The Tumor Microenvironment Innately Modulates Cancer Progression
The Tumor Microenvironment Innately Modulates Cancer Progression
Review Research
Abstract
Cancer development and progression occurs in concert phoid cells, myeloid-derived suppressor cells, and natural
with alterations in the surrounding stroma. Cancer cells can killer cells) as well as adaptive immune cells (T cells and B
functionally sculpt their microenvironment through the cells) contribute to tumor progression when present in the
secretion of various cytokines, chemokines, and other fac- tumor microenvironment (TME). Cross-talk between cancer
tors. This results in a reprogramming of the surrounding cells and the proximal immune cells ultimately results in an
cells, enabling them to play a determinative role in tumor environment that fosters tumor growth and metastasis.
Introduction edge to current therapies that target dysfunctional cells in the TME.
In this review, we summarize the current knowledge on the ability
The tumor microenvironment (TME) is complex and contin-
of the TME to co-opt innate immune cells for cancer promotion
uously evolving. In addition to stromal cells, fibroblasts, and
and clinical strategies targeting these innate immune responses in
endothelial cells, the TME comprises innate and adaptive immune
the context of cancer.
cells. Previous studies have focused predominantly on adaptive
immune cells in the context of cancer. T lymphocytes, in partic-
Macrophages
ular, have been a target of interest for their potent cytotoxic
Of all of the innate immune cells, monocyte-derived macro-
capabilities, so much so that their differentiation status became
phages (Mj) best reflect the Th1/Th2 paradigm. Simplistically,
a model for other cell types and was coined the "Th1/Th2
Mjs can be polarized into inflammatory M1 (classically activat-
paradigm" (1). This dichotomy posits that T cells orchestrate
ed) or immune-suppressive M2 Mjs (alternatively activated) (2).
pathogen-dependent immune responses by differential produc-
Mjs modulate immune responses through pathogen phagocyto-
tion of cytokines: Th1 cells govern a proinflammatory phenotype
sis and antigen presentation, and also function in wound healing
and Th2 cells orchestrate an immunosuppressive phenotype.
and tissue repair, thus necessitating them for immune homeo-
Current TME-targeted treatments have focused predominantly
stasis (3). Mjs are tissue-specific and ubiquitous; they contribute
on T cells; prime examples include checkpoint blockade and
to all stages of wound healing, tissue formation, coagulation,
chimeric antigen receptor (CAR) T-cell therapies. With an expan-
inflammation, and tissue reorganization (4). Mjs first appear in
sion of the literature regarding the TME, it is now evident that the
the yolk sac on embryonic day 7, and from there they disseminate
innate immune response not only indirectly influences the TME
to peripheral tissues to establish tissue-resident Mjs, although a
by controlling T-cell fate, but also critically sculpts the TME. These
majority of adult tissue Mj populations (including the spleen,
innate immune cell types include macrophages, dendritic cells
lung, and skin), originate in the fetal liver, indicating that the Mjs
(DC), neutrophils, myeloid-derived suppressor cells (MDSC),
established by the yolk sac are replaced by those that originate in
natural killer cells (NK), and innate lymphoid cells (ILC). Mech-
the fetal liver. Specifically, hematopoietic stem cells colonizing
anistically, cytokines within the TME manipulate immune func-
the fetal liver give rise to all hematopoietic lineages, including
tions that culminate in muted immune responses that guide
monocytes (5). In the context of cancer, one form of Mj recruit-
tumor progression. It is essential to develop a comprehensive
ment includes recruitment from the bone marrow as monocytes
understanding of the innate immune cells and extend this knowl-
by chemokines (CCL1, CCL2, CCL3, CCL4, CCL5, CCL7, CXCL1,
CXCL2, CXCL4, CX3CL1), leading to Mj differentiation in
1
response to cytokines (CSF-1) that are secreted by many different
Department of Pathology, The University of Alabama at Birmingham,
cell types, including tumor cells, osteoblasts, and uterine epithe-
Birmingham, Alabama. 2O'Neal Comprehensive Cancer Center, The University
of Alabama at Birmingham, Birmingham, Alabama. lial cells (4, 6).
The TME potentiates immune suppressive M2 Mjs through the
Corresponding Author: Lalita A. Shevde, University of Alabama at Birmingham,
secretion of cytokines such as IL4 (Fig. 1A and B). Cumulatively,
Birmingham, AL 35233. Phone: 205-975-6261; Fax: 205-975-6615; E-mail:
[email protected] this enables tumor growth and progression as Mjs can make up to
50% of tumor mass (7). High Mj infiltration of most tumor types
Cancer Res 2019;79:4557–66
including breast cancer, gastric cancer, lung cancer, hepatoma,
doi: 10.1158/0008-5472.CAN-18-3962 and other malignancies correlated with a negative prognosis,
2019 American Association for Cancer Research. further establishing their role in cancer progression (8–10). Also,
www.aacrjournals.org 4557
Hinshaw and Shevde
TC IL1
F & NK1 MDSC 8
an F
TC
me fa gio TC NK2
NKT1 Pe TC n zy rin ct g
rfo a o or en
rin F
TC Gr erf s i TC TC
TC 1
p
T
c Fβ
TC TC IL1 GF β TG 13
TC 3 G IL1 TC TC , IL
Phagocytosis M- 8, TC IL5
TC TC and inos CS
cytosis TC TC F TC TC TC TC IL4
Phago TC M1 NKT2 IL13
TC TC M2
DC1 F TC IL10
TC TC F
TC IL2
TC TC TC 5,
TC enesis IL3
angiog TC 3
r in &
TG
rfo e
α F Fβ
pe z y m
IFN
TNF F
Phagocytosis
, IL
M1 IL6 DC2 TGFβ EG
γT
13
an
TG
,A ,V
NF
IL4
Gr
Fβ
PC 2
IL6, IL10
α
L ILC2
IL4
, TG
0
CC
L1
IFNγ
3,
,I
Fβ
Fβ
NK1 IL
TG
Th1 APC
ILC1
3, APC
γ
IF
ILC1 Th2
NKT3
Figure 1.
Cross-talk in the tumor microenvironment. A, The impact of inflammatory or tumor-suppressive immune cells on tumor cells in the TME. The bold arrows show
the impact that immune cells ideally have on tumor cells (TC). The interactions between NKTs, DCs, T cells, neutrophils, ILCs, Mw, and NK cells and tumor cells are
depicted. Fibroblasts are denoted with the letter "F." B, The cross-talk between immune cells in the TME that have been polarized to an immune-suppressive
type and the cytokines secreted by the TCs that contribute to this Th2-like polarization.
an aspect of their normal tissue remodeling abilities includes ligands (16). This kindles a feed-forward loop that sustains
regulation of epithelial cell movement. This function of Mjs is alternatively polarized Mjs within the TME. Interfering with
co-opted by tumor cells within the TME; Mjs release factors (e.g., this cross-talk reprogrammed the TME to be immune reactive
EGF) that promote the movement and invasion of cancer and diminished the occurrence of metastasis. Given their role in
cells (11, 12). inducing a premetastatic niche ("a favorable microenvironment
While the M1/M2 classification is a simplified understanding for survival and outgrowth of tumor cells induced at distal sites
of Mj phenotype and function, in reality Mjs are plastic in by tumors"; ref. 17), aiding in extravasation of circulating
nature and exist in a continuum of functional states (7). M2 cancer cells, and promoting metastasis (18), Mjs present as
Mjs can further be classified into M2a, M2b, M2c, and M2d prime candidates for therapeutic intervention.
subsets (Table 1). These subsets are defined on the basis of their
different inducers namely: IFNg, and LPS for M1; IL4, IL10, Dendritic cells
IL13 for M2a; TLR agonists for M2b; IL10, TNFa, and gluco- DCs bridge the gap between the adaptive and innate immune
corticoids for M2c; and TLR and adenosine A2A receptor for systems. They initiate pathogen-specific T-cell responses and are
M2d (13). Furthermore, these differential Mj subtypes have therefore important for bolstering protective immunity. It is
different functional roles as outlined in Table 1. Therefore, it is important to note that B cells and Mjs also perform antigen
unsurprising that Mjs that exhibit properties of both M1 and presentation, albeit with lower activity than that of DCs. To
M2 exist in distinct proportions in the TME, depending on the effectively stimulate the adaptive immune response, DCs must
tumor type, although the M2 phenotype is typically favored. recognize, capture, and present antigens, upregulate costimula-
This poses a conundrum because Mj-mediated killing of cancer tory molecules, produce inflammatory cytokines, and then travel
cells is virtually nonexistent in the TME of tumors with high to secondary lymphoid organs for antigen presentation to T cells.
proportions of M2 Mjs. The wound-healing phenotype of M2 The inability of DCs to perform these functions greatly hampers
Mjs established by the TME enables tumor growth, prolifera- the immune response to pathogens, viruses, and tumors. DCs are
tion, angiogenesis, and epithelial–mesenchymal transition functionally classified into different subtypes such as classical DCs
(EMT; ref. 14). There are many aspects of the TME, including (cDC), plasmacytoid DCs (pDC), and monocyte-derived inflam-
cytokines and hypoxia, which orchestrate Mj polarization and matory DCs (moDC). cDCs can be further divided into cDC1 and
function (Table 1; ref. 15). IL4, commonly present in the TME, cDC2. cDC1s develop under the control of the transcription
initiates STAT6 signaling in Mjs, launching a transcriptional factors IRF8, ID2, and BATF3, and cDC2s develop under the
program that directs alternative polarization of Mjs. In a recent control of transcription factors IRF4, ID2, ZEB, and Notch2/
publication, Hanna and colleagues have identified that tumor KLF4 (19). These subsets are also functionally distinct: cDC1s
cells engage in a dialog with Mjs via secreted Hedgehog are capable of cross-presentation and thus are able to present both
endogenous and exogenous antigens, whereas cDC2s only pres- Analogous to Mjs, DCs are plastic in nature and can be
ent exogenous antigens and do not typically perform cross-pre- stratified into specific subtypes. In the context of cancer, DCs are
sentation. cDCs and pDCs are present and active during steady- broadly referred to as tumor-infiltrating dendritic cells (TIDC),
state conditions, while moDCs tend to only arise during inflam- which will be the predominant focus of this section. TIDCs can be
mation. DCs specialize in different functions dependent on their immunogenic or tolerogenic dependent upon environmental
stage of maturation and differentiation (Table 1). DCs can localize signals. Examples of DCs that contribute to immune suppression
and acclimate to different tissues such as skin, lung, intestine, and include CD5hi cDC2s that stimulate Th2, Th17, and T regulatory
liver and efficiently respond to environmental stimuli (20). responses (19). It is important to note that each of the subtypes
referred to in Table 1 can make up TIDCs that often adopt an (ROS/RNS). They also reconfigure the extracellular matrix
immune-suppressive phenotype due to the suppressive nature of through secretion of neutrophil elastase (NE) and matrix metal-
the TME. loproteinases (MMP8/9) in the TME and promote angiogenesis
Tumors classically reprogram their microenvironment to sup- (Oncostatin-M), tumor progression (PGE2), and invasion
port their survival. In the context of DCs, they do so by secreting (through the release of ROS/RNS, NE, MMP-9). NETs are com-
cytokines to upregulate transcriptional and metabolic pathways prised of MMPs, cathepsin G, and NE (34, 35). These proteases
that promote a tolerogenic phenotype, such as those that involve degrade proinflammatory cytokines and reposition the TME to
IDO, Arg1, iNOS, and STAT3 (21). These pathways trigger altera- enhance tumor progression and aid in metastasis (36).
tions in DC metabolism, metabolite production, energetic shifts, The plasticity of circulating neutrophils is an important feature
and/or alterations of chromatin accessibility (22). These modifi- in patients with cancer. These neutrophils, called high-density
cations impact every aspect of DC functionality, including their neutrophils (HDN) or low-density neutrophils (LDN), corre-
abilities to secrete inflammatory cytokines and to prime effector T spond to N1 and N2 phenotypes, respectively. In many cancer
cells. Generally, DCs patrolling the TME encounter immune- types, LDNs, which exhibit a more immature phenotype, pre-
suppressive factors such as VEGF, IL10, TGFb, prostaglandin E2 dominate in the circulation and may contribute to cancer pro-
(PGE2), and other cytokines (seen in Fig. 1B) that inhibit DC gression and metastasis (29). A detailed understanding of neu-
maturation into immunogenic cells and promote their develop- trophils and signals that pivot neutrophils to become immune
ment into a tolerogenic phenotype, not only stunting their Th1- suppressive holds much promise toward reprogramming the
factors in the TME can alter the metabolism of MDSCs toward breast cancer (55) in mouse models. However, NKTIIs have been
fatty acid oxidation, prompting an upregulation of Arg1 and reported to support MDSCs in a B-cell lymphoma mouse mod-
NOS2 production (47). The critical role of MDSCs in tumorigen- el (54, 56). As such, targeting NKTIIs and supplementation with
esis, growth, the establishment of the premetastatic niche, and NKTIs may provide an exciting therapeutic approach.
metastatic outgrowth warrants the need to effectively target them
by depletion or blockade. Although their critical role in the Innate lymphoid cells
survival and advancement of tumors is well known, there are Another crucial component of the TME is the ILCs that have
currently no FDA-approved drugs or therapies that directly target characteristics similar to those of NK cells. ILCs share a common
MDSCs. lymphoid progenitor with B and T cells, but lack B- and T-cell
receptors and are thus classified as innate immune cells (57). ILCs
Natural killer cells and natural killer T cells contribute to T-cell polarization through antigen presentation
NK cells are circulatory, innate lymphoid cells recognized for and cytokine secretion (58). There are three types of ILCs (ILC1,
their cytotoxic effector functions. Classically, there are two subsets ILC2, and ILC3) classified on the basis of their production of Th1,
of NKs defined by their expression of CD16 and CD56 levels: Th2, and Th17-based cytokines and distinct transcription fac-
namely, CD56hi CD16 and CD56lo CD16hi (48). CD56hi CD16 tors. (59). ILC1s tend to exhibit antitumor functions through
NKs secrete inflammatory cytokines, whereas CD56lo CD16hi NKs cytokine production (mainly IFNg). Furthermore, ILC1s can be
specialize in cytotoxic functions and cell-mediated killing. Within divided into NK ILC1s and non-NK ILC1s based on their expres-
immune cell cross-talk and tumor–stromal cross-talk, but also back into the patient. This therapy has achieved significant success
stromal–immune cell cross-talk as it contributes significantly to marked by diminished tumor burden in patients with prostate
tumor development. cancer. A new DC vaccine targeting glioblastoma is DCVax-L that
includes autologous DCs loaded with glioblastoma tumor lysate.
Current and future therapeutics This vaccine has been tested in a phase III clinical trial for
The tumor masterfully controls its surrounding environment to glioblastoma, and overall patient survival was shown to increase
promote its establishment, growth, survival, and spread. One of by 6 months (80).
the chief ways it does this is through reprogramming innate Despite success with DC vaccinations, there are challenges
immune cells to foster tumor growth and survival, leaving the associated with them, including high cost, the absence of univer-
patient with a weakened defense and often a worse prognosis. This sal vaccine, the need for massive amounts of DCs, and issues with
is a potential Achilles heel of the tumor; as such, reprogramming polarizing conventional DCs in vitro. Previous attempts at DC
the innate immune system is a potentially important approach to vaccinations focused on moDCs that are rare and do not func-
improve patient outcomes. tionally resemble cross-presenting DCs in vivo (81). It is now
recognized that cDCs comprise the DC subtype that is most likely
Macrophage therapies to come into contact with cancer cells in the TME and mount the
Previous clinical trials targeting Mjs in the TME have been ensuing immune response. While cDCs are challenging to isolate,
unsuccessful. Many prior trials involved the activation a cDC vaccine for melanoma has been reported to elicit a cytotoxic
References
1. McGuirk P, Mills KH.Pathogen-specific regulatory T cells provoke a shift in stimulating factor-1/epidermal growth factor paracrine loop. Cancer Res
the Th1/Th2 paradigm in immunity to infectious diseases. 2005;65:5278–83.
Trends Immunol 2002;23:450–5. 12. Wyckoff J, Wang W, Lin EY, Wang Y, Pixley F, Stanley ER, et al. A paracrine
2. Yang L, Zhang Y. Tumor-associated macrophages: from basic research to loop between tumor cells and macrophages is required for tumor cell
clinical application. J Hematol Oncol 2017;10:58. migration in mammary tumors. Cancer Res 2004;64:7022–9.
3. Chen H, Shi R, Luo B, Yang X, Qiu L, Xiong J, et al. Macrophage 13. Funes SC, Rios M, Escobar-Vera J, Kalergis AM. Implications of mac-
peroxisome proliferator-activated receptor gamma deficiency delays skin rophage polarization in autoimmunity. Immunology 2018;154:
wound healing through impairing apoptotic cell clearance in mice. 186–95.
Cell Death Dis 2015;6:e1597. 14. Zheng X, Turkowski K, Mora J, Br€ une B, Seeger W, Weigert A, et al.
4. Peiseler M, Kubes P.Macrophages play an essential role in trauma-induced Redirecting tumor-associated macrophages to become tumoricidal
sterile inflammation and tissue repair. Eur J Trauma Emerg Surg 2018;44: effectors as a novel strategy for cancer therapy. Oncotarget 2017;8:
335–49. 48436–52.
5. Varol C, Mildner A, Jung S. Macrophages: development and tissue spe- 15. Petrova V, Annicchiarico-Petruzzelli M, Melino G, Ivano Amelio I. The
cialization. Annu Rev Immunol 2015;33:643–75. hypoxic tumour microenvironment. Oncogenesis 2018;7:10.
6. Qin L, Wu YL, Toneff MJ, Li D, Liao L, Gao X, et al. NCOA1 Directly Targets 16. Hanna A, Metge BJ, Bailey SK, Chen D, Chandrashekar DS, Varambally S,
M-CSF1 expression to promote breast cancer metastasis. Cancer Res 2014; et al. Inhibition of Hedgehog signaling reprograms the dysfunctional
74:3477–88. immune microenvironment in breast cancer. Oncoimmunology 2019;8:
7. Kim J, Bae JS.Tumor-associated macrophages and neutrophils in tumor 1548241.
microenvironment. Mediators Inflamm 2016;2016:6058147. 17. Peinado H, Zhang H, Matei IR, Costa-Silva B, Hoshino A, Rodrigues G,
8. Cardoso AP, Pinto ML, Pinto AT, Oliveira MI, Pinto MT, Gonçalves R, et al. et al. Pre-metastatic niches: organ-specific homes for metastases. Nat Rev
Macrophages stimulate gastric and colorectal cancer invasion through Cancer 2017;17:302–17.
EGFR Y(1086), c-Src, Erk1/2 and Akt phosphorylation and smallGTPase 18. Doak GR, Schwertfeger KL, Wood DK. Distant Relations: Macrophage
activity. Oncogene 2014;33:2123–33. Functions in the Metastatic Niche. Trends Cancer 2018;4:445–59.
9. Wang N, Liu W, Zheng Y, Wang S, Yang B, Li M, et al. CXCL1 derived from 19. Collin M, Bigley V. Human dendritic cell subsets: an update. Immunology
tumor-associated macrophages promotes breast cancer metastasis via 2018;154:3–20.
activating NF-kappaB/SOX4 signaling. Cell Death Dis 2018;9:880. 20. Motta JM, Rumjanek VM.Sensitivity of Dendritic Cells to Microenviron-
10. Zhao X, Qu J, Sun Y, Wang J, Liu X, Wang F, et al. Prognostic significance of ment Signals. J Immunol Res 2016;2016:4753607.
tumor-associated macrophages in breast cancer: a meta-analysis of the 21. Sumpter TL, Dangi A, Matta BM, Huang C, Stolz DB, Vodovotz Y, et al.
literature. Oncotarget 2017;8:30576–86. Hepatic stellate cells undermine the allostimulatory function of liver
11. Goswami S, Sahai E, Wyckoff JB, Cammer M, Cox D, Pixley FJ, et al. myeloid dendritic cells via STAT3-dependent induction of IDO.
Macrophages promote the invasion of breast carcinoma cells via a colony- J Immunol 2012;189:3848–58.
22. Basit F, Mathan T, Sancho D, de Vries IJM. Human dendritic cell subsets pressor cells in the tumor microenvironment. J Exp Med 2010;207:
undergo distinct metabolic reprogramming for immune response. 2439–53.
Front Immunol 2018;9:2489. 47. Hossain F, Al-Khami AA, Wyczechowska D, Hernandez C, Zheng L, Reiss
23. Fridlender ZG, Sun J, Kim S, Kapoor V, Cheng G, Ling L, et al. Polarization K, et al. Inhibition of fatty acid oxidation modulates immunosuppressive
of tumor-associated neutrophil phenotype by TGF-beta: "N1" versus functions of myeloid-derived suppressor cells and enhances cancer ther-
"N2" TAN. Cancer Cell 2009;16:183–94. apies. Cancer Immunol Res 2015;3:1236–47.
24. Chaput N, Conforti R, Viaud S, Spatz A, Zitvogel L. The Janus face of 48. Stabile H, Fionda C, Gismondi A, Santoni A. Role of distinct natural killer
dendritic cells in cancer. Oncogene 2008;27:5920–31. cell subsets in anticancer response. Front Immunol 2017;8:293.
25. Aspord C, Pedroza-Gonzalez A, Gallegos M, Tindle S, Burton EC, Su D, 49. Glasner A, Levi A, Enk J, Isaacson B, Viukov S, Orlanski S, et al. NKp46
et al. Breast cancer instructs dendritic cells to prime interleukin 13- receptor-mediated interferon-gamma production by natural killer cells
secreting CD4þ T cells that facilitate tumor development. J Exp Med increases fibronectin 1 to alter tumor architecture and control metastasis.
2007;204:1037–47. Immunity 2018;48:396–398.
26. Tran Janco JM, Lamichhane P, Karyampudi L, Knutson KL. Tumor- 50. Imai K, Matsuyama S, Miyake S, Suga K, Nakachi K. Natural cytotoxic
infiltrating dendritic cells in cancer pathogenesis. J Immunol 2015; activity of peripheral-blood lymphocytes and cancer incidence: an 11-
194:2985–91. year follow-up study of a general population. Lancet 2000;356:
27. Michea P, No€el F, Zakine E, Czerwinska U, Sirven P, Abouzid O, et al. 1795–9.
Adjustment of dendritic cells to the breast-cancer microenvironment is 51. Langers I, Renoux VM, Thiry M, Delvenne P, Jacobs N, et al. Natural
subset specific. Nat Immunol 2018;19:885–97. killer cells: role in local tumor growth and metastasis. Biologics 2012;
28. Wang J, Jia Y, Wang N, Zhang X, Tan B, Zhang G, et al. The clinical 6:73–82.
significance of tumor-infiltrating neutrophils and neutrophil-to-CD8þ 52. Maurer S, Kropp KN, Klein G, Steinle A, Haen SP, Walz JS, et al. Platelet-
maturation and function via Mac-1/ICAM-1-dependent mechanism. 84. Colak S,Ten Dijke P.Targeting TGF-beta signaling in cancer. Trends Cancer
J Immunol 2009;183:1767–79. 2017;3:56–71.
68. Lakins MA, Ghorani E, Munir H, Martins CP, Shields JD. Cancer- 85. Connolly EC, Freimuth J, Akhurst RJ. Complexities of TGF-beta targeted
associated fibroblasts induce antigen-specific deletion of CD8 (þ) T cells cancer therapy. Int J Biol Sci 2012;8:964–78.
to protect tumour cells. Nat Commun 2018;9:948. 86. Fleming V, Hu X, Weber R, Nagibin V, Groth C, Altevogt P, et al. Targeting
69. Faradji A, Bohbot A, Frost H, Schmitt-Goguel M, Siffert JC, Dufour P, et al. myeloid-derived suppressor cells to bypass tumor-induced immunosup-
Phase I study of liposomal MTP-PE-activated autologous monocytes pression. Front Immunol 2018;9:398.
administered intraperitoneally to patients with peritoneal carcinomato- 87. Iclozan C, Antonia S, Chiappori A, Chen DT, Gabrilovich D. Therapeutic
sis. J Clin Oncol 1991;9:1251–60. regulation of myeloid-derived suppressor cells and immune response to
70. Hennemann B, Beckmann G, Eichelmann A, Rehm A, Andreesen R. Phase cancer vaccine in patients with extensive stage small cell lung cancer.
I trial of adoptive immunotherapy of cancer patients using monocyte- Cancer Immunol Immunother 2013;62:909–18.
derived macrophages activated with interferon gamma and lipopolysac- 88. Weber R, Fleming V, Hu X, Nagibin V, Groth C, Altevogt P, et al. Myeloid-
charide. Cancer Immunol Immunother 1998;45:250–6. derived suppressor cells hinder the anti-cancer activity of immune check-
71. Stevenson HC, Keenan AM, Woodhouse C, Ottow RT, Miller P, Steller EP, point inhibitors. Front Immunol 2018;9:1310.
et al. Fate of gamma-interferon-activated killer blood monocytes adop- 89. Yang Y, Lim O, Kim TM, Ahn YO, Choi H, Chung H, et al. Phase I Study
tively transferred into the abdominal cavity of patients with peritoneal of random healthy donor-derived allogeneic natural killer cell therapy
carcinomatosis. Cancer Res 1987;47:6100–3. in patients with malignant lymphoma or advanced solid tumors.
72. Ries CH, Cannarile MA, Hoves S, Benz J, Wartha K, Runza V, et al. Cancer Immunol Res 2016;4:215–24.
Targeting tumor-associated macrophages with anti-CSF-1R antibody 90. Veluchamy JP, Kok N, van der Vliet HJ, Verheul HMW, de Gruijl TD,
reveals a strategy for cancer therapy. Cancer Cell 2014;25:846–59. Spanholtz J. The rise of allogeneic natural killer cells as a platform for