid-Derived Suppressor Cell Subsets Recover Rapidly after Allogeneic Hematopoietic Stem/Progenitor Cell Transplantation. Biol Blood Marrow Transplant. 2015;21:1205–1214. doi: 10.1016/j.bbmt.2015.04.015. [DOI] [PubMed] [Google Scholar]
- 93.Zarkos K, Favaloro J, Liyadipitiya T, Brown R, Yang S, Suen H, et al. Myeloid derived suppressor cells are expanded in patients with multiple myeloma, induce Treg cells and delay T-cell recovery post transplantation. Cytotherapy. 2014;16:S65. [Google Scholar]
- 94.Kalos M, Levine BL, Porter DL, Katz S, Grupp SA, Bagg A, et al. T cells with chimeric antigen receptors have potent antitumor effects and can establish memory in patients with advanced leukemia. Sci Transl Med. 2011;3:95ra73. doi: 10.1126/scitranslmed.3002842. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Grupp SA, Kalos M, Barrett D, Aplenc R, Porter DL, Rheingold SR, et al. Chimeric antigen receptor–modified T cells for acute lymphoid leukemia. N Engl J Med. 2013;368:1509–1518. doi: 10.1056/NEJMoa1215134. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Garfall AL, Fraietta JA, Maus MV. Immunotherapy with chimeric antigen receptors for multiple myeloma. Discov Med. 2014;17:37–46. [PubMed] [Google Scholar]
- 97.Maus MV, June CH. Zoom zoom: racing CARs for multiple myeloma. Clin Cancer Res. 2013;19:1917–1919. doi: 10.1158/1078-0432.CCR-13-0168. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Maus MV, Grupp SA, Porter DL, June CH. Antibody-modified T cells: CARs take the front seat for hematologic malignancies. Blood. 2014;123:2625–2635. doi: 10.1182/blood-2013-11-492231. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Van Valckenborgh E, Schouppe E, Movahedi K, De Bruyne E, Menu E, De Baetselier P, et al. Multiple myeloma induces the immunosuppressive capacity of distinct myeloid-derived suppressor cell subpopulations in the bone marrow. Leukemia. 2012;26:2424–2428. doi: 10.1038/leu.2012.113. [DOI] [PubMed] [Google Scholar]
- 100.De Veirman K, Van Ginderachter JA, Van Riet I, De Beule N, Lub S, Menu E, et al. Myeloid Derived Suppressor Cell Mediated AMPK Activation Regulates Multiple Myeloma Cell Survival. Blood. 2014;124:2009–2009. [Google Scholar]
- 101.Sawant A, Deshane J, Jules J, Lee CM, Harris BA, Feng X, et al. Myeloid-derived suppressor cells function as novel osteoclast progenitors enhancing bone loss in breast cancer. Cancer Res. 2013;73:672–682. doi: 10.1158/0008-5472.CAN-12-2202. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Zhuang J, Zhang J, Lwin ST, Edwards JR, Edwards CM, Mundy GR, et al. Osteoclasts in multiple myeloma are derived from Gr-1+ CD11b+ myeloid-derived suppressor cells. PLoS One. 2012;7:e48871. doi: 10.1371/journal.pone.0048871. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.De Veirman K, Van Ginderachter J, Lub S, De Beule N, Thielemans K, Bautmans I, et al. Multiple myeloma induces Mcl-1 expression and survival of myeloid-derived suppressor cells. Oncotarget. 2015;6:10532–10547. doi: 10.18632/oncotarget.3300. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Wu L, Du H, Li Y, Qu P, Yan C. Signal transducer and activator of transcription 3 (Stat3C) promotes myeloid-derived suppressor cell expansion and immune suppression during lung tumorigenesis. Am J Pathol. 2011;179:2131–2141. doi: 10.1016/j.ajpath.2011.06.028. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Xin H, Zhang C, Herrmann A, Du Y, Figlin R, Yu H. Sunitinib inhibition of Stat3 induces renal cell carcinoma tumor cell apoptosis and reduces immunosuppressive cells. Cancer Res. 2009;69:2506–2513. doi: 10.1158/0008-5472.CAN-08-4323. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Ko JS, Rayman P, Ireland J, Swaidani S, Li G, Bunting KD, et al. Direct and differential suppression of myeloid-derived suppressor cell subsets by sunitinib is compartmentally constrained. Cancer Res. 2010;70:3526–3536. doi: 10.1158/0008-5472.CAN-09-3278. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Nefedova Y, Fishman M, Sherman S, Wang X, Beg AA, Gabrilovich DI. Mechanism of all-trans retinoic acid effect on tumor-associated myeloid-derived suppressor cells. Cancer Res. 2007;67:11021–11028. doi: 10.1158/0008-5472.CAN-07-2593. [DOI] [PubMed] [Google Scholar]
- 108.Shah MA, Muro K, Shitara K, Tebbutt NC, Bang Y-J, Lordick F, et al. 51st ASCO Annual Meeting Proceedings; 2015. abstr. TPS4139. [Google Scholar]
- 109.Ghosh N, Rudraraju L, Ye X, Noonan K, Huff CA, Borrello IM. Administration of an oral PDE5 inhibitor, tadalafil In conjunction with a lenalidomide containing regimen in patients with multiple myeloma. Blood. 2013;122:1959–1959. [Google Scholar]
- 110.Choi SH, Kim BG, Robinson J, Letterio JJ. The protective effects of the synthetic triterpenoid CDDO-methyl ester on colitis associated colon cancer. Cancer Res. 2011;71:827–827. [Google Scholar]
- 111.Nagaraj S, Youn JI, Weber H, Iclozan C, Lu L, Cotter MJ, et al. Anti-inflammatory triterpenoid blocks immune suppressive function of MDSCs and improves immune response in cancer. Clin Cancer Res. 2010;16:1812–1823. doi: 10.1158/1078-0432.CCR-09-3272. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Kroesen M, Gielen P, Brok IC, Armandari I, Hoogerbrugge PM, Adema GJ. HDAC inhibitors and immunotherapy; a double edged sword? Oncotarget. 2014;5:6558. doi: 10.18632/oncotarget.2289. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Gabrilovich DI, Velders MP, Sotomayor EM, Kast WM. Mechanism of immune dysfunction in cancer mediated by immature Gr-1+ myeloid cells. J Immunol. 2001;166:5398–5406. doi: 10.4049/jimmunol.166.9.5398. [DOI] [PubMed] [Google Scholar]
- 114.Mirza N, Fishman M, Fricke I, Dunn M, Neuger AM, Frost TJ, et al. All-trans-retinoic acid improves differentiation of myeloid cells and immune response in cancer patients. Cancer Res. 2006;66:9299–9307. doi: 10.1158/0008-5472.CAN-06-1690. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Lathers DM, Clark JI, Achille NJ, Young MRI. Phase 1B study to improve immune responses in head and neck cancer patients using escalating doses of 25-hydroxyvitamin D3. Cancer Immunol Immunother. 2004;53:422–430. doi: 10.1007/s00262-003-0459-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Finke JH, Rini B, Ireland J, Rayman P, Richmond A, Golshayan A, et al. Sunitinib reverses type-1 immune suppression and decreases T-regulatory cells in renal cell carcinoma patients. Clin Cancer Res. 2008;14:6674–6682. doi: 10.1158/1078-0432.CCR-07-5212. [DOI] [PubMed] [Google Scholar]
- 117.Melani C, Sangaletti S, Barazzetta FM, Werb Z, Colombo MP. Amino-biphosphonate–mediated MMP-9 inhibition breaks the tumor-bone marrow axis responsible for myeloid-derived suppressor cell expansion and macrophage infiltration in tumor stroma. Cancer Res. 2007;67:11438–11446. doi: 10.1158/0008-5472.CAN-07-1882. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Elamin YY, Rafee S, Toomey S, Hennessy BT. Immune effects of bevacizumab: killing two birds with one stone. Cancer Microenviron. 2015;8:15–21. doi: 10.1007/s12307-014-0160-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Hasnis E, Alishekevitz D, Gingis-Veltski S, Bril R, Fremder E, Voloshin T, et al. Anti-Bv8 antibody and metronomic gemcitabine improve pancreatic adenocarcinoma treatment outcome following weekly gemcitabine therapy. Neoplasia. 2014;16:501–510. doi: 10.1016/j.neo.2014.05.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Bose A, Taylor JL, Alber S, Watkins SC, Garcia JA, Rini BI, et al. Sunitinib facilitates the activation and recruitment of therapeutic anti-tumor immunity in concert with specific vaccination. Int J Cancer. 2011;129:2158–2170. doi: 10.1002/ijc.25863. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Suzuki E, Kapoor V, Jassar AS, Kaiser LR, Albelda SM. Gemcitabine selectively eliminates splenic Gr-1+/CD11b+ myeloid suppressor cells in tumor-bearing animals and enhances antitumor immune activity. Clin Cancer Res. 2005;11:6713–6721. doi: 10.1158/1078-0432.CCR-05-0883. [DOI] [PubMed] [Google Scholar]
- 122.Vincent J, Mignot G, Chalmin F, Ladoire S, Bruchard M, Chevriaux A, et al. 5-Fluorouracil selectively kills tumor-associated myeloid-derived suppressor cells resulting in enhanced T cell–dependent antitumor immunity. Cancer Res. 2010;70:3052–3061. doi: 10.1158/0008-5472.CAN-09-3690. [DOI] [PubMed] [Google Scholar]
- 123.Alizadeh D, Trad M, Hanke NT, Larmonier CB, Janikashvili N, Bonnotte B, et al. Doxorubicin eliminates myeloid-derived suppressor cells and enhances the efficacy of adoptive T-cell transfer in breast cancer. Cancer Res. 2014;74:104–118. doi: 10.1158/0008-5472.CAN-13-1545. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.Jamieson T, Clarke M, Steele CW, Samuel MS, Neumann J, Jung A, et al. Inhibition of CXCR2 profoundly suppresses inflammation-driven and spontaneous tumorigenesis. J Clin Invest. 2012;122:3127. doi: 10.1172/JCI61067. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125.Serafini P, Meckel K, Kelso M, Noonan K, Califano J, Koch W, et al. Phosphodiesterase-5 inhibition augments endogenous antitumor immunity by reducing myeloid-derived suppressor cell function. J Exp Med. 2006;203:2691–2702. doi: 10.1084/jem.20061104. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126.Corzo CA, Cotter MJ, Cheng P, Cheng F, Kusmartsev S, Sotomayor E, et al. Mechanism regulating reactive oxygen species in tumor-induced myeloid-derived suppressor cells. J Immunol. 2009;182:5693–5701. doi: 10.4049/jimmunol.0900092. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.Veltman JD, Lambers ME, van Nimwegen M, Hendriks RW, Hoogsteden HC, Aerts JG, et al. COX-2 inhibition improves immunotherapy and is associated with decreased numbers of myeloid-derived suppressor cells in mesothelioma. Celecoxib influences MDSC function. BMC Cancer. 2010;10:464. doi: 10.1186/1471-2407-10-464. [DOI] [PMC free article] [PubMed] [Google Scholar]