Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2018 May 15.
Published in final edited form as: Adv Drug Deliv Rev. 2017 Apr 25;114:206–221. doi: 10.1016/j.addr.2017.04.010

Progress in tumor-associated macrophage (TAM)-targeted therapeutics

Chayanon Ngambenjawong 1, Heather H Gustafson 1, Suzie H Pun 1,*
PMCID: PMC5581987  NIHMSID: NIHMS871371  PMID: 28449873

Abstract

As an essential innate immune population for maintaining body homeostasis and warding off foreign pathogens, macrophages display high plasticity and perform diverse supportive functions specialized to different tissue compartments. Consequently, aberrance in macrophage functions contributes substantially to progression of several diseases including cancer, fibrosis, and diabetes. In the context of cancer, tumor-associated macrophages (TAMs) in tumor microenvironment (TME) typically promote cancer cell proliferation, immunosuppression, and angiogenesis in support of tumor growth and metastasis. Oftentimes, the abundance of TAMs in tumor is correlated with poor disease prognosis. Hence, significant attention has been drawn towards development of cancer immunotherapies targeting these TAMs; either depleting them from tumor, blocking their pro-tumoral functions, or restoring their immunostimulatory/tumoricidal properties. This review aims to introduce readers to various aspects in development and evaluation of TAM-targeted therapeutics in pre-clinical and clinical stages.

Keywords: Tumor-associated macrophage, cancer immunotherapy, drug delivery, targeted therapy, tumor microenvironment

Graphical abstract

graphic file with name nihms871371u1.jpg

1. Introduction

Macrophages, key cells in the innate immune system, are the main component of the mononuclear phagocyte system (MPS) [1], which also include bone marrow progenitors and blood monocytes. Recent studies have revealed that the different components of the MPS system are derived in various stages of embryotic development and are not all from the same progenitor lineage [2][3]. Macrophage functions are as diverse as their lineages and play important roles in normal homeostasis and disease development [4][5]. This is especially true in acute and chronic inflammatory disease states such as wounds, malignancy, and autoimmune disorders. There is increasing evidence that macrophages play a central role in both normal and diseased tissue remodeling including angiogenesis, basement membrane breakdown, leukocyte infiltration, and immune suppression [4]. As such, the macrophage has emerged as a central drug target in a variety of disease states, including the tumor microenvironment (TME). This review focuses on drug strategies in malignancies that affect macrophage, while the reader is referred to excellent recent reviews for more information on other aspects of macrophage biology, including macrophage physiological function [5], the role of the macrophage in tumorigenesis [6], and the clinical applicability and translation [7] of macrophage drug targets.

The review first provides an introduction to macrophage and the role of macrophage populations in tumor development. Cancer therapy strategies involving macrophages in the following three areas are then presented: 1) identifying drug targets for modulation of TAM activities, 2) engineering carriers that promote, in increasing order of cell specificity, effective delivery of drug cargos to systemic macrophages, tumor-localized TAMs, or subtype-specific pro-tumoral M2-TAMs, and 3) utilizing the tumor-homing property of macrophages for cell-based therapies.

1.1 Mononuclear phagocyte system

First described in the starfish by Ilya Ilyich Mechnikov in 1882, phagocytes are an evolutionarily conserved subset of leukocytes that maintain organismal homeostasis in all multicellular species, [8]. This cell type, especially within mammalian species, is distributed throughout the body and present within each tissue to perform specific functions [3]. Microglia, the tissue-resident macrophages within the brain, facilitate neurological synapse pruning for memory maintenance [9] and serve as an immunological defense in an immune privileged environment [10]. Kupffer cells in the liver clear foreign, pathogenic, and waste materials from circulation [11]. Splenic red pulp macrophages are responsible for the recycling process of red blood cells [11][12][13]. Specific macrophage subsets in protective barriers, such as the Langerhans cells in the skin [14] and the alveolar macrophages within the lung [15], are responsible for both clearing pathogenic material and raising local inflammatory responses. Intraglomerular mesangial cells in the kidneys regulate blood flow [16]. The monocytes in blood circulation provide a reservoir of macrophages for mounting immune and inflammatory responses [17]. Further, the monocytes are derived from the bone marrow where hematopoietic progenitors generate monocytes and also osteoclastic cells which maintain bone function. Finally, there is mounting evidence to suggest that Hofbauer cells, the macrophages of the placenta, maintain signaling crosstalk from mother to baby during embryogenesis [18][19]. It is clear that MPS cells are part of a complex system that maintains the health and function of tissue and organs and therefore hold key roles in homeostasis and disease development. The complexities of different macrophage functions within different tissues are covered in several recent reviews [20][21][22].

1.2 Macrophage polarization

Macrophages are a plastic cell type capable of reacting to microenvironmental cues [23]. During pathogenesis, this cell type is among the first responders, recognizing pathogen-associated patterns (PAMPs) like lipopolysaccharide (LPS) [24]. LPS engages the Toll-like receptor 4 (TLR-4) on the surface of macrophages to activate transcription factors (e.g. interferon regulatory factors (IRFs) and nuclear factor kappaB (NF-κB)) to mount an inflammatory response [24][25]. These pro-inflammatory polarized macrophages (M(LPS)), release a variety cytokines, including IL-1β, IL-6, and TNF-α, facilitating the local recruitment of more macrophages and leukocyte infiltrate to fight against pathogenic insult [26]. In contrast, in wounding microenvironments, extensive release of inflammatory cytokines is potentially detrimental to overall tissue repair. Instead, macrophages respond to cytokines IL-4 and IL-13 released by damaged cells resulting in activation of the STAT6 transcription factor through the Janus kinase – signal transducers and activators of transcription (JAK-STAT) pathway, effectively turning on signature anti-inflammatory genes like arginase (Arg1) and resistin-like molecule α [26][27][28]. These gene regulatory pathways facilitate the recruitment of more immunoregulatory macrophages (M(IL-4)), release of immunoregulatory cytokines, and induction of processes like angiogenesis and basement membrane remodeling [29][30]. Based on pro-inflammatory and anti-inflammatory functions of macrophages under different stimuli, a broad classification has been proposed to generalize macrophages as classically activated M1 (pro-inflammatory, activated by LPS or interferon gamma (IFNγ)) or alternatively activated M2 (anti-inflammatory, activated by IL-4, IL-13, or IL-10) phenotypes [31]. Interestingly, these pro-inflammatory and anti-inflammatory pathways can converge with one another [32][33]. Oftentimes, wound sites are also susceptible to pathogen insult, so a balance between macrophage types is necessary for wound resolution. As such, both macrophage types exist within these environments, including macrophages that perform both functions and macrophages that are completely off the M1 and M2 spectrum [32][33]. Clearly, macrophage functions within a specific environment or in a specific disease or homeostatic state are complex. Nonetheless, macrophage activation within these environments and the key roles of macrophage in the development of resolution of complications or disease initiation are well-recognized. As such, macrophages are an important potential therapeutic target[26][34].

2. Macrophages in tumorigenesis

2.1 Macrophage polarization in tumor development

Macrophages are a large component of the leukocyte infiltrate into the TME. Over the last several decades, tumor-associated macrophages (TAMs) have been a subject of intense study for their impact on leukocytes, cytokines, and inflammatory mediators that either block or propagate tumor progression. Interestingly, the macrophage has been identified as a driver for inflammation, not only in cancer but in other disease states. Indeed, chronic inflammation that arises from diseases such as inflammatory bowel disease, silicosis, and asbestosis, pre-dispose these sites to cancer development. Readers are directed to Elinav et al.’s excellent review on this topic [35].

Interestingly, macrophages seem to have divided loyalties, on one hand promoting tumor resolution (M1/M(LPS)) and on the other propagating tumorigenesis (M2/M(IL-4)) (Fig. 1). Studies done almost a half century ago have shown that M1 macrophages have the capacity to kill and remove tumor cells, in line with the primary physiological function of M1 macrophages to remove foreign materials [36]. The M1 cells initiate cytokine production within the TME and facilitate tumor cell destruction through recruitment of pro-immunostimulating leukocytes and phagocytosis of tumor cells. However, studies within that same research period show that M2 macrophages have a central role in tumor propagation [37]. The M2 cells drive tumor development in both primary and metastatic sites through their contributions in basement membrane breakdown and deposition, angiogenesis, recruitment of leukocytes, and overall immune suppression [37][38][39]. It is important to note that, like normal homeostasis, macrophages within TME are not limited to M1/M(LPS) or M2/M(IL-4) states; they may reside in-between or off this spectrum. Removal of all macrophage populations regardless of polarization state has emerged as a potential therapeutic option, as there is a significant reduction in both primary and metastatic tumorigenesis [40]. However, as illustrated later within this review, the strategy has had a limited clinical impact, unless delivered in combination with other immunological agents, due to limitations in drug delivery and macrophage targeting [41]. On the other hand, the macrophage, regardless of polarization state retains the capacity for plasticity, including the ability to switch between phenotypes as a function of microenvironmental cues. Thus, altering the macrophage phenotype within TME from immune-suppressive to immune-promoting is currently being explored for therapeutic applications [41].

Fig. 1.

Fig. 1

Normal physiological macrophage functions that can promote (M2) or reduce tumor growth (M1).

2.2 Clinical implication of TAMs

Clinically, the presence of macrophages within primary tumors have been shown to be correlated with poorer prognosis in almost all tumors [7][42], with the exception of colon cancers [43]. Interestingly in recent years, clinicians have expanded these studies to investigate both M1 and M2 phenotypes within these microenvironments. Increasing levels M1 macrophages within these sites indicate better prognosis [44][45] whereas increasing levels of M2 macrophages [42] or decreased lymphocyte to monocyte ratios [46] predict poor outcomes. While these correlation studies have yet to be linked to causation, emerging therapeutic strategies aiming to remove macrophages and/or alter macrophage phenotypes are facilitating promising therapeutic benefits [41].

3 Pharmacological modulation of macrophages/TAMs

3.1 Bisphosphonate

Bisphophonates are a family of compounds structurally composed of a central carbon linked by two phosphate groups, R1 and R2 side groups where R1 is H, OH, or Cl and R2 comprises diverse functional groups which determine the potency of the compounds [47]. The first generation non-nitrogen containing bisphosphonates (etidronate, clodronate, and tiludronate) are intracellularly converted to a non-hydrolyzable ATP analogue leading to apoptosis [48]. The second generation (aliphatic amine R2) and third generation (aromatic amine R2) bisphosphonates induce apoptosis by inhibiting the essential enzyme farnesyl diphosphate (FPP) synthase. Bisphosphonates have a high affinity for hydroxyapatite and are frequently used in management of bone diseases such as osteoporosis, Paget disease, and bone metastases. Moreover, pre-clinical studies in murine breast tumor models suggest that bisphosphonate may also exhibit an extraskeletal therapeutic effect [49][50]. In this case, the bisphosphonate (zoledronic acid) primarily binds to microcalcifications present in breast tumors and is subsequently phagocytosed by TAMs to both induce apoptosis and promote M2-to-M1 repolarization. To improve pharmacokinetics, reduce toxic side effects (e.g. osteonecrosis of the jaw), and alter biodistribution away from bone for extraskeletal applications, bisphosphonates are typically formulated into liposomes or nanoparticles [51][52]. Although depletion of TAMs with liposomal clodronate (clodrolip) improves survival in some pre-clinical cancer models, to our knowledge, complete regression of tumors using bisphosphonate alone has not been achieved [51]. Some of the drugs shown to benefit from depletion of TAMs via bisphosphonates include anti-angiogenesis therapies (sorafenib and anti-VEGF antibody) and liposomal doxorubicin (Doxil) [51][53][54]. In addition to disrupting the pro-tumoral effect of TAMs, associated depletion of resident Kupffer cells has also been shown to reduce hepatic clearance of drug-loaded nanoparticles and hence prolong their plasma circulation time [54][55]. However, not all therapies benefit from depletion of macrophages/TAMs, especially immunotherapies designed to stimulate anti-tumor innate immunity [56][57][58]. In fact, an indiscriminate depletion of systemic macrophages such as via administration of clodronate liposomes may sometimes exacerbate the disease progression. As examples, depletion of subcapsular sinus CD169+ macrophages in tumor-draining lymph nodes is linked to increased tumor burden in B16F10 tumor model while the density of CD169+ macrophages in lymph nodes is also positively correlated with favorable prognosis in patients with colorectal and breast cancers [59][60][61]. These reports further suggest that TAM-targeted therapeutics that spare other potentially beneficial resident macrophages are more desirable than general macrophage depletion strategies. Clinically, bisphosphonates were among the first anti-cancer drug with reported activities on TAMs to be approved for human use, dating back to as early as 1995 for pamidronate and 2002 for zoledronic acid, where they were indicated for use in the management of multiple myeloma and bone-related metastasis (Table 1) [62].

Table 1.

FDA-approved drugs whose mechanisms of action may involve modulation of macrophages/TAMs

Drug Treatment Year approved (US FDA)
Bisphosphonate
Pamidronate/Aredia Multiple myeloma, metastatic breast cancer 1995
Zoledronic acid/Zometa Multiple myeloma, bone metastases from solid tumors 2002
Alkylating agent
Trabectedin/Yondelis Advanced soft-tissue sarcoma 2007 (EU) [63]
2015 (US)
Ovarian cancer 2009 (EU) [63]
Tyrosine kinase inhibitor
Imatinib/Gleevec Chronic myelogenous leukemia (CML) 2001
Dasatinib/Sprycel CML,Philadelphia chromosome-positive acute lymphoblastic leukemia (ALL) 2006
Chronic phase CML 2007
Chronic phase Philadelphia chromosome-positive CML 2010
Sunitinib/Sutent Gastrointestinal stromal tumor (GIST), advanced kidney cancer 2006
Pancreatic neuroendorcrine tumors 2011
Nilotinib/Tasigna CML 2007
Imaging agent
Tc 99m tilmanocept/Lymphoseek (CD206 targeting) Lymphatic mapping in breast cancer and melanoma 2013
Guiding Sentinel Lymph Node (SLN) Biopsy in squamous cell carcinoma of the oral cavity 2014

These drugs were not necessarily approved due to their actions on TAMs.

3.2 Inhibition of growth factor signaling

3.2.1 CSF-1R inhibition

Colony stimulating factor 1 receptor (CSF-1R) is a tyrosine kinase receptor expressed on mononuclear phagocytes [64]. Dimerization of the receptor upon binding to CSF-1 or IL-34 leads to a cascade of signaling events which promote proliferation, function, and survival of macrophages [64][65]. Antibodies targeting CSF-1 or CSF-1R as well as CSF-1R kinase inhibitors have been developed for therapeutic modulation of resident macrophages and TAMs [66]. AFS98 and M279 anti-mouse CSF-1R antibodies, capable of blocking both CSF-1 and IL-34, are widely used to study the effect of macrophage depletion in mouse tumor models and have been shown to reduce tumor size and improve survival in MMTV-PyMT spontaneous breast tumor model when administered both early and chronically [67][68][66]. While intraperitoneal administration of anti CSF-1R antibody systemically depletes resident macrophages in several major organs, a minimal effect is observed in brain, lung, ovary, and uterus likely due to anatomical accessibility (blood brain barrier), tissue turnover rate, or specific stimuli within the tissue environment [69]. On the other hand, a small molecule CSF-1R kinase inhibitor (BLZ945), which readily passes through blood brain barrier, has been shown to be effective in murine models of glioma, breast cancer, and cervical cancer [70][71]. It is worth noting that tumor recurrence in a murine glioma model following prolonged treatment with BLZ945 is attributed to increased level of IL-4 which enriches TAMs with the CD206+ M2-like phenotype [72]. These IL-4 stimulated TAMs secrete insulin-like growth factor-1 (IGF-1) to enhance survival of rebound glioma cells via stimulation of the phosphatidylinositol 3-kinase (PI3k) pathway. A combination therapy of BLZ495 with IGF-1R inhibitor significantly prolonged survival of mice with rebound glioma compared to monotherapy of either BLZ495 or IGF-1R inhibitor highlighting the need for rational design of combinatorial therapy for maximized response to TAM-directed anti-cancer therapy. Due to structural similarities among different receptor tyrosine kinases, tyrosine kinase inhibitors typically recognize multiple targets [73] and as such, several FDA-approved, multi-target tyrosine kinase inhibitors (Table 1), initially developed for other targets such as c-KIT, VEGFR, and PDGFR (reviewed in [74]), have now been shown to exhibit activity on the CSF-1R kinase domain [75][76][77]. New developments of more CSF-1R-specific inhibitors (e.g. PLX3397, PLX7486, BLZ945, and ARRY-382) are also being investigated in clinical trials (Table 2). In particular, PLX3397, developed by structure-guided design against the CSF-1R target, exhibits higher affinity and selectivity to CSF-1R than imatinib and demonstrates better efficacy in treatment of tenosynovial giant cell tumor [78]. As a result, the drug has been designated breakthrough therapy status and advanced into clinical trial phase III for the indication. Antibodies targeting CSF-1R are also entering Phase I and II clinical trials for treatment of solid tumors.

Table 2.

Selected compilation of drugs in clinical trials whose mechanisms of action may involve modulation of macrophages/TAMs

Drug/Description Sponsors and collaborators Phase/Status Tumor type Treatment ClinicalTrials.govIdentifier First received
CSF1R inhibitor
Pexidartinib/PLX3397 (Oral c-fms/KIT/FLT3 tyrosine kinase inhibitor) Centre Leon Berard and Plexxikon, AstraZeneca 1 Ongoing Advanced/metastatic colorectal or pancreatic cancer With Durvalumab (Anti-PD-L1 antibody) NCT02777710 May 12, 2016
Plexxikon and Merck Sharp & Dohme Corp. 1/2a Ongoing Advanced melanoma and other solid tumors With Pembrolizumab (Anti-PD-1 antibody) NCT02452424 May 20, 2015
Daiichi Sankyo Inc. 3 Ongoing Pigmented villonodular synovitis (PVNS), giant cell tumors of the tendon sheath (GCT-TS), or tenosynovial giant cell tumor (TGCT) Monotherapy NCT02371369 February 19, 2015
PLX7486 (Oral c-fms/Trk tyrosine kinase inhibitor, tosylate salt form) Plexxikon 1 Ongoing Solid tumors or tumors of any history with activating Trk point or NTRK fusion mutation Monotherapy NCT01804530 March 1, 2013
BLZ945 (Oral c-fms tyrosine kinase inhibitor) Novatis Pharmaceuticals 1/2 Ongoing Advanced solid tumors Monotherapy and with PDR001 (Anti-PD-1 antibody) NCT02829723 July 8, 2016
ARRY-382 (Oral c-fms tyrosine kinase inhibitor) Array BioPharma 1b/2 Ongoing Advanced solid tumors With Pembrolizumab (Anti-PD-1 antibody) NCT02880371 August 2, 2016
AMG 820 (Human IgG2 anti-CSF-1R antibody) Amgen 1b/2 Ongoing Advanced solid tumors With Pembrolizumab (Anti-PD-1 antibody) NCT02713529 March 3, 2016
Emactuzumab/RO5509554/RG7155 (Human IgG1 anti-CSF-1R antibody) Hoffmann-La Roche 1 Ongoing Solid cancers With MPDL3280A (Tecentriq/anti-PD-L1 antibody) NCT02323191 December 5, 2014
Hoffmann-La Roche 1 Ongoing Solid tumors With RO7009789 (Anti-CD40 agonistic antibody) NCT02760797 April 14, 2016
M.D. Anderson Cancer Center and Genentech Inc. 2 Ongoing Platinum-resistant, epithelial ovarian, fallopian tube or primary peritoneal Cancer With paclitaxel and Bevacizumab (Avastin/anti-VEGF antibody) NCT02923739 September 30, 2016
LY3022855/IMC-CS4 (Human IgG1 anti-CSF-1R antibody) Eli Lilly and Company and AstraZeneca 1a/1b Ongoing Advanced solid tumors With Durvalumab (Anti-PD-L1 antibody) or Tremelimumab (Anti-CTLA-4 antibody) NCT02718911 March 21, 2016
Cabiralizumab/FPA008 (Anti-CSF-1R antibody) Five Prime Therapeutics Inc. and Bristol-Myers Squibb 1a/1b Ongoing Advanced solid tumors Wth BMS-936558 (Nivolumab/anti-PD-1 antibody) NCT02526017 August 13, 2015
RON/MET inhibition
ASLAN002/BMS777607 (Oral RON/MET receptor tyrosine kinase inhibitor) Aslan Pharmaceuticals 1 Completed Malignant solid tumors Monotherapy NCT01721148 November 1, 2012
CD47-SIRPα inhibitor
Hu5F9-G4 (Human IgG4 Anti-CD47 antibody) Forty Seven Inc. 1b/2 Ongoing Colorectal neoplasms or solid tumors With Cetuximab (Erbitux/anti-EGFR antibody) NCT02953782 November 1, 2016
Forty Seven Inc. 1b/2 Ongoing Non-Hodgkin’s lymphoma or diffuse large B-cell diffuse lymphoma, indolent lymphoma With Rituximab (Anti-CD20 antibody) NCT02953509 November 1, 2016
CC-90002 (Anti-CD47 antibody) Celgene 1 Ongoing Acute myeloid leukemia or myelodysplastic syndromes Monotherapy NCT02641002 November 12, 2015
TTI-621 (SIRPα-Fc fusion) Trillium Therapeutics Inc. 1 Ongoing Solid tumors or mycosis fungoides Monotherapy NCT02890368 August 26, 2016
ALX148 (High affinity SIRPα variant) Alexo Therapeutics Inc. 1 Ongoing Metastatic cancer, solid tumor, advanced cancer, or non-Hodgkin’s lymphoma Monotherepay, with Atezolizumab (Tecentriq/anti-PD-L1 antibody) or Transtuzumab (Herceptin/anti-HER2 antibody) NCT03013218 December 15, 2016
CCR2-CCL2 inhibitor
PF-04136309/PF6309 (Small molecule CCR2 antagonist) Pfizer 1b/2 Ongoing Metastatic pancreatic ductal adenocarcinoma With Nab-paclitaxel and gemcitabine NCT02732938 February 29, 2016
MLN1202/S0916/plozalizumab (Human anti-CCR2 antibody) Millennium Pharmaceuticals Inc. 1 Ongoing Melanoma With Nivolumab (Anti-PD-1 antibody) NCT02723006 March 25, 2016
Carlumab/CNTO 888 (Human IgG1κ anti-CCL2 antibody) Centocor Inc. 1 Completed Cancer With DOXIL, gemcitabine, paclitaxel + carboplatin, or docetaxel NCT01204996 September 16, 2010
CXCR4-CXCL12 inhibitor
Mozobil/AMD-3100/plerixafor* (Small molecule CXCR4 antagonist) CCTU-Cancer Theme, Sanofi, Stand Up To Cancer, CRUK Cambridge Institute, Lustgarten Foundation, and National Institute for Health Research (UK) 1 Ongoing Metastatic pancreatic adenocarcinoma, ovarian serous adenocarcinoma, or metastatic colorectal cancer Monotherapy NCT02179970 July 1, 2014
LY2510924 (CXCR4 agonistic peptide) Eli Lilly and Company and AstraZeneca 1 Ongoing Solid tumors With Durvalumab (Anti-PD-L1 antibody) NCT02737072 April 8, 2016
CD40 agonist
CP-870,893/RO7009789 (Human IgG2 anti-CD40 antibody) Abramson Cancer Center of the University of Pennsylvania 1 Ongoing Recurrent/stage IV melanoma with Tremelimumab (Anti-CTLA-4 antibody) NCT01103635 April 12, 2010
Hoffmann-La Roche 1 Ongoing Solid cancers with MPDL3280A (Tecentriq/anti-PD-L1 antibody) NCT02304393 November 26, 2014
Abramson Cancer Center of the University of Pennsylvania 1 Ongoing Pancreatic cancer With nab-paclitaxel and gemcitabine NCT02588443 October 21, 2015
Hoffmann-La Roche 1 Ongoing Advanced/metastatic solid tumors With Vanucizumab (Bispecific anti-Ang2/VEGF-A antibody) NCT02665416 January 15, 2016
Hoffmann-La Roche 1 Ongoing Solid tumors With Emactuzumab (Anti-CSF-1R antibody) NCT02760797 April 14, 2016
Vasculature-modulating agent
Vanucizumab (Human bispecific anti-Ang2/VEGF antibody) Hoffmann-La Roche 1 Ongoing Neoplasms With Atezolizumab (Anti-PD-L1 antibody) NCT01688206 September 13, 2012
Hoffmann-La Roche 1 Ongoing Advanced/metastatic solid tumors With RO7009789 (Anti-CD40 antibody) NCT02665416 January 15, 2016
Vadimezan/5,6-dimethylxanthenone-4-acetic acid (DMXAA) (Vasculature-disrupting agent) Swiss Group for Clinical Cancer Research 2 Completed Lung cancer With carboplatin and paclitaxel NCT01057342 January 26, 2010
PI3kγ inhibitor
Duvelisib/IPI-549 (Oral PI3kδ/γ inhibitor) Infinity Pharmaceuticals Inc. 1 Ongoing Advanced solid tumors, non-small cell lung cancer (NSCLC), melanoma, or squamous cell cancer of the head and neck With Nivolumab (Anti-PD-1 antibody) NCT02637531 December 16, 2015

This compilation highlights the recently-initiated clinical trials with emphasis on combination therapy.

“Ongoing” includes both recruiting and non-recruiting stages.

*

Approved for use in combination with G-MCSF to mobilize hematopoietic stem cells to peripheral blood for collection and subsequent autologous transplantation in patients with non-Hodgkin’s lymphoma and multiple myeloma.

3.2.2 RON inhibition

RON (Recepteur d’Origine Nantais) is a receptor tyrosine kinase expressed on tissue-resident macrophages that binds to macrophage-stimulating protein (MSP) after its proteolytic activation at inflammation sites [79]. RON signaling in macrophages promotes cell spreading and phagocytosis as well as enhances M2 polarization via stimulation of arginase expression and attenuation of responses to pro-inflammatory stimuli e.g. IFNγ and LPS [80][81][82][83]. In prostate and breast cancer models, RON signaling in macrophages has been shown to impair anti-tumor functions of CD8+ T cells leading to promotion of tumor growth and metastatic outgrowth respectively [84][85]. Pharmacologic blockade of RON kinase with an inhibitor, BMS-777607/ASLAN002, boosts the population of pro-inflammatory TNF-α-secreting macrophages and reduces metastatic outgrowth in the lungs in the PyMT-MSP breast tumor model [85]. Alternatively, restoration of iNOS expression in MSP-stimulated macrophages may be achieved via inhibition of PI3k, one of the downstream targets of RON activation [83]. In fact, a synergistic therapeutic effect between the RON kinase inhibitor and a PI3k inhibitor NVP-BKM120 has been reported in a mouse xenograft model of patient-derived breast tumor where cancer cells express constitutively active isoform of RON (short-form RON or sfRON) [86]. Both RON and PI3k inhibitors are being investigated in clinical trials [85][87].

3.3 Modulation of macrophage phagocytic activity

3.3.1 Antibody-dependent cellular phagocytosis (ADCP)

Two important innate immune functions of macrophages are phagocytosis of foreign bodies, apoptotic cells, as well as cancer cells and processing of the engulfed materials for antigen presentation to stimulate adaptive immunity [88]. Tumor-targeted antibodies are a powerful class of anti-cancer biologics that act through direct inhibition of survival signaling, mediation of antibody-dependent cellular cytotoxicity (ADCC) by natural killer (NK) cells, induction of complement-dependent cytotoxicity (CDC) via complement cascade activation, and facilitation of antibody-dependent cellular phagocytosis (ADCP) by macrophages [89]. Clinically approved anti-cancer monoclonal antibodies such as rituximab and trastuzumab have been demonstrated to exert their therapeutic effects, primarily or in part, through macrophage-mediated ADCP [90][91]. Contrary to their capability to promote tumor invasion, TAMs are also able to phagocytose cancer cells in the presence of the opsonizing tumor-targeted antibodies [92]. Nonetheless, the therapeutic efficacy of these antibodies may be limited by counter mechanisms including antigenic modulation and trogocytosis, both of which remove antibody-antigen complexes off target cell plasma membrane [90][93]. Of note, trogocytosis has recently been shown to induce cell death complimentary to whole cell phagocytosis [94]. To further improve ADCP activity, the Fc region of tumor-targeted antibodies could be engineered to increase its interaction with activatory Fc receptors, most importantly FcγRIIa, on macrophages [95]. While development of clinical antibodies is primarily focused on IgG, therapeutic potentials of other Ig isotypes (IgA and IgE) are being investigated in several pre-clinical studies where monocytes/macrophages are also important in effecting the ADCC/ADCP functions [96][97].

3.3.2 Inhibition of CD47-SIRPα signaling

Successful development and maturation of tumors require effective evasion from such immune-surveillance activities [98]. In several tumor types, cancer cells block phagocytosis by upregulating surface expression of CD47 which interacts with signal regulatory protein alpha (SIRPα) on macrophages to transmit the “don’t eat me” signal. Inhibiting the CD47-SIRPα signaling axis with anti-CD47 antibodies or an engineered SIRPα-Fc fusion has been shown to restore macrophage ability to phagocytose cancer cells and prime a cytotoxic CD8+ T-cell response, leading to a significant reduction in tumor size and metastasis in several cancer models [98][99]. With promising results in pre-clinical studies, numerous clinical trials have been initiated in the past few years to investigate different therapeutic variants including anti-CD47 antibody (Hu5F9-G4 and CC-90002), engineered high affinity SIRPα (ALX148), and SIRPα-Fc fusion (TTI-621) (Table 2). One potential challenge in clinical translation of anti-CD47 therapies is the ubiquitous expression of CD47 on red blood cells and hematopoietic stem cells which may serve as an antibody sink to reduce efficacy while also causing transient anemia [100][101]. Alternative strategies have been investigated including targeted blocking of SIRPα with engineered high affinity CD47 ectodomain or development of bispecific antibody targeting both CD47 and tumor-associated antigen [102][101]. To minimize possible off-target toxicity, anti-CD47 antibody may be developed using the Fc scaffold of IgG4 to reduce Fc effector functions, or the engineered SIRPα without Fc fusion may be used in combination with Fc-active tumor-targeted antibodies [100][103].

3.4 Inhibition of inflammatory monocyte/macrophage recruitment

3.4.1 Inhibition of CCR2-CCL2 signaling

The number of intratumoral TAMs increases with tumor growth due to self-proliferation as well as recruitment and differentiation from circulating inflammatory Ly6C+CCR2+ monocytes [104]. The latter is mediated by elevated secretion of CCL2 (also known as monocyte chemoattractant protein-1 (MCP-1)), the ligand for CCR2, by cancer and stromal cells in tumors and associated metastases [105][106][107]. As a chemotactic factor, CCL2 mobilizes bone marrow-resident inflammatory monocytes into circulation and subsequently into the tumors. High expression of CCL2 in tumors and high count of CCR2+ monocytes in the peripheral blood are both correlated with poor prognosis in several cancer types [108][105][109]. Strategies to pharmacologically interrupt CCR2-CCL2 signaling axis (e.g. CCR2 small molecule inhibitor, anti-CCR2 antibody, and anti-CCL2 antibody) have been developed as anti-cancer therapies [110]. In a pancreatic mouse cancer model, a small molecule CCR2 inhibitor (PF-04136309) effectively reduces recruitment of inflammatory monocytes into the tumors resulting in reduced tumor growth and fewer liver metastases [105]. Anti-CCL2 antibodies have been shown to prevent metastasis in breast and prostate cancer models [111][112]. However, in the breast cancer model, discontinuation of the treatment accelerated metastases as a result of increased tumor localization of monocytes that were originally sequestered in bone marrow during the antibody treatment [111].

3.4.2 Inhibition of CXCR4-CXCL12 signaling

In addition to the CCR2-CCL2 signaling axis, CXCR4-CXCL12 (also known as stromal cell-derived factor-1 (SDF-1)) interaction is another signaling axis involved in recruitment of monocytes/macrophages and implicated in promotion of tumor invasiveness/regrowth [113][114]. Mechanistically, CXCL12 is secreted by both tumor and stromal cells including TAMs, and hence, mediates both autocrine and paracrine signaling in recruiting and differentiating monocytes expressing its receptor CXCR4 into tumor-infiltrating TAMs [115]. In glioma and breast cancer models, anti-cancer treatments with radiation therapy or chemotherapy have been shown to increase the level of CXCL12 in the perivascular region of tumors which in turn promotes accumulation of CXCR4+ TAMs [115][114]. Secretion of vascular endothelial growth factor A (VEGF-A) by these TAMs stimulates formation of new tumor vasculature and tumor relapse, the effects of which could be inhibited by treatment with CXCR4 inhibitor (AMD3100). Given multiple signaling pathways in monocyte/macrophage recruitment, simultaneous blockades of different signaling axes may be necessary to achieve the optimal effect.

3.5 Immunostimulation by anti-CD40 agonistic antibody

CD40 is a member of the tumor necrotic factor (TNF) receptor family expressed on antigen-presenting cells (APCs), monocytes, B cells, as well as in some tumors [116]. Since ligation between CD40 on APCs and CD40 ligand (CD40L) on T cells stimulates these APCs to activate effector T cells for anti-tumor response, the therapeutic effect of anti-CD40 agonistic antibody was thought to act through a similar mechanism. However, in a KPC mouse model of pancreatic ductal adenocarcinoma (PDA), depletion experiments of different immune cell populations revealed that the anti-tumor effect of the antibody could be mediated in a T cell-independent manner, requiring only the tumor infiltration of macrophages [117]. Mechanistically, systemic administration of anti-CD40 antibody raises the serum level of CCL2 and IFNγ where the former recruits inflammatory monocytes to the tumors, and the latter reprograms the recruited monocytes/macrophages for tumoricidal activities [118]. Interestingly, further optimization of anti-CD40 antibody by testing different IgG subclasses indicates that its agonistic immunostimulatory activity requires effective engagement of the antibody Fc domain with inhibitory FcγRIIb but not with other activatory Fc receptors (needed for ADCC/CDC) [119]. Consistent with the pre-clinical finding that the agonistic activity of anti-CD40 antibody is mediated more robustly when the activatory/inhibitory Fc receptor binding ratio (A/I) is lower [120], Pfizer’s human anti-CD40 antibody of IgG2 subclass (CP-870,893) has been shown in clinical trials to be more agonistic in immunostimulation compared to the other candidates (e.g. dacetuzumab, Chi Lob 7/4, and HCD122), the rest of which have IgG1 subclass (Table S1) [116].

3.6 Inhibition of angiogenic signaling

One of the essential pro-tumoral functions of macrophages is promotion of angiogenesis. The TIE2+ subpopulation, also called TIE2-expressing monocytes/macrophages (TEMs), has been shown to predominantly contribute to this role [6]. TEMs are typically recruited from circulation into the perivascular region of tumors where their pro-angiogenic activities are stimulated upon interaction of TIE2 with the corresponding ligand angiopoietin 2 (ANG2), secreted in several tumors [121][122]. Mazzieri et al. demonstrated that therapeutic blockade of ANG2 with an anti-ANG2 antibody reduced angiogenesis and tumor growth in mammary and pancreatic tumor models by preventing association between TEMs and angiogenic blood vessels as well as suppressing expression of TIE2 in TEMs [121]. Since upregulation of ANG2 is one of the resistance mechanisms to anti-VEGF therapy, dual targeting of both ANG2 and VEGF via either combination therapy or bispecific antibody has been investigated as a strategy to circumvent the therapeutic resistance [123][124]. Interestingly, in both highly vasculature-aberrant Gl261 as well as less aberrant MGG8 glioblastoma models, treatment with bispecific anti-ANG2/VEGF-A antibody (CrossMab, A2V) successfully improved survival the effect of which was attributed to the antibody-induced M2-to-M1 reprogramming of TAMs [124]. Similarly, a vascular-disrupting agent 5,6-dimethlxanthenone-4-acetic acid (DMXAA), initially developed for disrupting tumor vasculature, has also been shown to activate immunostimulatory functions of TAMs, which in turn orchestrate anti-tumor response of CD8+ T cells [125].

3.7 Metabolic modulation of macrophages/TAMs

3.7.1 mTOR inhibition

Macrophages of different functional states vary in their use of metabolic pathways for energy and metabolite production to support their specialized cellular activities [126]. Mammalian target of rapamycin (mTOR) signaling through mTOR complex 1 (mTORC1) and mTORC2 serves a central role in sensing nutrients, oxygen, and metabolites to direct metabolic programming of these macrophages [127]. In addition to its direct cytotoxic effect on cancer cells [128], rapamycin, an mTORC1-specific inhibitor, has been shown to stimulate macrophages to the M1-like phenotype with an anti-tumor effect in Huh-7 hepatocarcinoma mouse model [129]. Of note, other signaling molecules upstream of mTOR (e.g. PI3kγ, protein kinase B (Akt), and phosphatase and tensin homolog deleted on chromosome 10 (PTEN)) are also involved in shaping polarization and functions of macrophages/TAMs, making them potential targets for anti-cancer therapies [130][131][132]. As an example, Kaneda et al. recently showed that PI3kγ inhibitor (IPI-549) retarded tumor growth in implanted human papilloma virus positive head and neck squamous cell carcinoma (HPV+HNSCC), lung carcinoma, and breast carcinoma models in a TAM-dependent manner by promoting TAM-immunostimulatory responses [130]. Similarly, expression of PTEN (PI3k suppressor) or silencing of Akt1 can also promote anti-tumor M1 macrophage polarization [131][132].

3.7.2 Modulation of PPARγ-Gpr132-lactate signaling

The TME is enriched for lactate which is secreted by cancer cells performing glycolysis [133]. It is now known that the recognition of lactate by Gpr132 (G-protein coupled receptor) on TAMs promotes M2 polarization of these TAMs while the expression of Gpr132 is under a suppressive control of peroxisome proliferator-activated receptor (PPARγ) transcription factor [134][135]. Several strategies have been developed to interrupt this signaling axis. For example, supply of lactate for M2-TAM polarization could be blocked using an inhibitor of lactate dehydrogenase (oxamic acid) [134]. Alternatively, PPARγ agonists (e.g. thiazolidinedione family of drugs) or siRNA silencing of Gpr132 have been successfully used to insensitize TAMs to lactate stimulation with a tumor-shrinking effect in EO771 breast tumor model [135].

3.7.3 Inhibition of metabolism of amino acids and lipids

In addition to competition for glucose, TAMs readily catabolize arginine and tryptophan which are essential amino acids required for anti-tumor T cell functions [126]. Restoration of these nutrients by inhibiting arginase, nitric oxide synthase, and indoleamine 2, 3-dioxygenase (IDO) constitutes another promising therapeutic intervention to improve T cell survival and functions [136][137]. Moreover, TAMs are also known to mediate immunosuppressive and metastasis-promoting functions in response to cancer cell/TAM-secreted lipid metabolites such as prostaglandin E2 (PGE2) and sphingosine-1-phosphate (S1P) [138][139][140]. Pharmacological inhibition of PGE2-producing enzymes microsomal PGE2 synthase 1 (mPGES1) and cyclooxygenase-2 (COX-2), with CAY10526 and celecoxib respectively, effectively reduced PGE2 level in bone marrow cell/MBT-2 bladder cancer cell coculture with celecoxib being further shown to lower expression of T cell-suppressive programmed cell death ligand 1 (PD-L1) in an MBT-2 cancer model and also promote M2-to-M1 polarization of TAMs in an Apcmin/+ colon cancer model [139][141].

3.7.4 Modulation of essential elements and vitamins

Iron metabolism is vastly different between M1- and M2-polarized macrophages [142]. M1 macrophages upregulate ferritin to promote intracellular iron retention whereas M2 macrophages upregulate ferroportin to enhance iron efflux. Consistent with the M2-like phenotype, TAMs preferentially liberate iron and provide cancer cells with this essential element for promoting proliferation. Several iron chelators are being evaluated as anti-cancer agents [143]. Iron chelation therapy has recently been shown to reverse iron-processing function of M2 macrophages from iron release towards sequestration and block tumor-promoting effect of the macrophages [144]. Conversely, external supply of iron through administration of ferumoxytol iron oxide nanoparticles has also been shown to be therapeutic at inhibiting tumor growth and metastasis by stimulating pro-inflammatory macrophage polarization and production of ROS [145]. These seemingly contradictory results regarding iron supply and macrophage response emphasize the need for further study in this area.

In another example, macrophages may secrete a host-defense peptide, cathelicidin, shown to readily lyse proliferating B cell lymphoma cells but not normal B cells [146]. Cathelicidin production requires intracellular metabolism of 25-hydroxyvitamin D (25D) into bioactive 1,25-dihydroxyvitamin D (1,25D3) by vitamin D-1-hydroxylase CYP27B1, which is expressed at lower levels in M2 macrophages and TAMs compared to M1 macrophages. Treating M2 macrophages with 1,25D3 successfully restores cathelicidin production and cytotoxicity against the B cell lymphoma cells[146].

3.8 Modulation of macrophage scavenger receptors

Scavenger receptors comprise a family of receptors that broadly recognize modified low-density lipoproteins, polyanions, endogeneous proteins, as well as conserved microbial structures and are important for clearing foreign particles, pathogens, and apoptotic cells from the body [147]. Several scavenger receptors (e.g. scavenger receptor class A (SR-A1), macrophage receptor with collagenous structure (MARCO), CD36, CD68, CD163, and receptor for advanced glycation endproducts (RAGE)) are expressed on macrophages, some of which are involved in regulation of tumor immunity and are therefore potential targets for therapeutic modulation [148][149]. As an example, apolipoprotein A-I mimetic peptide 4F, a competing ligand for SR-A1, has been shown to inhibit tumor growth and metastasis in ovarian and pancreatic tumor models [150]. The proposed mechanisms of action involve interrupting TAM/cancer cell crosstalk, blocking SR-A1 from scavenging ECM components that allow for cancer cell migration, and scavenging of pro-inflammatory/angiogenic lysophosphatidic acid (LPA) [151]. However, in the context of glioma model, Zhang et al. demonstrated that recognition of tumor-secreted heat shock protein 70 (HSP70) by SR-A1 is important in suppressing M2 polarization in TAMs and is needed for inhibition of glioma proliferation and angiogenesis [152]. These studies highlight the context-dependent effect in modulation of scavenger receptor activity as a result of its recognition of a broad range of ligands.

3.9 Chemotherapy drugs

Although chemotherapy drugs are primarily developed to induce cell death in rapidly dividing cancer cells, many also have pharmacological effects on non-cancer cell populations. In particular, trabectedin and its second generation lurbinectedin are effective at killing TAMs in addition to cancer cells [153][154]. Mechanistically, trabectedin interacts with tumor necrosis factor-related apoptosis inducing ligand receptor 2 (TRAIL-R2) on mononuclear phagocytes leading to receptor clustering and subsequent caspase-8 dependent activation of apoptosis [154][155]. Its selective toxicity in TAMs over neutrophils and lymphocytes has been attributed to higher expression of the receptor in conjunction with lower expression of non-signaling decoy TRAIL-R3. In addition, trabectedin also inhibits production of chemoattractant cytokine CCL2 by TAMs and tumor cells [156]. Apart from exerting a cytotoxic effect, several chemotherapy drugs modulate macrophage responses to tumor [157,158]. In murine fibrosarcoma and mammary tumor models, microtubule-stabilizing agents such as docetaxel and paclitaxel have been shown to promote polarization of myeloid-derived suppresser cells (MDSCs) to macrophages with anti-tumor M1-phenotype via suppression of STAT3 phosphorylation [159,160]. Cyclophosphamide treatment promotes infiltration of macrophages, enhances secretion of pro-inflammatory cytokines (IL-6 and IL-12), and suppresses production of pro-tumoral M2-related cytokines (IL-4, IL-10, and IL-13) [161][162]. As a self-protective mechanism against chemotherapies, chemo-resistant cancer cells secrete IL-34 which enhances their survival as well as promotes M2 polarization of TAMs to further reinforce immunosuppressive functions [163]. In addition, chemotherapy treatments may also increase intratumoral level of CXCL12, which serves as a chemotactic factor in recruiting TIE2+CXCR4+ macrophages to the perivascular region of the tumor where they preferentially promote angiogenesis and tumor regrowth [113]. To combat these resistance mechanisms, an integration of chemotherapy and immunotherapy may allow for a more effective tumor regression. Of note, paclitaxel and gemcitabine are two chemotherapy drugs commonly investigated in a combination therapy with other TAM-modulating therapeutics due to their possible promotion of antigen release following cancer cell death (Table 2 and S1).

3.10 Activation of Toll-like receptors (TLRs)

Toll-like receptors (TLRs), expressed on either plasma membranes (TLR1, 2, 4, 5, 6, and 10) or endosomal membranes (TLR3, 7, 8, and 9) of APCs, recognize various PAMPs and damage-associated molecular patterns (DAMPs), constituting an indispensable system for innate immune surveillance [164]. In regard to modulation of TAMs, several TLR agonists (e.g. polyinosinic:polycytidylic acid (polyI:C) for TLR3, LPS for TLR4, imiquimod for TLR7, and CpG-olionucleotide for TLR9) have been shown to activate NF-κB leading to pro-inflammatory M1 polarization (reviewed in [165]). Moreover, TLR signaling may also be initiated by chemotherapy drugs via induction of immunogenic cell death (ICD) [166]. Oftentimes, engaging multiple TLRs is essential to effect a more potent anti-tumor response. As an example, a recent study by Zheng et al. reported development of a dual TLR4/5 stimulating bacterial cell therapy (BCT) based on attenuated ΔppGpp Salmonella typhimurium (TLR4 agonist) with inducible production of Vibrio vulnificus flagellin B (FlaB, TLR5 agonist) [167]. The dual stimulation was more effective at reprogramming TAMs from CD206+ M2 phenotype to immunostimulating CD86+ M1 phenotype. Despite the anti-tumor effect of NF-κB activation being observed in TLR agonistic therapies, an opposite finding has also been reported where inhibition of IκB kinase β (IKKβ), the main downstream activator molecule of NF-κB, repolarizes TAMs to IL-12-secreting, NO-producing M1 phenotype with a growth inhibition effect in an ID8 ovarian tumor model [168]. These opposing reports on pro-tumoral/anti-tumoral effects of NF-κB activation may imply involvement of other determining parameters (e.g. the presence of other signaling factors that shape TLR responses or the effects of TLR signaling on other cell populations in TME) that collectively dictate the overall response to the therapies and highlight the need to thoroughly understand the immunological context of tumor for effective tailoring of anti-cancer therapy.

4 Targeted drug delivery systems to macrophages/TAMs

With promising drugs for different molecular targets being identified that could potentially modulate TAMs to effect an anti-tumor response, the next challenge has been how to deliver these immunomodulating drugs effectively and selectively to TAMs while minimizing off-target side effects. The extensive number of studies in the cancer nanomedicine field investigating passive targeting to tumor via the enhanced permeability and retention (EPR) effect has provided information regarding the biodistribution of nanoparticle systems in vivo. From this work, we have also gained more understanding about parameters that influence internalization by macrophages (e.g. Kupffer cells), the main player in the mononuclear phagocyte system responsible for clearance of nanomedicines. This section describes parameters that dictate passive targeting of macrophages/TAMs as well as targeting ligands being investigated pre-clinically for active targeting of macrophages/TAMs.

4.1 Passive targeting to macrophages/TAMs

Due to their primary role in clearing cellular debris, pathogens, and foreign substances, macrophages possess high phagocytic capability. Micro/nanoparticles are relatively indiscriminately internalized by macrophages, although the rate and extent of uptake is affected by properties such as shape, size, rigidity, contact angle, and surface charge (Fig. 2) [169][170][171][172]. For nanoparticles/liposomes larger than 100 nm, most studies report increased uptake with increasing particle size up to about 1 - 3 μm, after which uptake declines [169][173]. On the other hand, for nanoparticles/liposomes smaller than 100 nm, most studies report increased uptake with decreasing particle size [174][175][176]. Contrasting trends are also observed which may be partially attributed to differences in nanoparticle composition and macrophage source, as well as involvement of other uptake mechanisms [177][178][179]. In term of charge, nanoparticles with either highly positive or highly negative zeta potentials are preferentially internalized compared to the ones with near neutral or slightly negative zeta potentials [169][177]. In general, rigid and spherical nanoparticles are more readily uptaken by macrophages than soft and cylindrical nanoparticles [172][180][181][182][183].

Fig. 2.

Fig. 2

Strategies to promote passive and active targeting of macrophages/TAMs.

Recently, MacParland et al. reported a comparative study on nanoparticle uptake by human monocyte-derived macrophages of different phenotypic polarization [184]. In general, M2-polarized human macrophages exhibit higher internalization of gold nanoparticles than the M1-polarized cells, and the extent of uptake is positively correlated with the expression of M2 markers CD163 and CD206. Further investigation on the nanoparticle uptake by unstimulated and stimulated M1- or M2-polarized Kupffer cells from deceased donors showed that the unstimulated Kupffer cells, possessing a mixed M1/M2 phenotype, exhibited comparable internalization of nanoparticles to the M2-polarized cells, both of which ingested more nanoparticles than the M1-polarized cells did.

While the natural tendency of M2 macrophages to more readily internalize nanoparticles could be beneficial for designing TAM-targeted therapy, the resident macrophages’ penchant for internalizing nanoparticles is still an important challenge in developing TAM-targeted therapies [185][186][181][187]. While this ability might be exploited to target TAMs in liver cancer or metastasis, delivery of NP-based formulations to TAMs at other sites must compete with avid internalization by hepatic and splenic macrophages [188][180]. In fact, the current mainstay for depletion of TAMs in animal cancer models utilizes liposomal clodronate, which relies on passive accumulation of liposomes in the macrophages and therefore indiscriminately affects both TAMs and resident macrophages [189][52].

4.2 Active targeting to macrophages/TAMs

Targeting ligands are also employed to facilitate preferential delivery to TAMs via specific ligand-receptor interaction (Fig. 2). Mannose receptor (CD206) is one of the most commonly targeted receptors for macrophage delivery due to its overexpression on M2 macrophages [190]. As a simple sugar, the native ligand mannose can be easily conjugated to carriers but binds with low affinity. Thus, anti-CD206 antibody has also been developed for targeting TAMs [191]. Although CD206-targeted drug delivery systems can achieve high M2/M1 selectivity, they are readily sequestered in liver by resident macrophages and liver sinusoidal endothelial cells which also express high level of the receptor [192].

TAMs also overexpress folate receptor beta (FRβ), which can be targeted using its ligand, folic acid [193][194]. To improve selectivity over folate receptor alpha (FRα), reduced folate carrier (RFC), and proton-coupled folate transporter (PCFT) which are expressed on both cancerous and non-cancerous tissues, various antifolate analogs have been developed a few of which exhibit significant FRβ selectivity over the rest [195]. Anti-FRβ Fv is also successfully used to selectively target FRβ+ TAMs [196]. While expression of FRβ is more restricted than FRα, several studies have reported that FRβ is also expressed on pro-inflammatory activated M1 macrophages [197][198][199].

Legumain and transferrin receptor are two other target receptors overexpressed on M2-TAMs compared to M1-TAMs but that are also present on several cancer cell types [200][201][202]. The proteolytic activity of legumain specific to the Ala-Ala-Asn substrate sequence has been used in the design of pro-drugs or smart drug delivery systems that target both TAMs and cancer cells [200][203][204]. Although many drug delivery systems targeting transferrin receptor have been reported, to our knowledge, all these systems are tailored toward targeting cancer cells expressing the receptor or enhancing transcytosis across blood brain barrier, and neither the effect on nor the application to affect TAMs has been reported [205][206][207].

In order to improve TAM selectivity over resident macrophages, a few groups have employed unique physical properties of TME to rationally design their targeting systems [203][190][208]. As an example, Zhu et al. reported the development of mannosylated PLGA nanoparticles that are masked with acid-sensitive PEG(2000) that prevents recognition by resident macrophages but is cleaved in the acidic TME to expose mannose for intratumoral TAM targeting [190]. In another study, Movahedi and Schoonooghe et al. demonstrated the use of excess unlabeled bivalent anti-MMR nanobody as a decoy to reduce background signal of 99mTc-labeled monovalent anti-MMR nanobody in liver and spleen [208]. The bivalent nanobody competitively binds to resident macrophages with higher avidity over the monovalent nanobody but has poor tumor penetration due to its bigger size, whereas the monovalent nanobody can diffuse into tumor to label M2-TAMs for imaging.

On our part, we have developed an M2 macrophage-targeting peptide (M2pep) via peptide phage display screening [209]. The peptide binds to M2 macrophages and M2-TAMs selectively over M1 and M0 macrophages as well as over other leukocytes. Further optimization of the peptide improves its serum stability and affinity, and enables enhanced targeting to M2-TAMs in syngeneic CT26 colon cancer and 4T1 breast cancer models [210][211]. Multivalent display of the peptide further improves binding avidity to M2 macrophages[212]. The small size and easy production of M2pep make it a promising and attractive targeting ligand for developing M2-TAM-targeted therapeutics.

5 Macrophages as a therapeutic depot

Macrophages receive considerable interest as a drug delivery carrier due to their tumor-homing property, high phagocytic capability of drug-loaded nanoparticles, and ability to directly kill cancer cells given the right immunostimulating environment [213]. For examples, peritoneal macrophages may be harvested, incubated with drugs, typically in nanoparticle/liposome formulations, for loading, and then infused back to animals/patients [214][215], or nanoparticles with appropriate size and/or ligand may be directly injected in vivo and get uptaken by resident macrophages/TAMs in the body for subsequent sustained release [216][217][188]. While both strategies significantly improve the circulation half-life of the drugs, the long-term survival of the macrophage host is limited by toxicity of the drug, and hence, most of the drugs are loaded in the nanoparticle or liposome formulations to reduce acute toxicity to the macrophage carrier [214,216]. Interestingly, Tanei et al. observed increased expression of p-glycoprotein efflux pump in albumin-bound paclitaxel nanoparticle-loaded macrophages compared to the unloaded control and attributed this as one of the possible mechanisms to preserve macrophage viability as well as to enable drug release to the surrounding e.g. TME [188]. In the case where drug is not toxic to macrophages, appropriate formulation enables sustained release of the loaded drug from macrophages for up to at least 2 weeks as demonstrated by Dou et al. for indinavir nanoparticle-loaded macrophages [218][219]. With proper nanoparticle encapsulation strategies to achieve intracellular stability, it is even possible to load biologics such as proteins into macrophages [220,221]. Equally important to effective drug loading is the ability of drug-laden macrophages to migrate to tumor. A study by Chang and Guo et al. shows that the size of nanoparticles internalized by macrophages can significantly alter locomotivity of the macrophages, with smaller nanoparticles (30 and 50 nm) having higher uptake into macrophages than larger nanoparticles (100 and 500 nm) but also more readily retarding the migration rate of the macrophages; this study suggests that 100 nm nanoparticles provide a good balance for effective drug loading and macrophage migration [174]. To increase control over drug activity from macrophage carriers, macrophage have been loaded with temperature-responsive liposomes for triggered release [216], with gold-silica nanoshells for photothermal therapy [215], and with iron oxide nanoparticles for dual tracking and trafficking using electromagnetic actuation [214][222][223].

Recently, an additional role of TAMs as a processing factory for antibody-drug conjugates (ADCs) has been reported [224]. Specifically, Li et al. observed an anti-tumor activity of non-targeted humanized IgG conjugated to protease-cleavable monomethyl auristatin E (hIgG-vaMMAE) in L-82 anaplastic large cell lymphoma and MCF-7 breast cancer xenograft models and further demonstrated, in a panel of xenograft models, that the efficacy is positively correlated with the extent of F4/80+ TAM infiltrate. Mechanistic investigation revealed the two key parameters for the TAM-mediated, antigen-independent, anti-tumor activity; (1) binding and internalization of ADCs to FcγR on TAMs for processing and release of drug and (2) membrane permeability of drug to mediate the by-stander effect once released from TAMs. This report provides the first evidence on the possible contribution of TAMs on efficacy of ADCs and presents an underexplored opportunity of TAM-targeted ADC engineering.

Instead of loading drugs into macrophages, it is also possible to genetically engineer them to produce therapeutic proteins. As an example, Palma and Mazzieri et al. transduced hematopoietic/progenitor stem cells (HPSCs) with lentiviral vector encoding Ifna1 under control of Tie2 promoter/enhancer elements. When transplanted into tumor-bearing mice, a subpopulation of these cells matured into TEMs and homed to the tumor where their TIE2 expression was further heightened resulting in localized production of IFNα that provided an anti-tumor effect and without systemic toxicity [225].

Since macrophages may exhibit both pro-tumoral and anti-tumoral activities in TME, it is important to fully understand the in vivo fate/functional state of these macrophages as well as ideally be able to promote and maintain the anti-tumor functional state of the macrophage carrier in order to maximize this cell-directed therapy. Although the early days of research were devoted to loading macrophages with cytotoxic drugs, it should be possible to load drugs/nanoparticles that may enhance in vivo anti-tumor properties of these macrophages or other immune cells. In light with the recent report on M1-inducing property of clinically approved iron oxide nanoparticles [145], it would be interesting to study how the iron oxide nanoparticle-loaded macrophages, already developed for diagnostic purpose, may exhibit enhanced anti-tumor properties. Finally, further development of the technology for delivery of biologics such as peptide or proteins drugs would greatly expand the utility of this technology in cancer therapy.

6 Perspectives on TAM-targeted therapeutics

With deeper understanding of cancer immunology, diverse strategies for modulation of TAMs are being uncovered and explored for therapeutic applications. Due to the complexity of tumors, combination therapy is typically needed to maximize an anti-tumor response. Thus, a clear understanding of the modes of drug action as well as mechanisms of resistance is needed in order to design an efficacious combination therapy that minimizes antagonistic effects. For example, identification of PI3k up-regulation in tumor as a resistance mechanism for CSF-1R kinase inhibitor in recurrent glioma suggests that a combination therapy between PI3k and CSF-1R inhibitors could be more beneficial [72]. Conversely, the therapeutics aiming to block macrophage recruitment signals (e.g. CCL2 or CXCL12 inhibition) may not be compatible with the ones that require the presence of macrophages for anti-tumor actions (e.g. anti-CD40 antibody) [118]. In congruence with the well-appreciated immunosuppressive roles of TAMs, a consensus was observed regarding the potential benefits of TAM-targeted therapies in potentiating immune checkpoint blockade therapies (anti PD-1/PD-L1/CTLA-4 antibodies) as evidenced in the race among pharmaceutical companies to investigate such combination therapies (Table 2). Improvement in gene sequencing and analysis technologies greatly facilitates the adoption of precision medicine where patients could be examined for genetic makeup and matched with appropriate therapeutic regimens. Documenting patients’ genetic profile and the corresponding therapeutic outcome are also beneficial in correlation studies to better predict patient response as well as in refining drug development. In the case of CSF-1R inhibition therapy, certain single nucleotide polymorphisms (SNPs) in CSF-1R have been identified that reduce the potency of emactuzumab [226]. Nonetheless, the study may help in the future design of the next-generation CSF-1R blockade therapy. To reap the benefit of a steady rise in molecularly targeted therapies that are promising for clinical translation, it is more than ever important to be resource-efficient. This may be possible through careful validation of pre-clinical studies and innovative design of clinical trials as seen, for example, in the I-SPY 2 trial (NCT01042379) [227]. With numerous therapeutic targets being identified and drug candidates being explored for modulation of TAMs, drug delivery technologies will soon come into play to further enhance therapeutic efficacy of these drugs, for example, by improving pharmacokinetics, stability, selectivity, or intracellular delivery while limiting systemic toxicity. Together with advancement in gene-editing technology, effective silencing of genes that promote pro-tumoral functions of TAMs (e.g. STAT3, SIRPα, PI3k, or Gpr132) may one day be a practicable therapeutic option. Finally, a cost barrier is another factor that could impede the clinical translation especially when multiple antibodies are used in a combination therapy as currently investigated in several trials. The problem may be alleviated with improvement in manufacturing efficiency or development of cheaper alternatives such as small molecule drugs or peptide analogs.

The last century of research has revealed that the mononuclear phagocyte system and macrophages play an essential role in normal physiological processes and disease development. Targeting TAMs has proven to be a promising strategy as TAM targeting agents are rapidly advancing to the clinic, both in combination with traditional therapeutics and with other immunomodulatory agents. With the increasing depth of TAM targeting agents within pre-clinical development and new research developments in understanding mechanistic TAM pathways, it is clear that TAM targeted therapies will be an important addition to the anti-cancer armamentarium.

Supplementary Material

supplement

Acknowledgments

This work was supported by NIH 1R01CA177272. Chayanon Ngambenjawong was supported by an Anandamahidol Foundation Fellowship. Heather H. Gustafson was supported by the Cardiovascular Pathology Training Grant (5 T32 HL 007312-37).

Footnotes

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.Hume DA. The mononuclear phagocyte system. Curr Opin Immunol. 2006;18:49–53. doi: 10.1016/j.coi.2005.11.008. [DOI] [PubMed] [Google Scholar]
  • 2.Mass E, Ballesteros I, Farlik M, Halbritter F, Gunther P, Crozet L, et al. Specification of tissue-resident macrophages during organogenesis. Science. 2016;353 doi: 10.1126/science.aaf4238. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Schulz C, Perdiguero E Gomez, Chorro L, Szabo-Rogers H, Cagnard N, Kierdorf K, et al. A lineage of myeloid cells independent of Myb and hematopoietic stem cells. Science. 2012;336:86–90. doi: 10.1126/science.1219179. [DOI] [PubMed] [Google Scholar]
  • 4.Wynn TA, Chawla A, Pollard JW. Macrophage biology in development, homeostasis and disease. Nature. 2013;496:445–455. doi: 10.1038/nature12034. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Ginhoux F, Jung S. Monocytes and macrophages: developmental pathways and tissue homeostasis. Nat Rev Immunol. 2014;14:392–404. doi: 10.1038/nri3671. [DOI] [PubMed] [Google Scholar]
  • 6.Lewis CE, Harney AS, Pollard JW. The Multifaceted Role of Perivascular Macrophages in Tumors. Cancer Cell. 2016;30365 doi: 10.1016/j.ccell.2016.07.009. [DOI] [PubMed] [Google Scholar]
  • 7.Mantovani A, Marchesi F, Malesci A, Laghi L, Allavena P. Tumour-associated macrophages as treatment targets in oncology. Nat Rev Clin Oncol. 2017 doi: 10.1038/nrclinonc.2016.217. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Mechnikov II. Immunity in Infective Diseases. Rev Infect Dis. 1988;10:223–227. http://dx.doi.org/10.1093/clinids/10.1.223. [PubMed] [Google Scholar]
  • 9.Paolicelli RC, Bolasco G, Pagani F, Maggi L, Scianni M, Panzanelli P, et al. Synaptic pruning by microglia is necessary for normal brain development. Science. 2011;333:1456–1458. doi: 10.1126/science.1202529. [DOI] [PubMed] [Google Scholar]
  • 10.Nayak D, Roth TL, McGavern DB. Microglia development and function. Annu Rev Immunol. 2014;32:367–402. doi: 10.1146/annurev-immunol-032713-120240. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Ganz T. Macrophages and systemic iron homeostasis. J Innate Immun. 2012;4:446–453. doi: 10.1159/000336423. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Hvidberg V, Maniecki MB, Jacobsen C, Hojrup P, Moller HJ, Moestrup SK. Identification of the receptor scavenging hemopexin-heme complexes. Blood. 2005;106:2572–2579. doi: 10.1182/blood-2005-03-1185. [DOI] [PubMed] [Google Scholar]
  • 13.Kohyama M, Ise W, Edelson BT, Wilker PR, Hildner K, Mejia C, et al. Role for Spi-C in the development of red pulp macrophages and splenic iron homeostasis. Nature. 2009;457:318–321. doi: 10.1038/nature07472. http://dx.doi.org/10.1038/nature07472. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Merad M, Manz MG, Karsunky H, Wagers A, Peters W, Charo I, et al. Langerhans cells renew in the skin throughout life under steady-state conditions. Nat Immunol. 2002;3:1135–1141. doi: 10.1038/ni852. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Hussell T, Bell TJ. Alveolar macrophages: plasticity in a tissue-specific context. Nat Rev Immunol. 2014;14:81–93. doi: 10.1038/nri3600. [DOI] [PubMed] [Google Scholar]
  • 16.Stockand JD, Sansom SC. Glomerular mesangial cells: electrophysiology and regulation of contraction. Physiol Rev. 1998;78:723–744. doi: 10.1152/physrev.1998.78.3.723. [DOI] [PubMed] [Google Scholar]
  • 17.Ajami B, Bennett JL, Krieger C, McNagny KM, Rossi FMV. Infiltrating monocytes trigger EAE progression, but do not contribute to the resident microglia pool. Nat Neurosci. 2011;14:1142–1149. doi: 10.1038/nn.2887. [DOI] [PubMed] [Google Scholar]
  • 18.Brown MB, von Chamier M, Allam AB, Reyes L. M1/M2 macrophage polarity in normal and complicated pregnancy. Front Immunol. 2014;5:606. doi: 10.3389/fimmu.2014.00606. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Renaud SJ, Graham CH. The role of macrophages in utero-placental interactions during normal and pathological pregnancy. Immunol Invest. 2008;37:535–564. doi: 10.1080/08820130802191375. [DOI] [PubMed] [Google Scholar]
  • 20.Davies LC, Jenkins SJ, Allen JE, Taylor PR. Tissue-resident macrophages. Nat Immunol. 2013;14:986–995. doi: 10.1038/ni.2705. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Lavin Y, Mortha A, Rahman A, Merad M. Regulation of macrophage development and function in peripheral tissues. Nat Rev Immunol. 2015;15:731–744. doi: 10.1038/nri3920. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Lavin Y, Winter D, Blecher-Gonen R, David E, Keren-Shaul H, Merad M, et al. Tissue-Resident Macrophage Enhancer Landscapes Are Shaped by the Local Microenvironment. Cell. 2014;159:1312–1326. doi: 10.1016/j.cell.2014.11.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Okabe Y, Medzhitov R. Tissue-specific signals control reversible program of localization and functional polarization of macrophages. Cell. 2014;157:832–844. doi: 10.1016/j.cell.2014.04.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Kawai T, Akira S. The role of pattern-recognition receptors in innate immunity: update on Toll-like receptors. Nat Immunol. 2010;11:373–384. doi: 10.1038/ni.1863. [DOI] [PubMed] [Google Scholar]
  • 25.Takeda K, Akira S. TLR signaling pathways. Semin Immunol. 2004;16:3–9. doi: 10.1016/j.smim.2003.10.003. [DOI] [PubMed] [Google Scholar]
  • 26.Malyshev I, Malyshev Y. Current Concept and Update of the Macrophage Plasticity Concept: Intracellular Mechanisms of Reprogramming and M3 Macrophage “Switch” Phenotype. Biomed Res Int. 2015;2015:341308. doi: 10.1155/2015/341308. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Takeda K, Tanaka T, Shi W, Matsumoto M, Minami M, Kashiwamura S, et al. Essential role of Stat6 in IL-4 signalling. Nature. 1996;380:627–630. doi: 10.1038/380627a0. [DOI] [PubMed] [Google Scholar]
  • 28.Gordon S, Martinez FO. Alternative activation of macrophages: mechanism and functions. Immunity. 2010;32:593–604. doi: 10.1016/j.immuni.2010.05.007. [DOI] [PubMed] [Google Scholar]
  • 29.Martinez FO, Helming L, Gordon S. Alternative activation of macrophages: an immunologic functional perspective. Annu Rev Immunol. 2009;27:451–483. doi: 10.1146/annurev.immunol.021908.132532. [DOI] [PubMed] [Google Scholar]
  • 30.Van Dyken SJ, Locksley RM. Interleukin-4- and interleukin-13-mediated alternatively activated macrophages: roles in homeostasis and disease. Annu Rev Immunol. 2013;31:317–343. doi: 10.1146/annurev-immunol-032712-095906. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Italiani P, Boraschi D. From Monocytes to M1/M2 Macrophages: Phenotypical vs. Functional Differentiation. Front Immunol. 2014;5:514. doi: 10.3389/fimmu.2014.00514. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Murray PJ, Allen JE, Biswas SK, Fisher EA, Gilroy DW, Goerdt S, et al. Macrophage activation and polarization: nomenclature and experimental guidelines. Immunity. 2014;41:14–20. doi: 10.1016/j.immuni.2014.06.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Martinez FO, Gordon S. The M1 and M2 paradigm of macrophage activation: time for reassessment. F1000Prime Rep. 2014;6:13. doi: 10.12703/P6-13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Zhou D, Huang C, Lin Z, Zhan S, Kong L, Fang C, et al. Macrophage polarization and function with emphasis on the evolving roles of coordinated regulation of cellular signaling pathways. Cell Signal. 2014;26:192–197. doi: 10.1016/j.cellsig.2013.11.004. [DOI] [PubMed] [Google Scholar]
  • 35.Elinav E, Nowarski R, Thaiss CA, Hu B, Jin C, Flavell RA. Inflammation-induced cancer: crosstalk between tumours, immune cells and microorganisms. Nat Rev Cancer. 2013;13:759–771. doi: 10.1038/nrc3611. http://dx.doi.org/10.1038/nrc3611. [DOI] [PubMed] [Google Scholar]
  • 36.Evans R, Alexander P. Cooperation of immune lymphoid cells with macrophages in tumour immunity. Nature. 1970;228:620–622. doi: 10.1038/228620a0. [DOI] [PubMed] [Google Scholar]
  • 37.Wang HW, Joyce JA. Alternative activation of tumor-associated macrophages by IL-4: priming for protumoral functions. Cell Cycle. 2010;9:4824–4835. doi: 10.4161/cc.9.24.14322. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Quail DF, Joyce JA. Microenvironmental regulation of tumor progression and metastasis. Nat Med. 2013;19:1423–1437. doi: 10.1038/nm.3394. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Caux C, Ramos RN, Prendergast GC, Bendriss-Vermare N, Ménétrier-Caux C. A Milestone Review on How Macrophages Affect Tumor Growth. Cancer Res. 2016;76:6439. doi: 10.1158/0008-5472.CAN-16-2631. LP-6442. http://cancerres.aacrjournals.org/content/76/22/6439.abstract. [DOI] [PubMed] [Google Scholar]
  • 40.Ostuni R, Kratochvill F, Murray PJ, Natoli G. Macrophages and cancer: from mechanisms to therapeutic implications. Trends Immunol. 2017;36:229–239. doi: 10.1016/j.it.2015.02.004. [DOI] [PubMed] [Google Scholar]
  • 41.Quail DF, Joyce JA. Molecular Pathways: Deciphering Mechanisms of Resistance to Macrophage-Targeted Therapies. Clin Cancer Res. 2016 doi: 10.1158/1078-0432.CCR-16-0133. http://clincancerres.aacrjournals.org/content/early/2016/11/24/1078-0432.CCR-16-0133.abstract. [DOI] [PMC free article] [PubMed]
  • 42.Yuan ZY, Luo RZ, Peng RJ, Wang SS, Xue C. High infiltration of tumor-associated macrophages in triple-negative breast cancer is associated with a higher risk of distant metastasis. Onco Targets Ther. 2014;7:1475–1480. doi: 10.2147/OTT.S61838. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Ruffell B, Affara NI, Coussens LM. Differential macrophage programming in the tumor microenvironment. Trends Immunol. 2012;33:119–126. doi: 10.1016/j.it.2011.12.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Ma J, Liu L, Che G, Yu N, Dai F, You Z. The M1 form of tumor-associated macrophages in non-small cell lung cancer is positively associated with survival time. BMC Cancer. 2010;10:112. doi: 10.1186/1471-2407-10-112. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Mei J, Xiao Z, Guo C, Pu Q, Ma L, Liu C, et al. Prognostic impact of tumor-associated macrophage infiltration in non-small cell lung cancer: A systemic review and meta-analysis. Oncotarget. 2016;7:34217–34228. doi: 10.18632/oncotarget.9079. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Gu L, Li H, Chen L, Ma X, Li X, Gao Y, et al. Prognostic role of lymphocyte to monocyte ratio for patients with cancer: evidence from a systematic review and meta-analysis. Oncotarget. 2016;7:31926–31942. doi: 10.18632/oncotarget.7876. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Drake MT, Clarke BL, Khosla S. Bisphosphonates: Mechanism of Action and Role in Clinical Practice. Mayo Clin Proc. 2008;83:1032–1045. doi: 10.4065/83.9.1032. http://www.ncbi.nlm.nih.gov/pmc/articles/PMC2667901/ [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Roelofs AJ, Thompson K, Gordon S, Rogers MJ. Molecular mechanisms of action of bisphosphonates: current status. Clin Cancer Res. 2006;12:6222s–6230s. doi: 10.1158/1078-0432.CCR-06-0843. [DOI] [PubMed] [Google Scholar]
  • 49.Junankar S, Shay G, Jurczyluk J, Ali N, Down J, Pocock N, et al. Real-time intravital imaging establishes tumor-associated macrophages as the extraskeletal target of bisphosphonate action in cancer. Cancer Discov. 2015;5:35–42. doi: 10.1158/2159-8290.CD-14-0621. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Coscia M, Quaglino E, Iezzi M, Curcio C, Pantaleoni F, Riganti C, et al. Zoledronic acid repolarizes tumour-associated macrophages and inhibits mammary carcinogenesis by targeting the mevalonate pathway. J Cell Mol Med. 2010;14:2803–2815. doi: 10.1111/j.1582-4934.2009.00926.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Zeisberger SM, Odermatt B, Marty C, Zehnder-Fjallman AHM, Ballmer-Hofer K, Schwendener RA. Clodronate-liposome-mediated depletion of tumour-associated macrophages: a new and highly effective antiangiogenic therapy approach. Br J Cancer. 2006;95:272–281. doi: 10.1038/sj.bjc.6603240. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Rogers TL, Holen I. Tumour macrophages as potential targets of bisphosphonates. J Transl Med. 2011;9:177. doi: 10.1186/1479-5876-9-177. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Zhang W, Zhu XD, Sun HC, Xiong YQ, Zhuang PY, Xu HX, et al. Depletion of tumor-associated macrophages enhances the effect of sorafenib in metastatic liver cancer models by antimetastatic and antiangiogenic effects. Clin Cancer Res. 2010;16:3420–3430. doi: 10.1158/1078-0432.CCR-09-2904. [DOI] [PubMed] [Google Scholar]
  • 54.Hattori Y, Shibuya K, Kojima K, Miatmoko A, Kawano K, Ozaki KI, et al. Zoledronic acid enhances antitumor efficacy of liposomal doxorubicin. Int J Oncol. 2015;47:211–219. doi: 10.3892/ijo.2015.2991. [DOI] [PubMed] [Google Scholar]
  • 55.Ohara Y, Oda T, Yamada K, Hashimoto S, Akashi Y, Miyamoto R, et al. Effective delivery of chemotherapeutic nanoparticles by depleting host Kupffer cells. Int J Cancer. 2012;131:2402–2410. doi: 10.1002/ijc.27502. [DOI] [PubMed] [Google Scholar]
  • 56.Moynihan KD, Opel CF, Szeto GL, Tzeng A, Zhu EF, Engreitz JM, et al. Eradication of large established tumors in mice by combination immunotherapy that engages innate and adaptive immune responses. Nat Med. 2016;22:1402–1410. doi: 10.1038/nm.4200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Chao MP, Alizadeh AA, Tang C, Myklebust JH, Varghese B, Gill S, et al. Anti-CD47 antibody synergizes with rituximab to promote phagocytosis and eradicate non-Hodgkin lymphoma. Cell. 2010;142:699–713. doi: 10.1016/j.cell.2010.07.044. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Lehmann B, Biburger M, Brückner C, Ipsen-Escobedo A, Gordan S, Lehmann C, et al. Tumor location determines tissue-specific recruitment of tumor-associated macrophages and antibody-dependent immunotherapy response. Sci Immunol. 2017;2 doi: 10.1126/sciimmunol.aah6413. http://immunology.sciencemag.org/content/2/7/eaah6413.abstract. [DOI] [PubMed] [Google Scholar]
  • 59.Pucci F, Garris C, Lai CP, Newton A, Pfirschke C, Engblom C, et al. SCS macrophages suppress melanoma by restricting tumor-derived vesicle-B cell interactions. Science. 2016;352:242–246. doi: 10.1126/science.aaf1328. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Ohnishi K, Komohara Y, Saito Y, Miyamoto Y, Watanabe M, Baba H, et al. CD169-positive macrophages in regional lymph nodes are associated with a favorable prognosis in patients with colorectal carcinoma. Cancer Sci. 2013;104:1237–1244. doi: 10.1111/cas.12212. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Shiota T, Miyasato Y, Ohnishi K, Yamamoto-Ibusuki M, Yamamoto Y, Iwase H, et al. The Clinical Significance of CD169-Positive Lymph Node Macrophage in Patients with Breast Cancer. PLoS One. 2016;11:e0166680. doi: 10.1371/journal.pone.0166680. http://dx.doi.org/10.1371%2Fjournal.pone.0166680. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Michaelson MD, Smith MR. Bisphosphonates for treatment and prevention of bone metastases. J Clin Oncol. 2005;23:8219–8224. doi: 10.1200/JCO.2005.02.9579. [DOI] [PubMed] [Google Scholar]
  • 63.http://www.fda.gov/NewsEvents/Newsroom/PressAnnouncements/ucm468832.htm
  • 64.Otero K, Turnbull IR, Poliani PL, Vermi W, Cerutti E, Aoshi T, et al. Macrophage colony-stimulating factor induces the proliferation and survival of macrophages via a pathway involving DAP12 and beta-catenin. Nat Immunol. 2009;10:734–743. doi: 10.1038/ni.1744. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Lin H, Lee E, Hestir K, Leo C, Huang M, Bosch E, et al. Discovery of a cytokine and its receptor by functional screening of the extracellular proteome. Science. 2008;320:807–811. doi: 10.1126/science.1154370. [DOI] [PubMed] [Google Scholar]
  • 66.Hume DA, MacDonald KPA. Therapeutic applications of macrophage colony-stimulating factor-1 (CSF-1) and antagonists of CSF-1 receptor (CSF-1R) signaling. Blood. 2012;119:1810–1820. doi: 10.1182/blood-2011-09-379214. [DOI] [PubMed] [Google Scholar]
  • 67.Lohela M, Casbon AJ, Olow A, Bonham L, Branstetter D, Weng N, et al. Intravital imaging reveals distinct responses of depleting dynamic tumor-associated macrophage and dendritic cell subpopulations. Proc Natl Acad Sci U S A. 2014;111:E5086–95. doi: 10.1073/pnas.1419899111. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Fend L, Accart N, Kintz J, Cochin S, Reymann C, Le Pogam F, et al. Therapeutic effects of anti-CD115 monoclonal antibody in mouse cancer models through dual inhibition of tumor-associated macrophages and osteoclasts. PLoS One. 2013;8:e73310. doi: 10.1371/journal.pone.0073310. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.MacDonald KPA, Palmer JS, Cronau S, Seppanen E, Olver S, Raffelt NC, et al. An antibody against the colony-stimulating factor 1 receptor depletes the resident subset of monocytes and tissue- and tumor-associated macrophages but does not inhibit inflammation. Blood. 2010;116:3955–3963. doi: 10.1182/blood-2010-02-266296. [DOI] [PubMed] [Google Scholar]
  • 70.Pyonteck SM, Akkari L, Schuhmacher AJ, Bowman RL, Sevenich L, Quail DF, et al. CSF-1R inhibition alters macrophage polarization and blocks glioma progression. Nat Med. 2013;19:1264–1272. doi: 10.1038/nm.3337. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Strachan DC, Ruffell B, Oei Y, Bissell MJ, Coussens LM, Pryer N, et al. CSF1R inhibition delays cervical and mammary tumor growth in murine models by attenuating the turnover of tumor-associated macrophages and enhancing infiltration by CD8+ T cells. Oncoimmunology. 2013;2:e26968. doi: 10.4161/onci.26968. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Quail DF, Bowman RL, Akkari L, Quick ML, Schuhmacher AJ, Huse JT, et al. The tumor microenvironment underlies acquired resistance to CSF-1R inhibition in gliomas. Science. 2016;352:aad3018. doi: 10.1126/science.aad3018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Jeong W, Doroshow JH, Kummar S. US FDA Approved Oral Kinase Inhibitors for the Treatment of Malignancies. Curr Probl Cancer. 2013;37:110–144. doi: 10.1016/j.currproblcancer.2013.06.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Broekman F, Giovannetti E, Peters GJ. Tyrosine kinase inhibitors: Multi-targeted or single-targeted? World J Clin Oncol. 2011;2:80–93. doi: 10.5306/wjco.v2.i2.80. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Kogan M, Fischer-Smith T, Kaminsky R, Lehmicke G, Rappaport J. CSF-1R up-regulation is associated with response to pharmacotherapy targeting tyrosine kinase activity in AML cell lines. Anticancer Res. 2012;32:893–899. http://www.ncbi.nlm.nih.gov/pmc/articles/PMC3601026/ [PMC free article] [PubMed] [Google Scholar]
  • 76.Brownlow N, Russell AE, Saravanapavan H, Wiesmann M, Murray JM, Manley PW, et al. Comparison of nilotinib and imatinib inhibition of FMS receptor signaling, macrophage production and osteoclastogenesis. Leukemia. 2007;22:649–652. doi: 10.1038/sj.leu.2404944. http://dx.doi.org/10.1038/sj.leu.2404944. [DOI] [PubMed] [Google Scholar]
  • 77.Brownlow N, Mol C, Hayford C, Ghaem-Maghami S, Dibb NJ. Dasatinib is a potent inhibitor of tumour-associated macrophages, osteoclasts and the FMS receptor. Leukemia. 2008;23:590–594. doi: 10.1038/leu.2008.237. http://dx.doi.org/10.1038/leu.2008.237. [DOI] [PubMed] [Google Scholar]
  • 78.Tap WD, Wainberg ZA, Anthony SP, Ibrahim PN, Zhang C, Healey JH, et al. Structure-Guided Blockade of CSF1R Kinase in Tenosynovial Giant-Cell Tumor. N Engl J Med. 2015;373:428–437. doi: 10.1056/NEJMoa1411366. [DOI] [PubMed] [Google Scholar]
  • 79.Chaudhuri A. Regulation of Macrophage Polarization by RON Receptor Tyrosine Kinase Signaling. Front Immunol. 2014;5:546. doi: 10.3389/fimmu.2014.00546. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Lutz MA, Correll PH. Activation of CR3-mediated phagocytosis by MSP requires the RON receptor, tyrosine kinase activity, phosphatidylinositol 3-kinase, and protein kinase C zeta. J Leukoc Biol. 2003;73:802–814. doi: 10.1189/jlb.0602319. [DOI] [PubMed] [Google Scholar]
  • 81.Sharda DR, Yu S, Ray M, Squadrito ML, De Palma M, Wynn TA, et al. Regulation of macrophage arginase expression and tumor growth by the Ron receptor tyrosine kinase. J Immunol. 2011;187:2181–2192. doi: 10.4049/jimmunol.1003460. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Morrison AC, Wilson CB, Ray M, Correll PH. Macrophage-stimulating protein, the ligand for the stem cell-derived tyrosine kinase/RON receptor tyrosine kinase, inhibits IL-12 production by primary peritoneal macrophages stimulated with IFN-gamma and lipopolysaccharide. J Immunol. 2004;172:1825–1832. doi: 10.4049/jimmunol.172.3.1825. [DOI] [PubMed] [Google Scholar]
  • 83.Chen YQ, Fisher JH, Wang MH. Activation of the RON receptor tyrosine kinase inhibits inducible nitric oxide synthase (iNOS) expression by murine peritoneal exudate macrophages: phosphatidylinositol-3 kinase is required for RON-mediated inhibition of iNOS expression. J Immunol. 1998;161:4950–4959. [PubMed] [Google Scholar]
  • 84.Gurusamy D, Gray JK, Pathrose P, Kulkarni RM, Finkleman FD, Waltz SE. Myeloid-Specific Expression of Ron Receptor Kinase Promotes Prostate Tumor Growth. Cancer Res. 2013;73:1752–1763. doi: 10.1158/0008-5472.CAN-12-2474. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Eyob H, Ekiz HA, DeRose YS, Waltz SE, Williams MA, Welm AL. Inhibition of Ron kinase blocks conversion of micrometastases to overt metastases by boosting anti-tumor immunity. Cancer Discov. 2013;3:751–760. doi: 10.1158/2159-8290.CD-12-0480. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Bieniasz M, Radhakrishnan P, Faham N, De La O J-P, Welm AL. Preclinical Efficacy of Ron Kinase Inhibitors Alone and in Combination with PI3K Inhibitors for Treatment of sfRon-Expressing Breast Cancer Patient-Derived Xenografts. Clin Cancer Res. 2015;21:5588–5600. doi: 10.1158/1078-0432.CCR-14-3283. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Maira SM, Pecchi S, Huang A, Burger M, Knapp M, Sterker D, et al. Identification and characterization of NVP-BKM120, an orally available pan-class I PI3-kinase inhibitor. Mol Cancer Ther. 2012;11:317–328. doi: 10.1158/1535-7163.MCT-11-0474. [DOI] [PubMed] [Google Scholar]
  • 88.Lavin Y, Merad M. Macrophages: gatekeepers of tissue integrity. Cancer Immunol Res. 2013;1:201–209. doi: 10.1158/2326-6066.CIR-13-0117. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Gul N, van Egmond M. Antibody-Dependent Phagocytosis of Tumor Cells by Macrophages: A Potent Effector Mechanism of Monoclonal Antibody Therapy of Cancer. Cancer Res. 2015;75:5008–5013. doi: 10.1158/0008-5472.CAN-15-1330. [DOI] [PubMed] [Google Scholar]
  • 90.Tipton TRW, Roghanian A, Oldham RJ, Carter MJ, Cox KL, Mockridge CI, et al. Antigenic modulation limits the effector cell mechanisms employed by type I anti-CD20 monoclonal antibodies. Blood. 2015;125:1901–1909. doi: 10.1182/blood-2014-07-588376. [DOI] [PubMed] [Google Scholar]
  • 91.Shi Y, Fan X, Deng H, Brezski RJ, Rycyzyn M, Jordan RE, et al. Trastuzumab triggers phagocytic killing of high HER2 cancer cells in vitro and in vivo by interaction with Fcgamma receptors on macrophages. J Immunol. 2015;194:4379–4386. doi: 10.4049/jimmunol.1402891. [DOI] [PubMed] [Google Scholar]
  • 92.Grugan KD, McCabe FL, Kinder M, Greenplate AR, Harman BC, Ekert JE, et al. Tumor-associated macrophages promote invasion while retaining Fc-dependent anti-tumor function. J Immunol. 2012;189:5457–5466. doi: 10.4049/jimmunol.1201889. [DOI] [PubMed] [Google Scholar]
  • 93.Taylor RP, Lindorfer MA. Analyses of CD20 monoclonal antibody-mediated tumor cell killing mechanisms: rational design of dosing strategies. Mol Pharmacol. 2014;86:485–491. doi: 10.1124/mol.114.092684. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Velmurugan R, Challa DK, Ram S, Ober RJ, Ward ES. Macrophage-Mediated Trogocytosis Leads to Death of Antibody-Opsonized Tumor Cells. Mol Cancer Ther. 2016;15:1879–1889. doi: 10.1158/1535-7163.MCT-15-0335. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Richards JO, Karki S, Lazar GA, Chen H, Dang W, Desjarlais JR. Optimization of antibody binding to FcgammaRIIa enhances macrophage phagocytosis of tumor cells. Mol Cancer Ther. 2008;7:2517–2527. doi: 10.1158/1535-7163.MCT-08-0201. [DOI] [PubMed] [Google Scholar]
  • 96.Brandsma AM, Ten Broeke T, Nederend M, Meulenbroek LAPM, van Tetering G, Meyer S, et al. Simultaneous Targeting of FcgammaRs and FcalphaRI Enhances Tumor Cell Killing. Cancer Immunol Res. 2015;3:1316–1324. doi: 10.1158/2326-6066.CIR-15-0099-T. [DOI] [PubMed] [Google Scholar]
  • 97.Josephs DH, Bax HJ, Dodev T, Georgouli M, Nakamura M, Pellizzari G, et al. Anti-folate receptor-α IgE but not IgG recruits macrophages to attack tumors via TNF-α/MCP-1 signaling. Cancer Res. 2017 doi: 10.1158/0008-5472.CAN-16-1829. http://cancerres.aacrjournals.org/content/early/2017/01/14/0008-5472.CAN-16-1829.abstract. [DOI] [PMC free article] [PubMed]
  • 98.Willingham SB, Volkmer JP, Gentles AJ, Sahoo D, Dalerba P, Mitra SS, et al. The CD47-signal regulatory protein alpha (SIRPa) interaction is a therapeutic target for human solid tumors. Proc Natl Acad Sci U S A. 2012;109:6662–6667. doi: 10.1073/pnas.1121623109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Weiskopf K, Ring AM, Ho CCM, Volkmer JP, Levin AM, Volkmer AK, et al. Engineered SIRPalpha variants as immunotherapeutic adjuvants to anticancer antibodies. Science. 2013;341:88–91. doi: 10.1126/science.1238856. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Liu J, Wang L, Zhao F, Tseng S, Narayanan C, Shura L, et al. Pre-Clinical Development of a Humanized Anti-CD47 Antibody with Anti-Cancer Therapeutic Potential. PLoS One. 2015;10:e0137345. doi: 10.1371/journal.pone.0137345. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Ho CCM, Guo N, Sockolosky JT, Ring AM, Weiskopf K, Ozkan E, et al. “Velcro” engineering of high affinity CD47 ectodomain as signal regulatory protein alpha (SIRPalpha) antagonists that enhance antibody-dependent cellular phagocytosis. J Biol Chem. 2015;290:12650–12663. doi: 10.1074/jbc.M115.648220. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Dheilly E, Moine V, Broyer L, Salgado-Pires S, Johnson Z, Papaioannou A, et al. Selective Blockade of the Ubiquitous Checkpoint Receptor CD47 Is Enabled by Dual-Targeting Bispecific Antibodies. Mol Ther. 2017 doi: 10.1016/j.ymthe.2016.11.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Weiskopf K, Anderson KL, Ito D, Schnorr PJ, Tomiyasu H, Ring AM, et al. Eradication of Canine Diffuse Large B-Cell Lymphoma in a Murine Xenograft Model with CD47 Blockade and Anti-CD20. Cancer Immunol Res. 2016;4:1072–1087. doi: 10.1158/2326-6066.CIR-16-0105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Franklin RA, Liao W, Sarkar A, Kim MV, Bivona MR, Liu K, et al. The cellular and molecular origin of tumor-associated macrophages. Science. 2014;344:921–925. doi: 10.1126/science.1252510. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Sanford DE, Belt BA, Panni RZ, Mayer A, Deshpande AD, Carpenter D, et al. Inflammatory monocyte mobilization decreases patient survival in pancreatic cancer: a role for targeting the CCL2/CCR2 axis. Clin Cancer Res. 2013;19:3404–3415. doi: 10.1158/1078-0432.CCR-13-0525. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Ren G, Zhao X, Wang Y, Zhang X, Chen X, Xu C, et al. CCR2-dependent recruitment of macrophages by tumor-educated mesenchymal stromal cells promotes tumor development and is mimicked by TNFalpha. Cell Stem Cell. 2012;11:812–824. doi: 10.1016/j.stem.2012.08.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Qian BZ, Li J, Zhang H, Kitamura T, Zhang J, Campion LR, et al. CCL2 recruits inflammatory monocytes to facilitate breast-tumour metastasis. Nature. 2011;475:222–225. doi: 10.1038/nature10138. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Engblom C, Pfirschke C, Pittet MJ. The role of myeloid cells in cancer therapies. Nat Rev Cancer. 2016;16:447–462. doi: 10.1038/nrc.2016.54. [DOI] [PubMed] [Google Scholar]
  • 109.Ji J, Eggert T, Budhu A, Forgues M, Takai A, Dang H, et al. Hepatic stellate cell and monocyte interaction contributes to poor prognosis in hepatocellular carcinoma. Hepatology. 2015;62:481–495. doi: 10.1002/hep.27822. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Lim SY, Yuzhalin AE, Gordon-Weeks AN, Muschel RJ. Targeting the CCL2-CCR2 signaling axis in cancer metastasis. Oncotarget. 2016;7:28697–28710. doi: 10.18632/oncotarget.7376. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Bonapace L, Coissieux MM, Wyckoff J, Mertz KD, Varga Z, Junt T, et al. Cessation of CCL2 inhibition accelerates breast cancer metastasis by promoting angiogenesis. Nature. 2014;515:130–133. doi: 10.1038/nature13862. [DOI] [PubMed] [Google Scholar]
  • 112.Kirk PS, Koreckij T, Nguyen HM, Brown LG, Snyder LA, Vessella RL, et al. Inhibition of CCL2 signaling in combination with docetaxel treatment has profound inhibitory effects on prostate cancer growth in bone. Int J Mol Sci. 2013;14:10483–10496. doi: 10.3390/ijms140510483. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Hughes R, Qian BZ, Rowan C, Muthana M, Keklikoglou I, Olson OC, et al. Perivascular M2 Macrophages Stimulate Tumor Relapse after Chemotherapy. Cancer Res. 2015;75:3479–3491. doi: 10.1158/0008-5472.CAN-14-3587. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Wang SC, Yu CF, Hong JH, Tsai CS, Chiang CS. Radiation therapy-induced tumor invasiveness is associated with SDF-1-regulated macrophage mobilization and vasculogenesis. PLoS One. 2013;8:e69182. doi: 10.1371/journal.pone.0069182. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Sanchez-Martin L, Estecha A, Samaniego R, Sanchez-Ramon S, Vega MA, Sanchez-Mateos P. The chemokine CXCL12 regulates monocyte-macrophage differentiation and RUNX3 expression. Blood. 2011;117:88–97. doi: 10.1182/blood-2009-12-258186. [DOI] [PubMed] [Google Scholar]
  • 116.Vonderheide RH, Glennie MJ. Agonistic CD40 antibodies and cancer therapy. Clin Cancer Res. 2013;19:1035–1043. doi: 10.1158/1078-0432.CCR-12-2064. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Beatty GL, Chiorean EG, Fishman MP, Saboury B, Teitelbaum UR, Sun W, et al. CD40 Agonists Alter Tumor Stroma and Show Efficacy Against Pancreatic Carcinoma in Mice and Humans. Science. 2011;331:1612–1616. doi: 10.1126/science.1198443. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Long KB, Gladney WL, Tooker GM, Graham K, Fraietta JA, Beatty GL. IFNgamma and CCL2 Cooperate to Redirect Tumor-Infiltrating Monocytes to Degrade Fibrosis and Enhance Chemotherapy Efficacy in Pancreatic Carcinoma. Cancer Discov. 2016;6:400–413. doi: 10.1158/2159-8290.CD-15-1032. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Li F, Ravetch JV. Inhibitory Fcgamma receptor engagement drives adjuvant and anti-tumor activities of agonistic CD40 antibodies. Science. 2011;333:1030–1034. doi: 10.1126/science.1206954. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.White AL, Chan HTC, Roghanian A, French RR, Mockridge CI, Tutt AL, et al. Interaction with FcgammaRIIB is critical for the agonistic activity of anti-CD40 monoclonal antibody. J Immunol. 2011;187:1754–1763. doi: 10.4049/jimmunol.1101135. [DOI] [PubMed] [Google Scholar]
  • 121.Mazzieri R, Pucci F, Moi D, Zonari E, Ranghetti A, Berti A, et al. Targeting the ANG2/TIE2 axis inhibits tumor growth and metastasis by impairing angiogenesis and disabling rebounds of proangiogenic myeloid cells. Cancer Cell. 2011;19:512–526. doi: 10.1016/j.ccr.2011.02.005. [DOI] [PubMed] [Google Scholar]
  • 122.Coffelt SB, Tal AO, Scholz A, De Palma M, Patel S, Urbich C, et al. Angiopoietin-2 regulates gene expression in TIE2-expressing monocytes and augments their inherent proangiogenic functions. Cancer Res. 2010;70:5270–5280. doi: 10.1158/0008-5472.CAN-10-0012. [DOI] [PubMed] [Google Scholar]
  • 123.Coutelle O, Schiffmann LM, Liwschitz M, Brunold M, Goede V, Hallek M, et al. Dual targeting of Angiopoetin-2 and VEGF potentiates effective vascular normalisation without inducing empty basement membrane sleeves in xenograft tumours. Br J Cancer. 2015;112:495–503. doi: 10.1038/bjc.2014.629. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Kloepper J, Riedemann L, Amoozgar Z, Seano G, Susek K, Yu V, et al. Ang-2/VEGF bispecific antibody reprograms macrophages and resident microglia to anti-tumor phenotype and prolongs glioblastoma survival. Proc Natl Acad Sci U S A. 2016;113:4476–4481. doi: 10.1073/pnas.1525360113. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.Jassar AS, Suzuki E, Kapoor V, Sun J, Silverberg MB, Cheung L, et al. Activation of tumor-associated macrophages by the vascular disrupting agent 5,6-dimethylxanthenone-4-acetic acid induces an effective CD8+ T-cell-mediated antitumor immune response in murine models of lung cancer and mesothelioma. Cancer Res. 2005;65:11752–11761. doi: 10.1158/0008-5472.CAN-05-1658. [DOI] [PubMed] [Google Scholar]
  • 126.Ghesquiere B, Wong BW, Kuchnio A, Carmeliet P. Metabolism of stromal and immune cells in health and disease. Nature. 2014;511:167–176. doi: 10.1038/nature13312. [DOI] [PubMed] [Google Scholar]
  • 127.Weichhart T, Hengstschlager M, Linke M. Regulation of innate immune cell function by mTOR. Nat Rev Immunol. 2015;15:599–614. doi: 10.1038/nri3901. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Rodrik-Outmezguine VS, Okaniwa M, Yao Z, Novotny CJ, McWhirter C, Banaji A, et al. Overcoming mTOR resistance mutations with a new-generation mTOR inhibitor. Nature. 2016;534:272–276. doi: 10.1038/nature17963. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129.Chen W, Ma T, Shen X, Xia X, Xu G, Bai X, et al. Macrophage-induced tumor angiogenesis is regulated by the TSC2-mTOR pathway. Cancer Res. 2012;72:1363–1372. doi: 10.1158/0008-5472.CAN-11-2684. [DOI] [PubMed] [Google Scholar]
  • 130.Kaneda MM, Messer KS, Ralainirina N, Li H, Leem CJ, Gorjestani S, et al. PI3Kγ is a molecular switch that controls immune suppression. Nature. 2016;539:437–442. doi: 10.1038/nature19834. http://dx.doi.org/10.1038/nature19834. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Arranz A, Doxaki C, Vergadi E, Martinez de la Torre Y, Vaporidi K, Lagoudaki ED, et al. Akt1 and Akt2 protein kinases differentially contribute to macrophage polarization. Proc Natl Acad Sci U S A. 2012;109:9517–9522. doi: 10.1073/pnas.1119038109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132.Sahin E, Haubenwallner S, Kuttke M, Kollmann I, Halfmann A, Dohnal AM, et al. Macrophage PTEN regulates expression and secretion of arginase I modulating innate and adaptive immune responses. J Immunol. 2014;193:1717–1727. doi: 10.4049/jimmunol.1302167. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133.Colegio OR, Chu N-Q, Szabo AL, Chu T, Rhebergen AM, Jairam V, et al. Functional polarization of tumour-associated macrophages by tumour-derived lactic acid. Nature. 2014;513:559–563. doi: 10.1038/nature13490. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134.Chen P, Zuo H, Xiong H, Kolar MJ, Chu Q, Saghatelian A, et al. Gpr132 sensing of lactate mediates tumor-macrophage interplay to promote breast cancer metastasis. Proc Natl Acad Sci U S A. 2017;114:580–585. doi: 10.1073/pnas.1614035114. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135.Cheng WY, Huynh H, Chen P, Pena-Llopis S, Wan Y. Macrophage PPARgamma inhibits Gpr132 to mediate the anti-tumor effects of rosiglitazone. Elife. 2016;5 doi: 10.7554/eLife.18501. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.Kusmartsev S, Gabrilovich DI. STAT1 signaling regulates tumor-associated macrophage-mediated T cell deletion. J Immunol. 2005;174:4880–4891. doi: 10.4049/jimmunol.174.8.4880. [DOI] [PubMed] [Google Scholar]
  • 137.Munn DH, Shafizadeh E, Attwood JT, Bondarev I, Pashine A, Mellor AL. Inhibition of T cell proliferation by macrophage tryptophan catabolism. J Exp Med. 1999;189:1363–1372. doi: 10.1084/jem.189.9.1363. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138.Beloribi-Djefaflia S, Vasseur S, Guillaumond F. Lipid metabolic reprogramming in cancer cells. Oncogenesis. 2016;5:e189. doi: 10.1038/oncsis.2015.49. http://dx.doi.org/10.1038/oncsis.2015.49. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139.Prima V, Kaliberova LN, Kaliberov S, Curiel DT, Kusmartsev S. COX2/mPGES1/PGE2 pathway regulates PD-L1 expression in tumor-associated macrophages and myeloid-derived suppressor cells. Proc Natl Acad Sci U S A. 2017 doi: 10.1073/pnas.1612920114. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140.Jung M, Oren B, Mora J, Mertens C, Dziumbla S, Popp R, et al. Lipocalin 2 from macrophages stimulated by tumor cell-derived sphingosine 1-phosphate promotes lymphangiogenesis and tumor metastasis. Sci Signal. 2016;9:ra64. doi: 10.1126/scisignal.aaf3241. [DOI] [PubMed] [Google Scholar]
  • 141.Nakanishi Y, Nakatsuji M, Seno H, Ishizu S, Akitake-Kawano R, Kanda K, et al. COX-2 inhibition alters the phenotype of tumor-associated macrophages from M2 to M1 in ApcMin/+ mouse polyps. Carcinogenesis. 2011;32:1333–1339. doi: 10.1093/carcin/bgr128. [DOI] [PubMed] [Google Scholar]
  • 142.Torti SV, Torti FM. Ironing out cancer. Cancer Res. 2011;71:1511–1514. doi: 10.1158/0008-5472.CAN-10-3614. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143.Torti SV, Torti FM. Iron and cancer: more ore to be mined. Nat Rev Cancer. 2013;13:342–355. doi: 10.1038/nrc3495. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144.Mertens C, Akam EA, Rehwald C, Brüne B, Tomat E, Jung M. Intracellular Iron Chelation Modulates the Macrophage Iron Phenotype with Consequences on Tumor Progression. PLoS One. 2016;11:e0166164. doi: 10.1371/journal.pone.0166164. http://dx.doi.org/10.1371%2Fjournal.pone.0166164. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145.Zanganeh S, Hutter G, Spitler R, Lenkov O, Mahmoudi M, Shaw A, et al. Iron oxide nanoparticles inhibit tumour growth by inducing pro-inflammatory macrophage polarization in tumour tissues. Nat Nanotechnol. 2016;11:986–994. doi: 10.1038/nnano.2016.168. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146.Bruns H, Buttner M, Fabri M, Mougiakakos D, Bittenbring JT, Hoffmann MH, et al. Vitamin D-dependent induction of cathelicidin in human macrophages results in cytotoxicity against high-grade B cell lymphoma. Sci Transl Med. 2015;7:282ra47. doi: 10.1126/scitranslmed.aaa3230. [DOI] [PubMed] [Google Scholar]
  • 147.Canton J, Neculai D, Grinstein S. Scavenger receptors in homeostasis and immunity. Nat Rev Immunol. 2013;13:621–634. doi: 10.1038/nri3515. http://dx.doi.org/10.1038/nri3515. [DOI] [PubMed] [Google Scholar]
  • 148.Yu X, Guo C, Fisher PB, Subjeck JR, Wang XY. Scavenger Receptors: Emerging Roles in Cancer Biology and Immunology. Adv Cancer Res. 2015;128:309–364. doi: 10.1016/bs.acr.2015.04.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 149.Georgoudaki AM, Prokopec KE, Boura VF, Hellqvist E, Sohn S, Ostling J, et al. Reprogramming Tumor-Associated Macrophages by Antibody Targeting Inhibits Cancer Progression and Metastasis. Cell Rep. 2016;15:2000–2011. doi: 10.1016/j.celrep.2016.04.084. [DOI] [PubMed] [Google Scholar]
  • 150.Neyen C, Plüddemann A, Mukhopadhyay S, Maniati E, Bossard M, Gordon S, et al. Macrophage Scavenger Receptor A promotes tumour progression in murine models of ovarian and pancreatic cancer. J Immunol. 2013;190:3798–3805. doi: 10.4049/jimmunol.1203194. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151.Neyen C, Mukhopadhyay S, Gordon S, Hagemann T. An apolipoprotein A-I mimetic targets scavenger receptor A on tumor-associated macrophages: A prospective anticancer treatment? Oncoimmunology. 2013;2:e24461. doi: 10.4161/onci.24461. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 152.Zhang H, Zhang W, Sun X, Dang R, Zhou R, Bai H, et al. Class A1 scavenger receptor modulates glioma progression by regulating M2-like tumor-associated macrophage polarization. Oncotarget. 2016;7:50099–50116. doi: 10.18632/oncotarget.10318. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 153.Cespedes MV, Guillen MJ, Lopez-Casas PP, Sarno F, Gallardo A, Alamo P, et al. Lurbinectedin induces depletion of tumor-associated macrophages, an essential component of its in vivo synergism with gemcitabine, in pancreatic adenocarcinoma mouse models. Dis Model Mech. 2016;9:1461–1471. doi: 10.1242/dmm.026369. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 154.Germano G, Frapolli R, Belgiovine C, Anselmo A, Pesce S, Liguori M, et al. Role of macrophage targeting in the antitumor activity of trabectedin. Cancer Cell. 2013;23:249–262. doi: 10.1016/j.ccr.2013.01.008. [DOI] [PubMed] [Google Scholar]
  • 155.Liguori M, Buracchi C, Pasqualini F, Bergomas F, Pesce S, Sironi M, et al. Functional TRAIL receptors in monocytes and tumor-associated macrophages: A possible targeting pathway in the tumor microenvironment. Oncotarget. 2016;7:41662–41676. doi: 10.18632/oncotarget.9340. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 156.Allavena P, Signorelli M, Chieppa M, Erba E, Bianchi G, Marchesi F, et al. Anti-inflammatory properties of the novel antitumor agent yondelis (trabectedin): inhibition of macrophage differentiation and cytokine production. Cancer Res. 2005;65:2964–2971. doi: 10.1158/0008-5472.CAN-04-4037. [DOI] [PubMed] [Google Scholar]
  • 157.Mantovani A, Allavena P. The interaction of anticancer therapies with tumor-associated macrophages. J Exp Med. 2015;212:435–445. doi: 10.1084/jem.20150295. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 158.De Palma M, Lewis CE. Macrophage regulation of tumor responses to anticancer therapies. Cancer Cell. 2013;23:277–286. doi: 10.1016/j.ccr.2013.02.013. [DOI] [PubMed] [Google Scholar]
  • 159.Mullins DW, Burger CJ, Elgert KD. Paclitaxel enhances macrophage IL-12 production in tumor-bearing hosts through nitric oxide. J Immunol. 1999;162:6811–6818. [PubMed] [Google Scholar]
  • 160.Kodumudi KN, Woan K, Gilvary DL, Sahakian E, Wei S, Djeu JY. A novel chemoimmunomodulating property of docetaxel: suppression of myeloid-derived suppressor cells in tumor bearers. Clin Cancer Res. 2010;16:4583–4594. doi: 10.1158/1078-0432.CCR-10-0733. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 161.Guerriero JL, Ditsworth D, Catanzaro JM, Sabino G, Furie MB, Kew RR, et al. DNA alkylating therapy induces tumor regression through an HMGB1-mediated activation of innate immunity. J Immunol. 2011;186:3517–3526. doi: 10.4049/jimmunol.1003267. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 162.Bryniarski K, Szczepanik M, Ptak M, Zemelka M, Ptak W. Influence of cyclophosphamide and its metabolic products on the activity of peritoneal macrophages in mice. Pharmacol Rep. 2009;61:550–557. doi: 10.1016/s1734-1140(09)70098-2. [DOI] [PubMed] [Google Scholar]
  • 163.Baghdadi M, Wada H, Nakanishi S, Abe H, Han N, Putra WE, et al. Chemotherapy-Induced IL34 Enhances Immunosuppression by Tumor-Associated Macrophages and Mediates Survival of Chemoresistant Lung Cancer Cells. Cancer Res. 2016;76:6030–6042. doi: 10.1158/0008-5472.CAN-16-1170. [DOI] [PubMed] [Google Scholar]
  • 164.Kaczanowska S, Joseph AM, Davila E. TLR agonists: our best frenemy in cancer immunotherapy. J Leukoc Biol. 2013;93:847–863. doi: 10.1189/jlb.1012501. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 165.Tang X, Mo C, Wang Y, Wei D, Xiao H. Anti-tumour strategies aiming to target tumour-associated macrophages. Immunology. 2013;138:93–104. doi: 10.1111/imm.12023. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 166.Gebremeskel S, Johnston B. Concepts and mechanisms underlying chemotherapy induced immunogenic cell death: impact on clinical studies and considerations for combined therapies. Oncotarget. 2015;6:41600–41619. doi: 10.18632/oncotarget.6113. http://www.ncbi.nlm.nih.gov/pmc/articles/PMC4747176/ [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 167.Zheng JH, Nguyen VH, Jiang SN, Park SH, Tan W, Hong SH, et al. Two-step enhanced cancer immunotherapy with engineered <em>Salmonella typhimurium</em> secreting heterologous flagellin. Sci Transl Med. 2017;9 doi: 10.1126/scitranslmed.aak9537. http://stm.sciencemag.org/content/9/376/eaak9537.abstract. [DOI] [PubMed] [Google Scholar]
  • 168.Hagemann T, Lawrence T, McNeish I, Charles KA, Kulbe H, Thompson RG, et al. Re-educating” tumor-associated macrophages by targeting NF-κB. J Exp Med. 2008;205:1261. doi: 10.1084/jem.20080108. LP-1268. http://jem.rupress.org/content/205/6/1261.abstract. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 169.Tabata Y, Ikada Y. Effect of the size and surface charge of polymer microspheres on their phagocytosis by macrophage. Biomaterials. 1988;9:356–362. doi: 10.1016/0142-9612(88)90033-6. [DOI] [PubMed] [Google Scholar]
  • 170.Champion JA, Mitragotri S. Role of target geometry in phagocytosis. Proc Natl Acad Sci U S A. 2006;103:4930–4934. doi: 10.1073/pnas.0600997103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 171.He C, Hu Y, Yin L, Tang C, Yin C. Effects of particle size and surface charge on cellular uptake and biodistribution of polymeric nanoparticles. Biomaterials. 2010;31:3657–3666. doi: 10.1016/j.biomaterials.2010.01.065. [DOI] [PubMed] [Google Scholar]
  • 172.Sosale NG, Spinler KR, Alvey C, Discher DE. Macrophage engulfment of a cell or nanoparticle is regulated by unavoidable opsonization, a species-specific “Marker of Self” CD47, and target physical properties. Curr Opin Immunol. 2015;35:107–112. doi: 10.1016/j.coi.2015.06.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 173.Chono S, Tanino T, Seki T, Morimoto K. Uptake characteristics of liposomes by rat alveolar macrophages: influence of particle size and surface mannose modification. J Pharm Pharmacol. 2007;59:75–80. doi: 10.1211/jpp.59.1.0010. [DOI] [PubMed] [Google Scholar]
  • 174.Chang YN, Guo H, Li J, Song Y, Zhang M, Jin J, et al. Adjusting the balance between effective loading and vector migration of macrophage vehicles to deliver nanoparticles. PLoS One. 2013;8:e76024. doi: 10.1371/journal.pone.0076024. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 175.Clift MJD, Rothen-Rutishauser B, Brown DM, Duffin R, Donaldson K, Proudfoot L, et al. The impact of different nanoparticle surface chemistry and size on uptake and toxicity in a murine macrophage cell line. Toxicol Appl Pharmacol. 2008;232:418–427. doi: 10.1016/j.taap.2008.06.009. http://dx.doi.org/10.1016/j.taap.2008.06.009. [DOI] [PubMed] [Google Scholar]
  • 176.Schwendener RA, Lagocki PA, Rahman YE. The effects of charge and size on the interaction of unilamellar liposomes with macrophages. Biochim Biophys Acta. 1984;772:93–101. doi: 10.1016/0005-2736(84)90521-2. [DOI] [PubMed] [Google Scholar]
  • 177.Yu SS, Lau CM, Thomas SN, Jerome WG, Maron DJ, Dickerson JH, et al. Size- and charge-dependent nonspecific uptake of PEGylated nanoparticles by macrophages. Int J Nanomedicine. 2012;7:799–813. doi: 10.2147/IJN.S28531. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 178.Herd H, Daum N, Jones AT, Huwer H, Ghandehari H, Lehr CM. Nanoparticle geometry and surface orientation influence mode of cellular uptake. ACS Nano. 2013;7:1961–1973. doi: 10.1021/nn304439f. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 179.Allen TM, Austin GA, Chonn A, Lin L, Lee KC. Uptake of liposomes by cultured mouse bone marrow macrophages: influence of liposome composition and size. Biochim Biophys Acta. 1991;1061:56–64. doi: 10.1016/0005-2736(91)90268-d. [DOI] [PubMed] [Google Scholar]
  • 180.Key J, Palange AL, Gentile F, Aryal S, Stigliano C, Di Mascolo D, et al. Soft Discoidal Polymeric Nanoconstructs Resist Macrophage Uptake and Enhance Vascular Targeting in Tumors. ACS Nano. 2015;9:11628–11641. doi: 10.1021/acsnano.5b04866. [DOI] [PubMed] [Google Scholar]
  • 181.Li Z, Sun L, Zhang Y, Dove AP, O’Reilly RK, Chen G. Shape Effect of Glyco-Nanoparticles on Macrophage Cellular Uptake and Immune Response. ACS Macro Lett. 2016;5:1059–1064. doi: 10.1021/acsmacrolett.6b00419. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 182.Qiu Y, Liu Y, Wang L, Xu L, Bai R, Ji Y, et al. Surface chemistry and aspect ratio mediated cellular uptake of Au nanorods. Biomaterials. 2010;31:7606–7619. doi: 10.1016/j.biomaterials.2010.06.051. [DOI] [PubMed] [Google Scholar]
  • 183.Bartneck M, Keul HA, Singh S, Czaja K, Bornemann J, Bockstaller M, et al. Rapid uptake of gold nanorods by primary human blood phagocytes and immunomodulatory effects of surface chemistry. ACS Nano. 2010;4:3073–3086. doi: 10.1021/nn100262h. [DOI] [PubMed] [Google Scholar]
  • 184.MacParland SA, Tsoi KM, Ouyang B, Ma XZ, Manuel J, Fawaz A, et al. Phenotype Determines Nanoparticle Uptake by Human Macrophages from Liver and Blood. ACS Nano. 2017;11:2428–2443. doi: 10.1021/acsnano.6b06245. [DOI] [PubMed] [Google Scholar]
  • 185.Blanco E, Shen H, Ferrari M. Principles of nanoparticle design for overcoming biological barriers to drug delivery. Nat Biotechnol. 2015;33:941–951. doi: 10.1038/nbt.3330. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 186.Tsai CY, Lu SL, Hu CW, Yeh CS, Lee GB, Lei HY. Size-dependent attenuation of TLR9 signaling by gold nanoparticles in macrophages. J Immunol. 2012;188:68–76. doi: 10.4049/jimmunol.1100344. [DOI] [PubMed] [Google Scholar]
  • 187.Herd HL, Bartlett KT, Gustafson JA, McGill LD, Ghandehari H. Macrophage silica nanoparticle response is phenotypically dependent. Biomaterials. 2015;53:574–582. doi: 10.1016/j.biomaterials.2015.02.070. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 188.Tanei T, Leonard F, Liu X, Alexander JF, Saito Y, Ferrari M, et al. Redirecting Transport of Nanoparticle Albumin-Bound Paclitaxel to Macrophages Enhances Therapeutic Efficacy against Liver Metastases. Cancer Res. 2016;76:429–439. doi: 10.1158/0008-5472.CAN-15-1576. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 189.Van Rooijen N, Sanders A. Liposome mediated depletion of macrophages: mechanism of action, preparation of liposomes and applications. J Immunol Methods. 1994;174:83–93. doi: 10.1016/0022-1759(94)90012-4. [DOI] [PubMed] [Google Scholar]
  • 190.Zhu S, Niu M, O’Mary H, Cui Z. Targeting of Tumor-Associated Macrophages Made Possible by PEG-Sheddable, Mannose-Modified Nanoparticles. Mol Pharm. 2013;10:3525–3530. doi: 10.1021/mp400216r. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 191.Sun X, Gao D, Gao L, Zhang C, Yu X, Jia B, et al. Molecular imaging of tumor-infiltrating macrophages in a preclinical mouse model of breast cancer. Theranostics. 2015;5:597–608. doi: 10.7150/thno.11546. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 192.Irache JM, Salman HH, Gamazo C, Espuelas S. Mannose-targeted systems for the delivery of therapeutics. Expert Opin Drug Deliv. 2008;5:703–724. doi: 10.1517/17425247.5.6.703. [DOI] [PubMed] [Google Scholar]
  • 193.Hattori Y, Yamashita J, Sakaida C, Kawano K, Yonemochi E. Evaluation of antitumor effect of zoledronic acid entrapped in folate-linked liposome for targeting to tumor-associated macrophages. J Liposome Res. 2015;25:131–140. doi: 10.3109/08982104.2014.954128. [DOI] [PubMed] [Google Scholar]
  • 194.Puig-Kroger A, Sierra-Filardi E, Dominguez-Soto A, Samaniego R, Corcuera MT, Gomez-Aguado F, et al. Folate receptor beta is expressed by tumor-associated macrophages and constitutes a marker for M2 anti-inflammatory/regulatory macrophages. Cancer Res. 2009;69:9395–9403. doi: 10.1158/0008-5472.CAN-09-2050. [DOI] [PubMed] [Google Scholar]
  • 195.Golani LK, Wallace-Povirk A, Deis SM, Wong J, Ke J, Gu X, et al. Tumor Targeting with Novel 6-Substituted Pyrrolo [2,3-d] Pyrimidine Antifolates with Heteroatom Bridge Substitutions via Cellular Uptake by Folate Receptor α and the Proton-Coupled Folate Transporter and Inhibition of de Novo Purine Nucleotide Biosynthesis. J Med Chem. 2016;59:7856–7876. doi: 10.1021/acs.jmedchem.6b00594. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 196.Nagai T, Tanaka M, Tsuneyoshi Y, Xu B, Michie SA, Hasui K, et al. Targeting tumor-associated macrophages in an experimental glioma model with a recombinant immunotoxin to folate receptor beta. Cancer Immunol Immunother. 2009;58:1577–1586. doi: 10.1007/s00262-009-0667-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 197.Xia W, Hilgenbrink AR, Matteson EL, Lockwood MB, Cheng JX, Low PS. A functional folate receptor is induced during macrophage activation and can be used to target drugs to activated macrophages. Blood. 2009;113:438–446. doi: 10.1182/blood-2008-04-150789. [DOI] [PubMed] [Google Scholar]
  • 198.Han W, Zaynagetdinov R, Yull FE, Polosukhin VV, Gleaves LA, Tanjore H, et al. Molecular imaging of folate receptor beta-positive macrophages during acute lung inflammation. Am J Respir Cell Mol Biol. 2015;53:50–59. doi: 10.1165/rcmb.2014-0289OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 199.Furusho Y, Miyata M, Matsuyama T, Nagai T, Li H, Akasaki Y, et al. Novel Therapy for Atherosclerosis Using Recombinant Immunotoxin Against Folate Receptor beta-Expressing Macrophages. J Am Heart Assoc. 2012;1:e003079. doi: 10.1161/JAHA.112.003079. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 200.Lin Y, Wei C, Liu Y, Qiu Y, Liu C, Guo F. Selective ablation of tumor-associated macrophages suppresses metastasis and angiogenesis. Cancer Sci. 2013;104:1217–1225. doi: 10.1111/cas.12202. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 201.Corna G, Campana L, Pignatti E, Castiglioni A, Tagliafico E, Bosurgi L, et al. Polarization dictates iron handling by inflammatory and alternatively activated macrophages. Haematologica. 2010;95:1814–1822. doi: 10.3324/haematol.2010.023879. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 202.Wu W, Luo Y, Sun C, Liu Y, Kuo P, Varga J, et al. Targeting cell-impermeable prodrug activation to tumor microenvironment eradicates multiple drug-resistant neoplasms. Cancer Res. 2006;66:970–980. doi: 10.1158/0008-5472.CAN-05-2591. [DOI] [PubMed] [Google Scholar]
  • 203.Liu Z, Xiong M, Gong J, Zhang Y, Bai N, Luo Y, et al. Legumain protease-activated TAT-liposome cargo for targeting tumours and their microenvironment. Nat Commun. 2014;5:4280. doi: 10.1038/ncomms5280. [DOI] [PubMed] [Google Scholar]
  • 204.Ruan S, Hu C, Tang X, Cun X, Xiao W, Shi K, et al. Increased Gold Nanoparticle Retention in Brain Tumors by in Situ Enzyme-Induced Aggregation. ACS Nano. 2016;10:10086–10098. doi: 10.1021/acsnano.6b05070. [DOI] [PubMed] [Google Scholar]
  • 205.Choi CHJ, Alabi CA, Webster P, Davis ME. Mechanism of active targeting in solid tumors with transferrin-containing gold nanoparticles. Proc Natl Acad Sci U S A. 2010;107:1235–1240. doi: 10.1073/pnas.0914140107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 206.Clark AJ, Davis ME. Increased brain uptake of targeted nanoparticles by adding an acid-cleavable linkage between transferrin and the nanoparticle core. Proc Natl Acad Sci U S A. 2015;112:12486–12491. doi: 10.1073/pnas.1517048112. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 207.Kang T, Jiang M, Jiang D, Feng X, Yao J, Song Q, et al. Enhancing Glioblastoma-Specific Penetration by Functionalization of Nanoparticles with an Iron-Mimic Peptide Targeting Transferrin/Transferrin Receptor Complex. Mol Pharm. 2015;12:2947–2961. doi: 10.1021/acs.molpharmaceut.5b00222. [DOI] [PubMed] [Google Scholar]
  • 208.Movahedi K, Schoonooghe S, Laoui D, Houbracken I, Waelput W, Breckpot K, et al. Nanobody-based targeting of the macrophage mannose receptor for effective in vivo imaging of tumor-associated macrophages. Cancer Res. 2012;72:4165–4177. doi: 10.1158/0008-5472.CAN-11-2994. [DOI] [PubMed] [Google Scholar]
  • 209.Cieslewicz M, Tang J, Yu JL, Cao H, Zavaljevski M, Motoyama K, et al. Targeted delivery of proapoptotic peptides to tumor-associated macrophages improves survival. Proc Natl Acad Sci U S A. 2013;110:15919–15924. doi: 10.1073/pnas.1312197110. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 210.Ngambenjawong C, Gustafson HH, Pineda JM, Kacherovsky NA, Cieslewicz M, Pun SH. Serum Stability and Affinity Optimization of an M2 Macrophage-Targeting Peptide (M2pep) Theranostics. 2016;6:1403–1414. doi: 10.7150/thno.15394. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 211.Ngambenjawong C, Pineda JMB, Pun SH. Engineering an Affinity-Enhanced Peptide through Optimization of Cyclization Chemistry. Bioconjug Chem. 2016;27:2854–2862. doi: 10.1021/acs.bioconjchem.6b00502. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 212.Ngambenjawong C, Cieslewicz M, Schellinger JG, Pun SH. Synthesis and evaluation of multivalent M2pep peptides for targeting alternatively activated M2 macrophages. J Control Release. 2016;224:103–111. doi: 10.1016/j.jconrel.2015.12.057. http://dx.doi.org/10.1016/j.jconrel.2015.12.057. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 213.Lee S, Kivimae S, Dolor A, Szoka FC. Macrophage-based cell therapies: The long and winding road. J Control Release. 2016;240:527–540. doi: 10.1016/j.jconrel.2016.07.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 214.Choi J, Kim HY, Ju EJ, Jung J, Park J, Chung HK, et al. Use of macrophages to deliver therapeutic and imaging contrast agents to tumors. Biomaterials. 2012;33:4195–4203. doi: 10.1016/j.biomaterials.2012.02.022. [DOI] [PubMed] [Google Scholar]
  • 215.Madsen SJ, Baek SK, Makkouk AR, Krasieva T, Hirschberg H. Macrophages as cell-based delivery systems for nanoshells in photothermal therapy. Ann Biomed Eng. 2012;40:507–515. doi: 10.1007/s10439-011-0415-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 216.Ikehara Y, Niwa T, Biao L, Ikehara SK, Ohashi N, Kobayashi T, et al. A carbohydrate recognition-based drug delivery and controlled release system using intraperitoneal macrophages as a cellular vehicle. Cancer Res. 2006;66:8740–8748. doi: 10.1158/0008-5472.CAN-06-0470. [DOI] [PubMed] [Google Scholar]
  • 217.Miller MA, Zheng YR, Gadde S, Pfirschke C, Zope H, Engblom C, et al. Tumour-associated macrophages act as a slow-release reservoir of nano-therapeutic Pt(IV) pro-drug. Nat Commun. 2015;6:8692. doi: 10.1038/ncomms9692. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 218.Dou H, Destache CJ, Morehead JR, Mosley RL, Boska MD, Kingsley J, et al. Development of a macrophage-based nanoparticle platform for antiretroviral drug delivery. Blood. 2006;108:2827–2835. doi: 10.1182/blood-2006-03-012534. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 219.Dou H, Grotepas CB, McMillan JM, Destache CJ, Chaubal M, Werling J, et al. Macrophage delivery of nanoformulated antiretroviral drug to the brain in a murine model of neuroAIDS. J Immunol. 2009;183:661–669. doi: 10.4049/jimmunol.0900274. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 220.Zhao Y, Haney MJ, Klyachko NL, Li S, Booth SL, Higginbotham SM, et al. Polyelectrolyte Complex Optimization for Macrophage Delivery of Redox Enzyme Nanoparticles. Nanomedicine (Lond) 2011;6:25–42. doi: 10.2217/nnm.10.129. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 221.Klyachko NL, Haney MJ, Zhao Y, Manickam DS, Mahajan V, Suresh P, et al. Macrophages offer a paradigm switch for CNS delivery of therapeutic proteins. Nanomedicine (Lond) 2014;9:1403–1422. doi: 10.2217/nnm.13.115. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 222.Muthana M, Kennerley AJ, Hughes R, Fagnano E, Richardson J, Paul M, et al. Directing cell therapy to anatomic target sites in vivo with magnetic resonance targeting. Nat Commun. 2015;6:8009. doi: 10.1038/ncomms9009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 223.Han J, Zhen J, Du Nguyen V, Go G, Choi Y, Ko SY, et al. Hybrid-Actuating Macrophage-Based Microrobots for Active Cancer Therapy. Sci Rep. 2016;6:28717. doi: 10.1038/srep28717. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 224.Li F, Ulrich M, Jonas M, Stone IJ, Linares G, Zhang X, et al. Tumor associated macrophages can contribute to antitumor activity through FcgammaRmediated processing of antibody-drug conjugates. Mol Cancer Ther. 2017 doi: 10.1158/1535-7163.MCT-17-0019. [DOI] [PubMed] [Google Scholar]
  • 225.De Palma M, Mazzieri R, Politi LS, Pucci F, Zonari E, Sitia G, et al. Tumor-targeted interferon-alpha delivery by Tie2-expressing monocytes inhibits tumor growth and metastasis. Cancer Cell. 2008;14:299–311. doi: 10.1016/j.ccr.2008.09.004. [DOI] [PubMed] [Google Scholar]
  • 226.Pradel LP, Ooi CH, Romagnoli S, Cannarile MA, Sade H, Ruttinger D, et al. Macrophage Susceptibility to Emactuzumab (RG7155) Treatment. Mol Cancer Ther. 2016;15:3077–3086. doi: 10.1158/1535-7163.MCT-16-0157. [DOI] [PubMed] [Google Scholar]
  • 227.Carey LA, Winer EP. I-SPY 2 — Toward More Rapid Progress in Breast Cancer Treatment. N Engl J Med. 2016;375:83–84. doi: 10.1056/NEJMe1603691. [DOI] [PubMed] [Google Scholar]

Associated Data

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

Supplementary Materials

supplement

RESOURCES