In a retrospective analysis of patients with unresectable melanoma, higher pretreatment tissue densities of CD16+ macrophages were associated with clinical benefit from combined CTLA-4 and PD-1 blockade. With further validation, this biomarker could serve as a tool in selecting between immune checkpoint inhibitor regimens.

See related article by Lee et al., p. 2513

In this issue of Clinical Cancer Research, Lee and colleagues report an association between CD16+ macrophage cell density in pretreatment melanoma tissue specimens and clinical outcomes following administration of combined CTLA-4 and PD-1 blockade to patients with treatment-naïve metastatic disease (1). CD16+ macrophage densities were higher in patients that experienced partial or complete responses to therapy compared with those with stable or progressive disease (P = 0.0041), and patients with higher CD16+ macrophage densities were also found to have significantly improved 3-year progression-free survival (87% vs. 42%; P = 0.0056). Importantly, no association between CD16+ macrophage density and clinical outcome was observed in a separate cohort of 50 patients treated with PD-1 monotherapy, indicating specificity of this predictive biomarker to combination CTLA-4 plus PD-1 checkpoint blockade.

Predictive biomarkers for combined CTLA-4 and PD-1 blockade, administered in melanoma as ipilimumab and nivolumab (ipi-nivo), remain a major unmet clinical need in the treatment of unresectable melanoma. Across tumor types including melanoma, tumor-intrinsic and immune biomarkers of response to PD-1 monotherapy have been well characterized including PD-L1 expression, IFNγ gene expression signatures, tumor mutational burden, and microsatellite instability (2, 3). In clinical practice, however, these features are not typically used to select patients with melanoma for anti-PD-1 monotherapy as response rates to pembrolizumab and nivolumab are considerably higher in unselected melanoma populations (4, 5) compared with other options such as chemotherapy. While most patients with unresectable melanoma will be treated with an initial regimen including a PD-1 inhibitor, oncologists must decide which patients warrant initial treatment with ipi-nivo, which is associated with higher response rates but also higher rates of severe immune-related toxicity compared with anti-PD-1 monotherapy (6). The recent regulatory approval of the LAG-3 inhibitor relatlimab in combination with nivolumab for previously untreated, unresectable melanoma has further complicated decision-making. There are currently no widely accepted biomarkers to guide the choice of upfront therapy, and clinicians rely on a combination of patient factors including overall fitness and the presence of high-risk features such as brain metastases (7) to identify patients that may need ipi-nivo as their first treatment. In this context, the findings by Lee and colleagues may represent an important step toward developing a clinically applicable, biological rationale for choosing among different first-line ICI regimens in the treatment of advanced melanoma.

Identification of CD16+ macrophages as a predictive biomarker for combined PD-1 and CTLA-4 blockade potentially supports a proposed role of the Fc region of ipilimumab in its mechanism of action (Fig. 1). While ipilimumab is known to facilitate costimulatory signaling during T-cell priming by antigen-presenting cells (8), some studies have highlighted the importance of immune microenvironment remodeling through the binding of ipilimumab's Fc region to Fc gamma receptors (FcγR), expressed on innate immune cells. CD16 is a low-affinity FcγR that is expressed on monocytes and macrophages and plays a role in antibody-dependent cellular toxicity (9). Downstream sequelae of ipilimumab-FcγR binding in murine tumor models includes depletion of suppressive regulatory T cells (Treg; ref. 10), and in Treg-ablated conditions, reduction in suppressive macrophages and concomitant activation of type I IFN signaling (11). In humans, while Treg depletion has not been demonstrated following treatment with ipilimumab (12), polymorphisms in the CD16a gene that increase binding to IgG1 antibodies are associated with improved overall survival after treatment with ipilimumab (13). Further translational research elucidating changes in the tumor immune microenvironment with CD16 engagement by ipilimumab may inform the identification of additional biomarkers and potential targets for novel immunotherapies.

Figure 1.

Proposed ipilimumab mechanisms of action in the tumor immune microenvironment. The ipilimumab variable region binds to CTLA-4 on T cells, facilitating costimulatory T-cell signaling through CD28. The Fc region binds to CD16 on immunostimulatory macrophages, which express CD86 and complement pathway genes. Downstream effects may include depletion of Tregs and immunosuppressive M2 macrophages. Upregulation of T cell–recruiting chemokines CXCL9, CXL10, and CXCL11 in the tumor microenvironment is also associated with CD16 expression on macrophages.

Figure 1.

Proposed ipilimumab mechanisms of action in the tumor immune microenvironment. The ipilimumab variable region binds to CTLA-4 on T cells, facilitating costimulatory T-cell signaling through CD28. The Fc region binds to CD16 on immunostimulatory macrophages, which express CD86 and complement pathway genes. Downstream effects may include depletion of Tregs and immunosuppressive M2 macrophages. Upregulation of T cell–recruiting chemokines CXCL9, CXL10, and CXCL11 in the tumor microenvironment is also associated with CD16 expression on macrophages.

Close modal

As most research on tumor-associated macrophages (TAM) has focused on immunosuppressive populations that abrogate immune checkpoint inhibitor (ICI) efficacy through molecules such as IL10 and TGFβ, it may be noteworthy that CD16 expression in macrophages was found to be associated with benefit from combined CTLA-4 and PD-1 blockade (1). TAMs exist on a spectrum from a proinflammatory M1-like phenotype expressing MHC type II (MHC II) to an immunosuppressive M2 CD163+ phenotype (14), and have been shown to both help and hinder ICI efficacy. In a YUMM1.7 murine model of melanoma, selective depletion of CD163+ macrophages led to an increase in effector T-cell infiltrates and concomitant tumor regression (15). Studies in other model systems have suggested that PD-1 blockade itself may repolarize macrophages from an M2- to an M1-like phenotype, and that macrophages are necessary for antitumor efficacy of PD-1 blockade (16). Given their expression of complement pathway genes and CD86, the CD16+ macrophages described by Lee and colleagues are likely more M1-like, but functional characterization of this population in model systems and patients are needed to understand their role in mediating combined ICI efficacy.

The complex interplay between tumor-infiltrating T-cell and myeloid populations complicates a simplistic conception of “hot” versus “cold” immune microenvironments as predictors of ICI response. Canonically, T cell–inflamed RNA signatures (17, 18) and higher levels of CD8+ tumor-infiltrating lymphocytes (19) have been associated with better outcomes of PD-1 response. However, how T cells influence the infiltration and behavior of TAM subsets is an area of ongoing investigation. T cell–induced melanoma expression of CSF1 has been associated with higher levels of CD163+ TAMs and impaired response to PD-1 blockade (20). Conversely, in the current study, CD16+ macrophages are associated with expression with T cell–recruiting cytokines (CXCL9, CXCL10, CXCL11), and are higher in on-treatment biopsies of responding than nonresponding patients. Dissecting the specificity and directionality of cross-talk between tumor-infiltrating T cells and macrophage subpopulations may provide additional opportunities to enhance antitumor immune responses.

In parallel with lab-based experiments to elucidate the functional role of CD16+ macrophages in facilitating response to combined CTLA-4 and PD-1 blockade, there are concrete steps that can be taken to translate this observation into a clinical assay to inform patient care. In the immediate future, this candidate biomarker will need further validation in other ipi-nivo–treated melanoma cohorts, and can be prospectively incorporated into ongoing trials of novel agents used in combination with CTLA-4 and PD-1 blockade. A standardized scoring system of CD16+ macrophage staining will need to be developed collaboratively with melanoma pathologists to deploy an IHC assay at scale without dependence on imaging software. While not necessary for validation a predictive biomarker, additional on-treatment biopsies at later timepoints may also be informative to determine whether upregulation in CD16+ macrophages is durable over time, and to track concomitant changes in Treg populations.

Beyond the further development of CD16 macrophage expression as a predictive biomarker, there are related opportunities to leverage emerging high-plex spatial imaging technologies to identify additional biomarkers to address this important clinical question. For instance, identifying immune cell populations that positively correlate with response to combined ipi-nivo and are agnostic or negatively correlate with response to PD-1 monotherapy may be the most useful in identifying patients that truly need up-front addition of CTLA-4 blockade. Alternatively, biopsies of PD-1 refractory melanoma that subsequently respond to ipi-nivo may represent another rich setting for biomarker discovery. In these and other contexts, robust bedside-to-bench characterization of the tumor immune microenvironment will ultimately allow for the application of precision medicine concepts to the field of immuno-oncology.

J.J. Luke reports service on data and safety monitoring boards for AbbVie, Agenus, Immutep, and Evaxion; participation on scientific advisory boards for (no stock) 7Hills, Affivant, BioCytics, Bright Peak, Exo, Fstar, Inzen, RefleXion, and Xilio and (stock) Actym, Alphamab Oncology, Arch Oncology, Duke Street Bio, Kanaph, Mavu, NeoTx, Onc.AI, OncoNano, physIQ, Pyxis, Saros, STipe, and Tempest; consultancy with compensation for AbbVie, Agenus, Alnylam, AstraZeneca, Atomwise, Bayer, Bristol Myers Squibb, Castle, Checkmate, Codiak, Crown, Cugene, Curadev, Day One, Eisai, EMD Serono, Endeavor, Flame, G1 Therapeutics, Genentech, Gilead, Glenmark, HotSpot, Kadmon, KSQ, Janssen, Ikena, Inzen, Immatics, Immunocore, Incyte, Instil, IOBiotech, Macrogenics, Merck, Mersana, Nektar, Novartis, Partner, Pfizer, Pioneering Medicines, PsiOxus, Regeneron, Replimmune, Ribon, Roivant, Servier, STINGthera, Synlogic, and Synthekine; and research support (all to institution for clinical trials unless noted) from AbbVie, Astellas, AstraZeneca, Bristol Myers Squibb, Corvus, Day One, EMD Serono, Fstar, Genmab, Hot Spot, Ikena, Immatics, Incyte, Kadmon, KAHR, Macrogenics, Merck, Moderna, Nektar, Next Cure, Numab, Palleon, Pfizer, Replimmune, Rubius, Servier, Scholar Rock, Synlogic, Takeda, Trishula, Tizona, and Xencor. In addition, J.J. Luke holds patents (both provisional) #15/612,657 (Cancer Immunotherapy) and PCT/US18/36052 (Microbiome Biomarkers for Anti-PD-1/PD-L1 Responsiveness: Diagnostic, Prognostic and Therapeutic Uses Thereof). No disclosures were reported by the other author.

J.W. Smithy acknowledges NIH grant P30CA008748. J.J. Luke acknowledges NIH grants UM1CA186690-06, P50CA254865-01A1, P30CA047904-32, and R01DE031729-01A1.

1.
Lee
H
,
Ferguson
AL
,
Quek
C
,
Vergara
IA
,
Pires daSilva
I
,
Allen
R
, et al
.
Intratumoral CD16+ macrophages are associated with clinical outcomes of patients with metastatic melanoma treated with combination anti-PD-1 and anti-CTLA-4 therapy
.
Clin Cancer Res
2023
;
29
:
2513
24
.
2.
Cristescu
R
,
Mogg
R
,
Ayers
M
,
Albright
A
,
Murphy
E
,
Yearley
J
, et al
.
Pan-tumor genomic biomarkers for PD-1 checkpoint blockade-based immunotherapy
.
Science
2018
;
362
:
eaar3593
.
3.
Litchfield
K
,
Reading
JL
,
Puttick
C
,
Thakkar
K
,
Abbosh
C
,
Bentham
R
, et al
.
Meta-analysis of tumor- and T cell-intrinsic mechanisms of sensitization to checkpoint inhibition
.
Cell
2021
;
184
:
596
614
.
4.
Robert
C
,
Schachter
J
,
Long
GV
,
Arance
A
,
Grob
JJ
,
Mortier
L
, et al
.
Pembrolizumab versus ipilimumab in advanced melanoma
.
N Engl J Med
2015
;
372
:
2521
32
.
5.
Robert
C
,
Long
GV
,
Brady
B
,
Dutriaux
C
,
Maio
M
,
Mortier
L
, et al
.
Nivolumab in previously untreated melanoma without BRAF mutation
.
N Engl J Med
2015
;
372
:
320
30
.
6.
Larkin
J
,
Chiarion-Sileni
V
,
Gonzalez
R
,
Grob
J-J
,
Rutkowski
P
,
Lao
CD
, et al
.
Five-year survival with combined nivolumab and ipilimumab in advanced melanoma
.
N Engl J Med
2019
;
381
:
1535
46
.
7.
Tawbi
HA
,
Forsyth
PA
,
Algazi
A
,
Hamid
O
,
Hodi
FS
,
Moschos
SJ
, et al
.
Combined nivolumab and ipilimumab in melanoma metastatic to the brain
.
N Engl J Med
2018
;
379
:
722
30
.
8.
Weber
J
.
Ipilimumab: controversies in its development, utility and autoimmune adverse events
.
Cancer Immunol Immunother
2009
;
58
:
823
30
.
9.
Yeap
WH
,
Wong
KL
,
Shimasaki
N
,
Teo
ECY
,
Quek
JKS
,
Yong
HX
, et al
.
CD16 is indispensable for antibody-dependent cellular cytotoxicity by human monocytes
.
Sci Rep
2016
;
6
:
34310
.
10.
Simpson
TR
,
Li
F
,
Montalvo-Ortiz
W
,
Sepulveda
MA
,
Bergerhoff
K
,
Arce
F
, et al
.
Fc-dependent depletion of tumor-infiltrating regulatory T cells co-defines the efficacy of anti-CTLA-4 therapy against melanoma
.
J Exp Med
2013
;
210
:
1695
710
.
11.
Yofe
I
,
Landsberger
T
,
Yalin
A
,
Solomon
I
,
Costoya
C
,
Demane
DF
, et al
.
Anti-CTLA-4 antibodies drive myeloid activation and reprogram the tumor microenvironment through FcgammaR engagement and type I interferon signaling
.
Nat Cancer
2022
;
3
:
1336
50
.
12.
Sharma
A
,
Subudhi
SK
,
Blando
J
,
Scutti
J
,
Vence
L
,
Wargo
J
, et al
.
Anti-CTLA-4 immunotherapy does not deplete FOXP3(+) regulatory T cells (tregs) in human cancers
.
Clin Cancer Res
2019
;
25
:
1233
8
.
13.
Arce Vargas
F
,
Furness
AJS
,
Litchfield
K
,
Joshi
K
,
Rosenthal
R
,
Ghorani
E
, et al
.
Fc effector function contributes to the activity of human anti-CTLA-4 antibodies
.
Cancer Cell
2018
;
33
:
649
63
.
14.
Pittet
MJ
,
Michielin
O
,
Migliorini
D
.
Clinical relevance of tumour-associated macrophages
.
Nat Rev Clin Oncol
2022
;
19
:
402
21
.
15.
Etzerodt
A
,
Tsalkitzi
K
,
Maniecki
M
,
Damsky
W
,
Delfini
M
,
Baudoin
E
, et al
.
Specific targeting of CD163(+) TAMs mobilizes inflammatory monocytes and promotes T cell-mediated tumor regression
.
J Exp Med
2019
;
216
:
2394
411
.
16.
Dhupkar
P
,
Gordon
N
,
Stewart
J
,
Kleinerman
ES
.
Anti-PD-1 therapy redirects macrophages from an M2 to an M1 phenotype inducing regression of OS lung metastases
.
Cancer Med
2018
;
7
:
2654
64
.
17.
Trujillo
JA
,
Sweis
RF
,
Bao
R
,
Luke
JJT
.
Cell-inflamed versus non-T cell-inflamed tumors: a conceptual framework for cancer immunotherapy drug development and combination therapy selection
.
Cancer Immunol Res
2018
;
6
:
990
1000
.
18.
Ayers
M
,
Lunceford
J
,
Nebozhyn
M
,
Murphy
E
,
Loboda
A
,
Kaufman
DR
, et al
.
IFN-gamma-related mRNA profile predicts clinical response to PD-1 blockade
.
J Clin Invest
2017
;
127
:
2930
40
.
19.
Wong
PF
,
Wei
W
,
Smithy
JW
,
Acs
B
,
Toki
MI
,
Blenman
KRM
, et al
.
Multiplex quantitative analysis of tumor-infiltrating lymphocytes and immunotherapy outcome in metastatic melanoma
.
Clin Cancer Res
2019
;
25
:
2442
9
.
20.
Neubert
NJ
,
Schmittnaegel
M
,
Bordry
N
,
Nassiri
S
,
Wald
N
,
Martignier
C
, et al
.
T cell-induced CSF1 promotes melanoma resistance to PD1 blockade
.
Sci Transl Med
2018
;
10
:
eaan3311
.