Download as pdf or txt
Download as pdf or txt
You are on page 1of 20

Peran Sel pada Sistem Imun dalam Jaringan Payudara dan Imunoterapi

sebagai Pilihan Terapi pada Kanker Payudara


(The Role of Immune Cells in Breast Tissue and Immunotherapy for the Treatment of Breast Cancer)

Disusun untuk memenuhi tugas dan melengkapi persyaratan dalam menempuh Program
Pendidikan Magister Biomedik

Widoasti Putri Utami


22010120410005

PROGRAM PENDIDIKAN MAGISTER BIOMEDIK


FAKULTAS KEDOKTERAN
UNIVERSITAS DIPONEGORO
SEMARANG
2020
Review

The Role of Immune Cells in Breast Tissue and


Immunotherapy for the Treatment of Breast
Cancer
Stephanie L. Goff, David N. Danforth
Abstract
Immune cells are present in normal breast tissue and in breast carcinoma. The nature and distribution of the immune
cell subtypes in these tissues are reviewed to promote a better understanding of their important role in breast cancer
prevention and treatment. We conducted a review of the literature to define the type, location, distribution, and role of
immune cells in normal breast tissue and in in situ and invasive breast cancer. Immune cells in normal breast tissue are
located predominantly within the epithelial component in breast ductal lobules. Immune cell subtypes representing
innate immunity (NK, CD68þ, and CD11cþ cells) and adaptive immunity (most commonly CD8þ, but CD4þ and CD20þ
as well) are present; CD8þ cells are the most common subtype and are primarily effector memory cells. Immune cells
may recognize neoantigens and endogenous and exogenous ligands and may serve in chronic inflammation and
immunosurveillance. Progression to breast cancer is characterized by increased immune cell infiltrates in tumor pa-
renchyma and stroma, including CD4þ and CD8þ granzyme Bþ cytotoxic T cells, B cells, macrophages and dendritic
cells. Tumor-infiltrating lymphocytes in breast cancer may serve as prognostic indicators for response to chemo-
therapy and for survival. Experimental strategies of adoptive transfer of breast tumor-infiltrating lymphocyte may allow
regression of metastatic breast cancer and encourage development of innovative T-cell strategies for the immuno-
therapy of breast cancer. In conclusion, immune cells in breast tissues play an important role throughout breast
carcinogenesis. An understanding of these roles has important implications for the prevention and the treatment of
breast cancer.

Clinical Breast Cancer, Vol. -,


No. -, --- Published by Elsevier Inc. This is an open access article under the CC BY-NC-ND
license (http://creativecommons.org/licenses/by-nc-nd/4.0/).
Keywords: Immunology, Breast ductal cells, Intraepithelial lymphocytes, Tumor immunology, Tumor-infiltrating lymphocytes

Introduction factors, cytokines, chemokines, and prostaglandins. Among the


Breast cancer is the most common malignancy in women, with most important of these elements are the immune cells, which are
over 316,000 cases annually in the United States.1 Most breast considered to play a critical role throughout the course of breast
cancers arise in the breast milk ducts, either as ductal carcinoma-in- carcinogenesis, beginning in normal breast tissue with immuno-
situ (DCIS) or as invasive ductal carcinoma. Breast cancer develops surveillance and continuing through primary and metastatic breast
through the accumulation of mutational changes, most commonly cancer. The ductal cellular layer in the normal breast contains a
in the ductal epithelium. The development and progression of these significant immune cell population consisting of CD8þ and CD4þ
changes can be influenced by multiple elements in the ductal T cells, B cells, dendritic cells, macrophages, NK (natural killer)
microenvironment, both cellular such as immune cells, adipocytes, cells, and other immune cell subtypes.2-4 Together, these immune
fibroblasts, and the microbiome, and soluble including growth cells provide important innate and adaptive immunity to the
epithelial layer for both protection against exogenous and endoge-
Surgery Branch, Center for Cancer Research, National Cancer Institute, National In-
nous agents, and elimination of transformed cells. An understanding
stitutes of Health, Bethesda, MD of the immune cellular population in normal breast tissue may thus
Submitted: Apr 3, 2020; Revised: Jun 10, 2020; Accepted: Jun 29, 2020 have important implications for breast cancer prevention, improved
methods of risk assessment, and the regulation of breast
Address for correspondence: David N. Danforth, MD, Surgery Branch, CCR, NCI, carcinogenesis.
NIH, Bethesda, MD 20892 Progression through the carcinogenic pathway from normal
E-mail contact: david_danforth@nih.gov
breast tissue to breast cancer is accompanied by quantitative and

-1
1526-8209/Published by Elsevier Inc. This is an open access article
under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/).
https://doi.org/10.1016/j.clbc.2020.06.011 Clinical Breast Cancer Month 2020
Role of Immune Cells
qualitative changes in the nature and location of the immune cell studies have indicated that hormone-related pathways are highly
population, including an increase in immune cell content in both enriched in the TDLU, including various hormone-related genes that
the parenchymal and stromal compartments. The immune cell are associated with breast carcinogenesis. It has been proposed that the
infiltration in breast cancer consists of multiple cellular subtypes, imbalanced hormone-reactions may result in the early onset of
including CD3þ (CD4þ and CD8þ) cells, B cells, monocytes/ neoplastic transformation that occurs mostly in breast TDLUs.12 The
macrophages, dendritic cells, and NK cells.4-6 The presence of breast ducts are surrounded by stroma that consists of extracellular
multiple immune cell subtypes in both the parenchyma and stroma matrix (ECM), fibroblasts, adipocytes, immune cells, microbiome,
places these cells in close proximity to tumor cells and other cells in and blood vessels. The breast ducts and stroma together comprise the
the microenvironment, and allows these cells to influence tumor breast microenvironment.
growth in multiple ways, either directly through CD4þ and CD8þ
cell-mediated cytotoxicity, or indirectly through immunosuppres- Distribution of Immune Cells within Normal Breast Tissue. Normal
sive or immunostimulatory actions from secreted cytokines, growth breast tissue contains immune cells of both myeloid (monocytes,
factors and other agents. Their distribution and characteristics may macrophages, dendritic cells) and lymphoid (T lymphocytes and B
also vary according to the subtype of breast cancer, their estrogen lymphocytes) lineage. Immune cells in normal breast tissue are
responsiveness, their mutational load, and the formation of tertiary predominantly localized to the lobules rather than the stroma and
lymphoid structures. Importantly, detailed analysis of tumor- fat (Figure 1),2 comprise cells of both the innate and adaptive im-
infiltrating lymphocytes (TILs) has demonstrated prognostic prop- mune systems, and thus may potentially provide protection against
erties for these cells,7-10 and collecting and administering TILs to bacterial and other pathogens as well as immune surveillance and
patients has resulted in durable complete regression of solid tumors elimination of epithelial cells with mutational changes. Immune
as well providing knowledge to allow development of cellular cells identified in early studies were predominantly lymphocytes,13
therapy with gene-modified T-cell receptors (TCRs) for treatment including CD8þ and CD4þ T lymphocytes,14,15 and macro-
of breast and other cancers.11 Together these findings indicate an phages.13-15 Five recent studies have further characterized this
important role for immune cells in the breast throughout breast intraepithelial immune cell population in normal breast tissue.2-
carcinogenesis, and significant changes in these cells during the 4,6,16
Overall, immune CD45þ cells are prominent among the
course of these events. To clarify these significant properties, we ductal epithelial cells. CD8þ T cells are the most common cells in
have conducted a review of the literature of the nature and char- all series, and in the study by Degnim et al,2 CD8þ T cells and
acteristics of ductal immune cells in normal breast tissue, of TILs in CD11cþ (dendritic) cells were present in virtually all lobules and
breast cancer, and the efforts to translate the latter findings into among the most numerous across lobules, while CD68þ cells
both valuable prognostic indicators and successful immunothera- (macrophages/monocytes) were also numerous across lobules;
peutic treatment options for breast cancer, including ongoing and CD4þ and CD20þ cells were less frequent. They also found both
innovative trials at our institution. dendritic cells and CD8þ cells were consistently observed in inti-
mate association with the epithelium of lobular acini and were
Materials and Methods primarily located at the basal aspect of the epithelium. The
A literature search was conducted through PubMed and cross- anatomical distribution of immune cells in normal breast tissue and
references to identify publications describing the nature and distri- their relationship to epithelial cells is depicted in Figure 1. The
bution of immune cells in normal breast tissue and in breast cancer. presence of the immune cells in the in TDLU places them at the
Normal breast tissue may include tissue from women at normal risk predominant site of the origin of breast cancer - TDLU epithelial12;
(such as from reduction mammoplasty), as well as from the this close proximity provides an important opportunity for the
contralateral breast or from tissue adjacent to a breast cancer in immune cells to influence the behavior of epithelial cells. Zumwalde
women with a breast cancer. For studies of breast cancer, emphasis et al,3 using flow cytometry of breast tissue organoids from reduc-
was placed on immune cells in the primary tumor. tion mammoplasty specimens, reported that CD8þ cells represented
75% of CD3þ cells; essentially all of the CD8aþ cells expressed
Results and Discussion CD8þb, indicating they were distinct from intestinal intraepithelial
Immune Cells in the Normal Breast cells, which are CD8þaa.6 Ruffell et al6 examined normal breast
Breast Ductal Anatomy. The ductal anatomy and its relationship to tissue either adjacent to breast cancer or from prophylactic mas-
the distribution of immune cells in the normal breast is illustrated in tectomy specimens and found CD3þ (CD4þ and CD8þ) cells were
Figure 1. The principle unit in the breast ductal system is the acini, the most common, while myeloid-lineage cells including macro-
which drains into an intralobular and then the extralobular duct; phages, dendritic cells and neutrophils were also prominent. The
together, these structures constitute the terminal ductal lobular unit distribution of immune cell types thus appears to be quite consistent
(TDLU). The intralobular terminal ducts are lined by cuboidal across multiple types of normal breast tissue. Interestingly, in the
epithelium, and the extralobular terminal duct and major ducts are study of normal breast organoids by Zumwalde et al, the authors
lined by pseudostratified columnar epithelium or a double layer of identified an important subtype of CD3þ cells, gd T cells. The
cuboidal epithelium. The TDLU is considered to be the predominant TCRs of these cells consists of g and d chains rather than the
site of the origin of breast cancer.12 The vast majority of breast cancers conventional a and b chains, and the receptor is considered to act as
are carcinomas that arise in the epithelium and include in-situ and a pattern recognition receptor and a bridge between innate and
invasive ductal and invasive lobular carcinoma. Gene expression adaptive immune responses.17 These cells possess cytotoxic activity,

2 - Clinical Breast Cancer Month 2020


Stephanie L. Goff, David N. Danforth
Figure 1 Nature and Distribution of Immune Cells in Normal Breast Tissue. A Variety of Immune Cells are Present in Normal Breast
Tissue, including Both Lymphocytes and Myeloid-Derived Cells. Immune Cells are Located Primarily Within the Ductal
Epithelium (Intraepithelial) but May be Present in the Stroma as Well (Not Depicted). The Stromal Extracellular Matrix (ECM)
Also Contains a Variety of Cell Types and Structures that May Interact With Immune Cells and the Epithelium. Adapted From
Degnim et al2

and serve to control the integrity of epithelium.18 The subtype intraepithelial lymphocyte population of the normal breast indicates
Vg2þ gd T cells was found to be consistently present in prepara- these cells have been antigen-activated, as well as indicating the
tions of mammary ductal epithelial organoids.3 presence of a dynamic immune network between ductal epithelium
and regional lymph nodes. Importantly, TEM may persist for
Immunogenicity of Immune Cells in Normal Breast Tissue. An considerable periods of time,19 and Hussein and Hassan5 have
important question is the immunogenicity of the immune cells in shown that the CD3þ population of cells in normal breast tissue are
normal breast tissue. It is noted that the immune cells in the breast granzyme Bþ cells and thus have cytotoxic functions, suggesting an
ducts are in proximity to draining lymph nodes in the ipsilateral important protective role for these intraepithelial immune cells.
axilla and mediastinum, and thus all of the principle cellular and There are several potential sources of antigens for activation of these
lymphatic components necessary for adaptive cellular immune CD8þ T cells, both exogenous (eg, viral, bacterial) and endogenous
response are present within the ductal system. In the study by (nuclear and cytosolic proteins, DNA, extracellular proteins).
Zumwalde et al3 further analysis of the CD8þ T cells indicated they Neoantigens, for example, could arise through mutational changes
were almost exclusively CD45ROþ/CD27 cells, and thus were in ductal epithelium from exposure to estrogens and other carcin-
effector memory T cells (TEM). The presence of TEM in the ogens.20 A recent review has demonstrated that normal breast tissue

Clinical Breast Cancer Month 2020 -3


Role of Immune Cells
at normal risk for breast cancer (such as from a reduction mam- diffusion and permeability through the ECM, and diffusion
moplasty) as well as normal breast tissue at increased risk for breast restrictions could foster tumorigenesis over short- and long-term
cancer (such as normal breast tissue adjacent to breast cancer) scales by restricting clearance of factors secreted by enveloped
contain widespread genomic changes including loss of heterozy- cells. Fibroblasts, a major component of the ECM, can be reprog-
gosity, small segmental deletions, DNA methylation, and telomere rammed into cancer-associated fibroblasts by cytokines and growth
shortening.21,22 This could provide an excellent source of neo- factors secreted by transformed epithelial cells.29 Cancer-associated
antigens through associated frameshift or other mutational changes. fibroblasts can be activated to promote tumor initiation, remodel-
In addition, the release of damage associatede or pathogen- ing of the ECM, and modulation of immune cells.29 Adipocytes are
associated molecular patterns from injured or dying epithelial or also important components of the ECM and are in close proximity
microbial cells may be recognized by macrophages (also present to epithelial cells, tumor and immune cells. Breast cancer cells
among ductal intraepithelial immune cells), eliciting release of induce production of endocrine and paracrine enzymes and bioac-
cytokines and chemokines, production of chronic inflammation, tive lipids by adipocytes, which in turn drive increased growth and
and production of reactive oxygen species and reactive nitrogen invasion of tumor cells.30 Leptin is also an important secretory
intermediates, which may then cause mutations in neighboring product of adipocytes, which can stimulate production of several
epithelial cells.23-25 As noted by Degnim et al,2 the consistent proinflammatory factors including interleukin (IL)-1, IL-6, and
presence of CD8þ cells and dendritic cells, interspersed within the tumor necrosis factor (TNF) a.31 Obesity is an important risk factor
breast epithelium, strongly suggests a role for antigen presentation for breast cancer in postmenopausal women and may promote
and immune effector function, as well as stress response and procarcinogenic processes in adipocytes. Several factors, including
maintenance of epithelial integrity. In a subsequent study by Deg- leptin, TNFa, IL-6, and resistin, are secreted by normal adipocytes,
nim et al,16 they compared the distribution of immune cells in the and are increased with obesity and have procarcinogenic effects.32
lobules of women with benign breast disease to breast samples from These in turn can modify the immune system and promote tu-
women with no known breast abnormalities and found benign mor growth.33 Together, these findings indicate a diverse popula-
breast disease lobules showed greater densities of CD8þ T cells, tion of cells within the ductal epithelial layer and microenvironment
CD11cþ dendritic cells, CD20þ B cells, and CD68þ macrophages of normal breast tissue that interact to play a major role in early
compared to the normal controls.16 The authors concluded that breast carcinogenesis.
elevated infiltration of both innate and adaptive immune effectors in
benign breast disease tissues suggests an immunogenic microenvi- Estrogenic Effects on Normal Breast Immune Cells. Estrogen is the
ronment. Finally, it has also been shown that many of the muta- predominant hormone in women during both premenopausal and
tional changes observed in normal high risk breast tissue are also postmenopausal time periods. Estrogens play an important role
present in the adjacent breast cancer, thus providing a potentially throughout breast carcinogenesis, from influencing risk, to the
comparable immunogenic environment for immune cells.26 Azizi development of contralateral breast cancer, to the progression of
et al,4 in a study of normal breast tissue from prophylactic mas- primary breast and metastatic breast cancer. These actions are
tectomy tissue, observed a large number of normal breast tissue achieved primarily through estrogenic actions on breast epithelial
resident immune cell states, including 13 myeloid and 19 T-cell cells, however there is important evidence that immune cells contain
clusters that were not observed in circulation or in the secondary estrogen receptors (ERs) and are regulated by estrogens as well. This
lymphoid tissue. The set of clusters found in normal breast tissue would provide an important opportunity for estrogens to influence
cells represented a subset of those observed in the breast cancers, immunosurveillance, as well as the development and progression of
supporting the widespread immunogenic environment of these breast cancer. Estrogenic actions result from binding to cytoplasmic
tissues. receptors ERa and ERb and activation of genomic pathways,
although estrogens may also activate growth factor receptor activity
Intraepithelial Immune Cells and the Microenvironment. The with signaling through nongenomic pathways by ligand-
immune cells within the intraepithelial layer are also surrounded by, independent ERa signaling involving binding of E2 to a
and may be influenced by, major components of the microenvi- membrane-anchored receptor, the G proteinecoupled estrogen re-
ronment including the ECM and the interstitial matrix (Figure 1). ceptor 1 (GPER1), with subsequent activation of G proteins.34
The ECM consists of the ductal and endothelial basement mem- Immune cells of both the innate (dendritic cells, macrophages,
branes, and the interstitial matrix is comprised of connective tissue neutrophils) and adaptive (CD4þ, CD8þ, B cells) cell types, and in
and cellular components (fibroblast, adipocyte, endothelial, in- both normal tissue and in breast cancer, have been shown to possess
flammatory).27 A dynamic interaction is thought to exist between ERs and to be responsive to estrogens.35-37 The response of immune
the multiple components of the microenvironment, which serves to cells to the estrogenic effects may include altered immune cell
maintain and promote the contribution of each component. The proliferation, secretion of cytokines, chemokines, and growth
components of the microenvironment are also considered to play a factors.35,38,39
significant role during breast carcinogenesis. With progression of the In breast cancer the distribution of immune cell types may vary
epithelial cells in the carcinogenic pathway, there is increasing in- according to tumor subtype as well as between stroma and tumor
fluence of these components, including the ECM, fibroblasts, and parenchyma (see below). The distribution of immune cells between
adipocytes on transformed cells. As Ghajar and Bissell28 have stroma and parenchyma/ductal epithelium provides an opportunity
pointed out, simple changes in ECM composition can alter for these cells, after estrogen stimulation, to influence multiple

4 - Clinical Breast Cancer Month 2020


Stephanie L. Goff, David N. Danforth
components of the microenvironment including other immune negativity, high Ki-67 proliferation index, and p53 over-
cells, epithelial cells, fibroblasts, adipose cells and endothelium. expression.44 Kim et al44 found the high infiltration of FOXP3þ
Ultimately, the response of immune cells will also be influenced by TIL and the presence of PD-L1þ immune cells were associated with
cell density, estrogen levels, ECM composition, blood supply, and tumor recurrence in patients with pure DCIS. Pure DCIS associated
the specific ER stimulated.38,40 Although estrogens are probably the with higher numbers of B lymphocytes has also been shown to be
major steroid that influences cell behavior, it is recognized that these associated with a shorter recurrence-free interval (P ¼ .04).53 Bates
cells may also contain receptors for other steroids, including pro- et al52 observed high numbers of FOXP3þ regulatory T cells
gesterone, glucocorticoids, and androgens,38 and respond and be identified in patients with DCIS at increased risk of relapse (P ¼
influenced by these hormones as well. Together, the cellular .04). CD8þ HLA-DRþ T cells, CD8þ HLA-DR T cells, and
responsiveness of immune cells to estrogens (and other hormones) CD115þ cell populations have also shown to be associated with risk
further clarifies events regulating progression in carcinogenesis, as of recurrence.48 Together, these findings indicate a prominent
well as suggest an additional potentially important role for anties- presence of TIL, which may play an important immunosuppressive
trogen therapy. role in DCIS.

Immune Cells in Ductal carcinoma in situ (DCIS) Immune Cells in Invasive Carcinoma of the Breast
DCIS is an important precursor lesion for invasive ductal carci- The immune cell content of breast tissue increases progressively
noma and accounts for 16% of all breast cancers annually in the from normal breast tissue to breast cancer.4-6,54,55 This is best
United States.1 DCIS contains, TILs and multiple studies have illustrated by studies comparing the immune cell distribution in
examined TIL in DCIS both according to cell density, according to breast cancer and related normal breast tissue. In a study of mas-
the presence of aggressive features, and according to cell type. DCIS tectomy specimens utilizing immunohistochemistry (IHC) and flow
is characterized by an increase in the density and extent of immune cytometry, Ruffell et al6 observed that breast cancer tissues con-
cell infiltration compared to normal breast tissue.5,41-43 The im- tained infiltrates dominated by CD8þ and CD4þ lymphocytes,
mune cell infiltrate in DCIS is also increased in more aggressive with minor populations of NK cells and B lymphocytes, whereas in
lesions, being greater in high-grade tumors,41-46 ER-negative tu- the normal breast tissue myeloid-lineage cells including macro-
mors,41,44 and human epidermal growth factor receptor 2 (HER2)- phages, mast cells, and neutrophils were more evident. A similar
positive or triple-negative breast cancer (TNBC),43,44,46,47 and also immune profile was observed in breast tissues obtained in prophy-
correlates with tumors with necrosis.5,43,44 High-grade DCIS has lactic mastectomy tissue. Activated T lymphocytes also predomi-
significantly higher percentages of FOXP3þ cells, CD68þ and nated in tumor tissue, with both CD4þ and CD8þ T cells
CD68þPCNAþ macrophages, human leukocyte antigen (HLA)- displaying increased expression of activation markers CD69 and
DRþ cells, CD4þ T cells, CD20þ B cells, and total TILs compared HLA-DR. They concluded these findings indicated a shift within
to nonehigh-grade DCIS.5,48 Hussein and Hassan5 noted a marked tumors toward a Th2-type response in breast cancer characterized
increase in the density of mononuclear inflammatory cell infiltrates, by the increased presence of B cells and CD4þ T cells, in com-
including CD20þ, CD68þ, CD3þ and granzyme Bþ cytotoxic T parison with normal breast tissue.6 At the transcriptional level, Azizi
cells from normal breast tissue to DCIS to invasive breast cancer. et al4 compared by singe cell sequencing the immune cell distri-
On average, ER DCIS has been found to contain higher numbers bution in breast cancer with that of normal adjacent breast tissue.
of all TIL subsets than ERþ DCIS, and ERþ DCIS was more likely They found a significant increase in the diversity of cell states, re-
to have to have a high CD8/FOXP3 ratio (> 4) than ER DCIS.41 flected in an increase in variance of gene expression, among T cells,
Thompson et al41 observed that CD3þ T cells predominated across monocytes, and NK cells in tumor compared to normal tissue. This
all DCIS subtypes at all ages, with slightly more CD4þ T cells than suggested that the increased heterogeneity of cell states and marked
CD8þT cells on average. CD20þ B cells were the next most phenotypic expansions found within the tumor were likely due to
common TILs, followed by FOXP3þ T regulatory cells (Tregs). more diverse local microenvironments within the tumor.4 Two
Others have observed B cells in DCIS and in invasive carcinoma, small studies utilizing high-throughput sequencing of the TCR beta
which were typically found in perivascular locales clustering in ag- chain to characterize diversity of the T-cell infiltrate have been
gregates with T cells, forming ectopic follicles,49,50 and indicating performed. Beausang et al56 demonstrated that there are unique
that the presence of B cells in neoplastic mammary tissue is the compartments of enriched clonotypes in tumor and tumor-adjacent
result of chronic activation rather than nonspecific chemo- normal breast tissue, as well as identifying sequences shared among
attraction.49 There is evidence also that TILs in DCIS are immu- patients and unlikely to be involved in specific tumor recognition.
nosuppressive. Programmed death ligand 1 (PD-L1) is an Park et al57 studied tumors before and after neoadjuvant therapy,
immunosuppressive surface ligand on lymphocytes; Thompson and demonstrated a clonal expansion and decrease in diversity in
et al41 demonstrated that 81% of DCIS lesions contained PD-L1þ those patients with pathologic complete response (pCR) to therapy.
TILs. They considered this to suggest an active immune response The complexity of the composition of immune cells in breast
within DCIS and supported TIL expression of PD-L1 as a marker cancer reflects a cross-talk between components of the innate im-
of downregulation of the body’s immune response within DCIS. mune response as it regulates the tumor microenvironment and the
FOXP3þ Treg cells are another important immunosuppressive cell polarity of the adaptive immune response within that tumor.49
that are increased in DCIS,41,44,48,51,52 and are associated with high Lymphocytes of the myeloid lineage, ie, tumor-associated macro-
nuclear grade, comedo-type necrosis, hormone receptor (HR) phages, myeloid-derived suppressor cells, and dendritic cells, can

Clinical Breast Cancer Month 2020 -5


Role of Immune Cells
create immunostimulatory or immunosuppressive milieus that affect luminal B (17.0%), or TNBC (14.5%). This, however, did not
the fate of T cells capable of homing to the tumor. In turn, apply to CD3þ or CD45ROþ type cells, and there was no signif-
macrophage polarization can be driven to M1 or M2 phenotypes by icant correlation between CD45ROþ TILs and clinicopathologic
the cytokines expressed by cytotoxic or regulatory T cells.49,58 parameters. Their study was conducted in the Sudan, and their
Within breast cancer, the distribution of immune cells may also finding that a higher CD8þ content was observed in HER2/neu-
differ between the tumor parenchyma and stroma. In a study of positive breast cancer is in agreement with the other studies, and
breast cancer mastectomy specimens, immune cells were delineated suggests that the biological factors regulating TIL distribution ac-
in the tumors and surrounding stroma by IHC staining to identify cording to subtype in North Africa may be similar to those in the
CD3 (T cells), CD20 (B cells), CD68 (macrophages), and gran- United States.
zyme B (cytotoxic subset of CD3þ cells).5 The breast samples
ranged the spectrum of breast disease: normal, benign proliferative Mutational Load of Breast Cancer Cells. The finding that immune
disease (usual ductal hyperplasia), DCIS, and invasive ductal car- cell types in breast cancer varied according to breast cancer subtype
cinoma. Although there was a progressive increase in all cell types in suggested the mutational load of breast cancer subtypes may also
both the parenchyma and stroma moving from normal mammary vary. When this was studied it was found there are also striking
tissue to ductal carcinoma, the most striking difference from benign differences in the mutational load of breast cancer between sub-
proliferative disease to DCIS to invasive cancer was the influx of types. The genomic characteristics of each subtype has recently been
CD3þ cells in the stroma of the latter two tissue types: average 4.2 analyzed and presented in detail in an analysis of cancer-adjacent
cells/mm2 versus 46.6 cells/mm2 versus 77.0 cells/mm2, respectively breast tissue from The Cancer Genome Atlas Network.22 The
(Figure 2). Interestingly, increases in the cytotoxic subset (granzyme findings for different subtypes can be summarized as follows: the
B) were restricted to immune cells in the parenchyma: 16.3 versus overall mutation rate was lowest in luminal A subtype and highest in
0.7 cells/mm2 in the parenchyma versus stroma of invasive cancer.5 the basal like and HER2 enriched (HER2E) subtypes. The luminal
Solinas et al,59 in their review, similarly found that the incidence of A subtype harbored the most significantly mutated genes, with the
stromal TIL (sTIL) ranged from 15% to 25%, whereas intratumoral most frequent being PIK3CA (45%), followed by MAP3K1,
TIL ranged from 5% to 10%. As will be discussed below, sTILs also GATA3, TP53, CDH1 and MAP2K4. Twelve percent of luminal A
have important prognostic value. tumors contained likely inactivating mutations in MAP3K1 and
MAP2K4. TP53 and PIK3CA mutations were the most common in
Immune Cell Distribution According to Breast Cancer Subtype. The Luminal B cancer (29% each); this contrasted with basal-like tu-
distribution of TIL varies quantitively and qualitatively according to mors where TP53 mutations occurred in 80% but PIK3CA muta-
subtype in breast cancer. This has been demonstrated in several tions were absent or rare. Ten percent of sporadic breast cancer may
series. Stanton et al,54 in a review of 13,914 patients, found a have a strong germline contribution. Luminal tumors were mostly
median of 20% of patients with TNBC demonstrated lymphocyte- diploid whereas luminal B tumors were mostly aneuploid.
predominant breast cancer (LPBC;  50%-60% lymphocytic Regarding HER2þ tumors, there are at least two types of clinically
infiltrate) at the time of diagnosis compared to 16% of HER2þ defined HER2 tumors, luminal messenger RNA (mRNA)-subtype/
tumors and 6% of HRþ cancers. A median of 60% of TNBC HER2þ tumors, and HER2E-mRNA subtype (HER2 enriched). A
samples had infiltrating CD8þ T cells in contrast to only 43% of comparison identified 302 differentially expressed genes. The
HRþ tumors. TNBC tumors were also more likely to have HER2E mRNA subtype typically showed high aneuploidy, the
FOXP3þ infiltrates than the HRþ subtype. Their findings indicated highest somatic mutation rate, and DNA amplification of other
that HR-positive disease may be the least immunogenic of the potential therapeutic targets including fibroblast growth factor re-
common breast cancer subtypes. Liu et al,60 in another study, re- ceptors, epidermal growth factor receptor, CDK4, and cyclin D1.
ported a high density infiltration of Treg and FOXP3PþTumor, but TP53 mutations were significantly enriched in HER2E or ER
not CD8þTumor, correlated significantly with HER2 overexpression. tumors whereas GATA3 mutations were only observed in luminal
Increased infiltration of Tregs and cytotoxic lymphocytes was subtypes or ERþ tumors. The basal-like subtype included TNBC
significantly more common in those tumors with unfavorable his- (75%) as well as other mRNA subtypes (25%), and showed basal-
tologic features, including high histologic grade and negative ER like tumors with a high frequency of TP53 mutations (80%).
and progesterone receptor status. They proposed that further studies PIK3CA was mutated in 9% of cases; however, inferred PI(3)K
to explore the functional status and action modes of cytotoxic pathway activity, whether from gene, protein, or high PI(3)K/AKT
lymphocytes and Tregs in different tissue locations and in different pathway activities was highest in basal-like cancers. Expression
breast cancer subtypes will lead to a better understanding of the features showed high expression of genes associated with cell pro-
nature of breast carcinoma immunity. Denkert et al61 observed the liferation. Although chromosome 8q24 is amplified across all sub-
percentage of tumors with high TILs was higher in TNBC (30%) types, high MYC activation seems to be a basal-like characteristic.
and HER2-positive breast cancer (19%) than in lumi- Other major genomic changes include ATM mutations (3%),
naleHER2enegative tumors (13%) (P < .0001). Their data sup- BRCA1 (30%) and BRCA2 (6%) inactivation, RB1 loss (20%), and
ported the hypothesis that breast cancer is immunogenic and might cyclin E1 amplification (9%). Together, these findings indicate a
be targetable by immune-modulating therapies. Mohamed et al62 significant mutational burden across breast cancer subtypes. This
reported HER2/neu-positive breast cancer had a significantly may have important clinical, therapeutic, and biological implica-
higher CD8þ content (26.1%) compared to luminal A (13.5%), tions. A high mutational burden would be associated with increased

6 - Clinical Breast Cancer Month 2020


Stephanie L. Goff, David N. Danforth
Figure 2 Immune Cells in Breast Proliferative Disease and Breast Cancer. Immune Cells, including CD68D (Macrophages/Monocytes),
CD3D (CD4D, CD8D), and Cytotoxic Lymphocytes (CTL) are Present in Breast Proliferative Disease and Increase in the
Parenchyma and Stroma With Progression to Breast Cancer. Relative Increases for Each Cell Type, With Benign Proliferation
as a Baseline, are Shown on a Logarithmic Scale in the Graphs Below the Figure. (Adapted from Data from within Hussein
and Hassan5)

genomic instability and neoantigen development, increased cell adaptive immunity and T-cell cytotoxicity.67 In the study by
injury and death, and development of chronic inflammation. At the Martinet et al,67 high densities of tumor HEVs independently
same time altered secretion of secretory products could influence the conferred a lower risk of relapse and significantly correlated this
activity, distribution, and interaction of associated immune and activity with longer metastasis-free, disease-free, and OS rates.
other cell types. Together, the presence and characteristics of TLS have important
clinical implications: their presence in poor-prognosis tumors may
Nature of Immune Cells in Tertiary Lymph Structures in Breast be important both for selection of therapy and entry into clinical
Cancer. Tertiary lymph structures (TLS) (aka tertiary lymph organs) trials. The identification of an organized immune cell collection
are ectopic lymphoid organs that develop at sites of chronic with effector memory cells in and adjacent to breast cancer and
inflammation including autoimmune diseases, infection, and tu- capable of antitumor activity may encourage efforts to promote its
mors. TLS have a defined structure, are comprised of multiple cell development and expansion in these tumors. The presence in TLS
types including both innate (dendritic, macrophages, neutrophils) of HEVs that can enhance movement of lymphocytes into these
and adaptive (B cells, T cells) as well as plasma cells and high tumors also encourages efforts to identify agents that can promote
endothelial venules (HEV).63,64 TLS are believed to be the site of the activity of these vessels.
immune response activation against tumor by recruiting and acti-
vating TIL.65 The TLS may be within the tumor or in a peritumoral Immune Cells as a Prognostic Biomarker. With renewed interest in
location. TLS are generally associated with tumors with a more immune cells, investigators have interrogated large prospectively
aggressive phenotype such as high-grade tumors, TNBC, and collected patient samples associated with seminal clinical trials to
HER2þ tumors.62,65,66 Liu et al65 reported TLS were associated evaluate the role of immune cells as prognostic markers. The first of
with higher tumor grade, apocrine phenotype, necrosis, extensive these studies was reported by Denkert et al10 after analysis of the
in-situ component, lymphovascular invasion, high TIL, HR nega- GeparDuo and GeparTrio trial cohorts. Pretreatment core biopsy
tivity, HER2 positivity, and c-kit expression. A favorable impact of samples of tumor and stroma were analyzed for lymphocytic infil-
TLS density on overall survival (OS) and disease-free survival (DFS) trate evaluated by routine hematoxylin and eosin staining. Two
of patients has been observed.62,63,66-71 An important component of categories of immune infiltrate were defined: iTu-Ly (intratumoral
TLS are HEV. These are specialized postcapillary venules found in lymphocytes) in direct contact with tumor cells or within tumor cell
lymphoid tissues that support high levels of lymphocyte extravasa- nests and str-Ly (stromal lymphocytes) without direct contact with
tion from the blood.72 High densities of tumor HEVs correlated tumor cells. LPBC was defined as tumors with > 60% of either
with increased naive, central memory and activated effector memory iTu-Ly or str-Ly and represented 11% of the study population.
T-cell infiltration and upregulation of genes related to T-helper 1 LPBC demonstrated an increased incidence of pCR (10/24, 41.7%)

Clinical Breast Cancer Month 2020 -7


Role of Immune Cells
when compared to those tumors with no lymphocytic infiltrate (1/ and 32% in low samples, but still associated with longer DFS.61
36, 2.8%) or focal infiltrate (17/158, 10.8%). This was also Together, these findings support a prognostic role of TILs in
demonstrated in gene expression data of the same cohort.10 In an breast cancer. The prognostic significance may correlate inversely
analysis of the BIG 02-98 multi-institutional randomized phase 3 with tumor HR status, suggesting there may also be a prominent
trial, increasing lymphocyte infiltration was associated with hormonal influence on these lymphocytes in HRþ tumors.
improved prognosis in the subgroup of node-positive patients with Lastly, the effect of neoadjuvant chemotherapy on immune cell
TNBC. LPBC represented only 10.6% of the TNBC cohort using a distribution has also been examined. In the study of Ruffell et al6
threshold of 50%, but prognosis improved linearly with each 10% their findings suggested that neoadjuvant chemotherapy further
increase in lymphocyte infiltration.8 altered the complexity of the immune microenvironment of the
Tumor samples from patients in the Eastern Cooperative residual tumors. They found residual tumors treated with
Oncology Group (ECOG) 1199 and 2197 trials have been analyzed neoadjuvant chemotherapy contained increased percentages of
for the presence of intraepithelial TIL or sTIL lymphocytes.9 infiltrating myeloid cells, accompanied by an increased CD8/CD4
Overall, the median intraepithelial TIL score was 0%, and LPBC T-cell ratio and higher numbers of granzyme Beexpressing cells,
was again a minority of cases (4.4%), although over 80% of cancers compared to tumors not treated with neoadjuvant chemotherapy.
had a sTIL score > 10. Higher sTIL scores were associated with The effect of chemotherapy has also been examined by García-
better prognosis; for every 10% increase in sTILs, a 14% reduction Martínez et al,77 who evaluated tumors using IHC to define CD3þ
of risk of recurrence or death was noted (P ¼ .02), and an 18% cells, subsets of CD4þ and CD8þ cells, and the presence of B cells
reduction of risk of distant recurrence (P ¼ .04), and 19% reduc- and monocytes before and after neoadjuvant chemotherapy.77 They
tion of risk of death. However, in Liu et al,60 the effects of observed a decrease in the number of CD3þ cells after chemo-
CD8þTumor infiltration, and the ratio of CD8þTumor/ therapy and a decrease in the CD4:CD8 ratio. They concluded that
FOXP3PþTumor on OS and progression-free survival, were not an IHC-based profile of immune cell subpopulations in breast
significant, whereas patients with high FOXP3þTumor had a cancer is able to identify a group of tumors highly sensitive to
significantly shorter OS (P ¼ .007) and progression-free survival neoadjuvant chemotherapy.
(P ¼ .003) both in the ERþ and ER groups. Interestingly, recent
studies have indicated that the spatial location and organization of Immune-Based Therapies for Breast Cancer. The majority of the
CD8þ TILs within the tumor may be important in relation to studies of lymphocytes in breast cancer has focused on character-
relapse-free survival.73 Egelston et al73 reported that the presence of izing the nature of the lymphocyte infiltrate, with some gene
islands of infiltrating CD8þ T cells was more significantly associated expression data that may hint at the functional role of these cells in
with relapse-free survival than CD8þ T-cell infiltration into either an anticancer response. Chemokines thought to be responsible for
tumor stroma or total tumor. The integrin CD103, a marker for lymphocyte migration and gene expression signatures associated
tissue resident memory T cells (TRM) appeared to mediate locali- with Type I effector responses have been shown to correlate with
zation into cancer islands within tumors. The TRM subset has been pCR.78,79 Programmed death receptor 1 (PD-1) and one of its li-
implicated in cancer surveillance of melanoma and lung cancer.74,75 gands (PD-L1) have also been correlated with higher pCR rate and
Savas et al76 utilized single-cell RNA sequencing techniques to improved prognosis in breast cancer.80 These molecules are part of
derive a CD8þ TRM signature from primary TNBC tumors; when the immune checkpoint pathway that limits T-cell response. PD-L1
applied to a bulk RNA data, the signature was associated with gene expression of immune cells in breast cancer has been shown to
improved patient survival. be positively associated with CD8þ and CD4þ memory-activated T
A review of 15 studies of TIL classification also identified dif- cells, but not with CD4þ memory resting or T-regulatory cells or
ferences in lymphocyte infiltration across tumor subtypes.54 LPBC other immune cell subpopulations.81 In other histologies, expres-
was more frequently identified in TNBC (20%) and HER2þ (16%) sion of PD-L1 may correlate with response to checkpoint inhibi-
specimens than in HRþ/HER2 tumors (6%). Efforts have been tion, but there are significant challenges to its use as a predictive
made to unify the classification of TILs within breast cancer spec- biomarker, highlighted by the KEYNOTE-86 trial of pem-
imens, with a focus on stromal lymphocytes as the most repro- brolizumab in patients with TNBC in which PD-L1 status was not
ducible and significant prognostic indicator of likelihood of the strongest discriminator between those with disease that did and
response.7 In an analysis by Denkert et al61 of pooled data from the did not respond to therapy.82 The overall response rate in that trial
German Breast Cancer Group, classification of tumors by degree of was 18.5%. Similarly modest results were seen in an ERþ popula-
stromal lymphocyte invasion allowed prediction of response to tion in KEYNOTE-28 with an overall response rate of 12%.83,84
neoadjuvant chemotherapy in all molecular subtypes, but differ- Checkpoint inhibitors have been combined with chemotherapy
ences in survival outcomes may suggest a different biology of the demonstrating improvement in median OS, more pronounced in
immunologic infiltrate in HRþ tumors. Although pCR was attained those patients with PD-L1þ tumors (Impassion130 trial),84 and an
in 28% of luminal HER2 tumors with high TILs (> 60%) increase in the pCR rate in women with TNBC (KEYNOTE-
compared to 6% and 11% in low ( 10%) and intermediate TILs, 522).85 A comprehensive review of current and proposed combi-
the presence of TILs was associated with shorter OS in that subtype. nation strategies identified 13 ongoing randomized phase 3 clinical
TNBC with high TILs demonstrated a higher pCR rate (50% high trials investigating the strategy in patients with breast cancer.86
vs. 31% intermediate and low) and was associated with longer DFS Given the presence of an immune infiltrate in stages as early as
and OS. The gains were more modest in HER2þ tumors with a DCIS, strategies to boost the immune response to potential tumor
48% pCR rate in high TILs specimens versus 39% in intermediate antigens by vaccination have been explored in prevention, adjuvant,

8 - Clinical Breast Cancer Month 2020


Stephanie L. Goff, David N. Danforth
neoadjuvant and metastatic settings.87 Additional efforts explore the use Disclosure
of tumor ablative strategies (ie, cryoablation, radiofrequency ablation, The authors have stated that they have no conflict of interest.
stereotactic radiation) for potential release of tumor-associated antigens
in conjunction with checkpoint inhibitor monoclonal antibodies to
increase response rates.88,89 A recent review highlighted the ways that the References
field of breast cancer immuno-oncology is building on information 1. American Cancer Society. Breast cancer facts and figures, 2017-2018, Available at:
https://www.cancer.org/content/dam/cancer-org/research/cancer-facts-and-statistics/
about TIL phenotypes to branch into different promising avenues of breast-cancer-facts-and-figures/breast-cancer-facts-and-figures-2017-2018.pdf.
research.90 Few groups have studied the phenotype and function of TILs Accessed: July 9, 2020.
2. Degnim AC, Brahmbhatt RD, Radisky DC, et al. Immune cell quantitation in
cultured from freshly resected breast cancer. In the Surgery Branch of the normal breast tissue lobules with and without lobulitis. Breast Cancer Res Treat
National Cancer Institute, TILs derived from fragments of metastatic 2014; 144:539-49.
3. Zumwalde NA, Haag JD, Sharma D, et al. Analysis of immune cells from human
breast cancer and cultured in IL-2 were predominantly CD4þ, in mammary ductal epithelial organoids reveals Vdelta2þ T cells that efficiently target
contrast to our experience with metastatic melanoma-derived TILs, breast carcinoma cells in the presence of bisphosphonate. Cancer Prev Res (Phila)
2016; 9:305-16.
which are predominantly CD8þ cells.91,92 Whole exome sequencing of 4. Azizi E, Carr AJ, Plitas G, et al. Single-cell map of diverse immune phenotypes in
the resected tumor identified candidate neoepitopes for further study, the breast tumor microenvironment. Cell 2018; 174:1293-308.
5. Hussein MR, Hassan HI. Analysis of the mononuclear inflammatory cell infiltrate
and TIL cultures were capable of identifying neoantigens processed by in the normal breast, benign proliferative breast disease, in situ and infiltrating
autologous antigen-presenting cells in vitro, in both class I and class II ductal breast carcinomas: preliminary observations. J Clin Pathol 2006; 59:972-7.
6. Ruffell B, Au A, Rugo HS, Esserman LJ, Hwang ES, Coussens LM. Leukocyte
restricted fashion.11,93 In a study of primary breast tumors, another composition of human breast cancer. Proc Natl Acad Sci U S A 2012; 109:
group used an agonistic antibody against 4-1BB/CD137 during the 2796-801.
7. Salgado R, Denkert C, Demaria S, et al. The evaluation of tumor-infiltrating
initial TIL cultures and were able to generate more robust CD8þ lymphocytes (TILs) in breast cancer: recommendations by an International TILs
populations. In two of these patient-derived cultures, the expanded Working Group 2014. Ann Oncol 2015; 26:259-71.
8. Loi S, Sirtaine N, Piette F, et al. Prognostic and predictive value of tumor-
CD8þ TILs were capable of mutation-specific recognition of class I infiltrating lymphocytes in a phase III randomized adjuvant breast cancer trial in
predicted neoepitopes.94 Tumor-specific TIL could be utilized to node-positive breast cancer comparing the addition of docetaxel to doxorubicin
with doxorubicin-based chemotherapy: BIG 02-98. J Clin Oncol 2013; 31:860-7.
identify tumor neoantigens that could then be utilized in patient-specific 9. Adams S, Gray RJ, Demaria S, et al. Prognostic value of tumor-infiltrating lym-
vaccine strategies or to create cell-based treatments with autologous TIL phocytes in triple-negative breast cancers from two phase III randomized adjuvant
breast cancer trials: ECOG 2197 and ECOG 1199. J Clin Oncol 2014; 32:2959-
or TIL-derived TCR gene-engineered products. We recently demon- 66.
strated proof of principle in a single case report of complete regression of 10. Denkert C, Loibl S, Noske A, et al. Tumor-associated lymphocytes as an inde-
pendent predictor of response to neoadjuvant chemotherapy in breast cancer.
refractory metastatic breast cancer after adoptive cell transfer with IL-2 J Clin Oncol 2010; 28:105-13.
and pembrolizumab.11 11. Zacharakis N, Chinnasamy H, Black M, et al. Immune recognition of somatic
mutations leading to complete durable regression in metastatic breast cancer. Nat
Med 2018; 24:724-30.
Conclusion 12. Yang J, Yu H, Zhang L, et al. Overexpressed genes associated with hormones in
terminal ductal lobular units identified by global transcriptome analysis: an insight
Immune cells are identified in breast tissue throughout carcino- into the anatomic origin of breast cancer. Oncol Rep 2016; 35:1689-95.
genesis, beginning in normal breast tissue in women at normal risk 13. Ferguson DJ. Intraepithelial lymphocytes and macrophages in the normal breast.
Virchows Arch A Pathol Anat Histopathol 1985; 407:369-78.
for breast cancer and continuing through breast cancer primary 14. Giorno R. Mononuclear cells in malignant and benign human breast tissue. Arch
tumors and breast cancer metastases. The immune cells in normal Pathol Lab Med 1983; 107:415-7.
15. Lwin KY, Zuccarini O, Sloane JP, Beverley PC. An immunohistological study of
breast tissue may play an important role in immunosurveillance and leukocyte localization in benign and malignant breast tissue. Int J Cancer 1985; 36:
clarify our understanding of the prevention (and development) of 433-8.
16. Degnim AC, Hoskin TL, Arshad M, et al. Alterations in the immune cell
early events in breast carcinogenesis. Our understanding of TILs is composition in premalignant breast tissue that precede breast cancer development.
increasing dramatically. These cells have prognostic value for breast Clin Cancer Res 2017; 23:3945-52.
17. Holtmeier W, Kabelitz D. gammadelta T cells link innate and adaptive immune
cancer outcomes and serve in adoptive transfer and other capacities responses. Chem Immunol Allergy 2005; 86:151-83.
to treat disseminated breast cancer. This knowledge is allowing us to 18. Kabelitz D, Wesch D. Features and functions of gamma delta T lymphocytes:
focus on chemokines and their receptors. Crit Rev Immunol 2003; 23:339-70.
recognize and to modify T cells in multiple ways, including selec- 19. Sallusto F, Lenig D, Forster R, Lipp M, Lanzavecchia A. Two subsets of memory T
tion of tumor neoantigen-specific T cells, to the engineering of lymphocytes with distinct homing potentials and effector functions. Nature 1999;
401:708-12.
TCRs to recognize specific tumor associated neoantigens and to 20. Roy D, Liehr JG. Estrogen, DNA damage and mutations. Mutat Res 1999; 424:
insert these TCR into lymphocytes (including peripheral blood 107-15.
21. Danforth DN Jr. Genomic changes in normal breast tissue in women at normal
lymphocytes), for the development of vaccines that recognize risk or at high risk for breast cancer. Breast Cancer (Auckl) 2016; 10:109-46.
tumor-specific antigens in breast cancer metastases. Together, these 22. Cancer Genome Atlas Network. Comprehensive molecular portraits of human
breast tumours. Nature 2012; 490:61-70.
findings allow immunology and immunotherapy to serve as the 23. Karin M, Lawrence T, Nizet V. Innate immunity gone awry: linking microbial
major fourth modality for the management of patients with cancer, infections to chronic inflammation and cancer. Cell 2006; 124:823-35.
24. Grivennikov SI, Greten FR, Karin M. Immunity, inflammation, and cancer. Cell
alongside surgery, chemotherapy, and radiotherapy. This should 2010; 140:883-99.
greatly improve our ability to prevent and treat breast cancer, as well 25. Chow MT, Moller A, Smyth MJ. Inflammation and immune surveillance in
cancer. Semin Cancer Biol 2012; 22:23-32.
as many other malignancies. 26. Troester MA, Hoadley KA, D’Arcy M, et al. DNA defects, epigenetics, and gene
expression in cancer-adjacent breast: a study from The Cancer Genome Atlas. NPJ
Breast Cancer 2016; 2. https://doi.org/10.1038/npjbcancer.2016.7, accessed:
August 29, 2020.
Acknowledgment 27. Frantz C, Stewart KM, Weaver VM. The extracellular matrix at a glance. J Cell Sci
This research was supported by the Intramural Research Pro- 2010; 123:4195-200.
28. Ghajar CM, Bissell MJ. Extracellular matrix control of mammary gland
gram, Center for Cancer Research, National Cancer Institute, Na- morphogenesis and tumorigenesis: insights from imaging. Histochem Cell Biol
tional Institutes of Health, Bethesda, MD. 2008; 130:1105-18.

Clinical Breast Cancer Month 2020 -9


Role of Immune Cells
29. Ohlund D, Elyada E, Tuveson D. Fibroblast heterogeneity in the cancer wound. 61. Denkert C, von Minckwitz G, Darb-Esfahani S, et al. Tumour-infiltrating lym-
J Exp Med 2014; 211:1503-23. phocytes and prognosis in different subtypes of breast cancer: a pooled analysis of
30. Hoy AJ, Balaban S, Saunders DN. Adipocyteetumor cell metabolic crosstalk in 3771 patients treated with neoadjuvant therapy. Lancet Oncol 2018; 19:40-50.
breast cancer. Trends Mol Med 2017; 23:381-92. 62. Mohamed M, Sarwath H, Salih N, et al. CD8þ tumor infiltrating lymphocytes
31. Delort L, Rossary A, Farges MC, Vasson MP, Caldefie-Chezet F. Leptin, adipo- strongly correlate with molecular subtype and clinico-pathological characteristics in
cytes and breast cancer: focus on inflammation and anti-tumor immunity. Life Sci breast cancer patients from Sudan. Transl Med Commun 2016; 1:4.
2015; 140:37-48. 63. Sautes-Fridman C, Petitprez F, Calderaro J, Fridman WH. Tertiary lymphoid
32. Cabia B, Andrade S, Carreira MC, Casanueva FF, Crujeiras AB. A role for novel structures in the era of cancer immunotherapy. Nat Rev Cancer 2019; 19:307-25.
adipose tissueesecreted factors in obesity-related carcinogenesis. Obes Rev 2016; 64. Engelhard VH, Rodriguez AB, Mauldin IS, Woods AN, Peske JD, Slingluff CL Jr.
17:361-76. Immune cell infiltration and tertiary lymphoid structures as determinants of
33. Madeddu C, Gramignano G, Floris C, Murenu G, Sollai G, Maccio A. Role of antitumor immunity. J Immunol 2018; 200:432-42.
inflammation and oxidative stress in post-menopausal oestrogen-dependent breast 65. Liu X, Tsang JYS, Hlaing T, et al. Distinct tertiary lymphoid structure associations
cancer. J Cell Mol Med 2014; 18:2519-29. and their prognostic relevance in HER2 positive and negative breast cancers.
34. Segovia-Mendoza M, Morales-Montor J. Immune tumor microenvironment in Oncologist 2017; 22:1316-24.
breast cancer and the participation of estrogen and its receptors in cancer phys- 66. Lee HJ, Park IA, Song IH, et al. Tertiary lymphoid structures: prognostic signif-
iopathology. Front Immunol 2019; 10:348. https://doi.org/10.3389/fimmu.2019. icance and relationship with tumour-infiltrating lymphocytes in triple-negative
00348, accessed: August 29, 2020. breast cancer. J Clin Pathol 2016; 69:422-30.
35. Kovats S. Estrogen receptors regulate innate immune cells and signaling pathways. 67. Martinet L, Garrido I, Filleron T, et al. Human solid tumors contain high
Cell Immunol 2015; 294:63-9. endothelial venules: association with T- and B-lymphocyte infiltration and favor-
36. Faas M, de Vos P, Melgert B. Sex hormones and immunoregulation. Brain Im- able prognosis in breast cancer. Cancer Res 2011; 71:5678-87.
mune, July 12, 2011, Available at: http://www.brainimmune.com/sex-hormones- 68. Martinet L, Filleron T, Le Guellec S, Rochaix P, Garrido I, Girard JP. High
and-immunoregulation, Accessed: July 9, 2020. endothelial venule blood vessels for tumor-infiltrating lymphocytes are associated
37. Khan D, Ansar Ahmed S. The immune system is a natural target for estrogen with lymphotoxin beta-producing dendritic cells in human breast cancer.
action: opposing effects of estrogen in two prototypical autoimmune diseases. Front J Immunol 2013; 191:2001-8.
Immunol 2015; 6:635. 69. Sofopoulos M, Fortis SP, Vaxevanis CK, et al. The prognostic significance of
38. Bereshchenko O, Bruscoli S, Riccardi C. Glucocorticoids, sex hormones, and peritumoral tertiary lymphoid structures in breast cancer. Cancer Immunol
immunity. Front Immunol 2018; 9:1332. Immunother 2019; 68:1733-45.
39. Bouman A, Heineman MJ, Faas MM. Sex hormones and the immune response in 70. Song IH, Heo SH, Bang WS, et al. Predictive value of tertiary lymphoid structures
humans. Hum Reprod Update 2005; 11:411-23. assessed by high endothelial venule counts in the neoadjuvant setting of triple-
40. Navarro FC, Herrnreiter C, Nowak L, Watkins SK. Estrogen regulation of T-cell negative breast cancer. Cancer Res Treat 2017; 49:399-407.
function and its impact on the tumor microenvironment. Gender Genome 2018; 2: 71. Lin L, Hu X, Zhang H, Hu H. Tertiary lymphoid organs in cancer immunology:
81-91. mechanisms and the new strategy for immunotherapy. Front Immunol 2019; 10:
41. Thompson E, Taube JM, Elwood H, et al. The immune microenvironment of 1398.
breast ductal carcinoma in situ. Mod Pathol 2016; 29:249-58. 72. Girard JP, Springer TA. High endothelial venules (HEVs): specialized endothe-
42. Tower H, Ruppert M, Britt K. The immune microenvironment of breast cancer lium for lymphocyte migration. Immunol Today 1995; 16:449-57.
progression. Cancers (Basel) 2019; 11:1375. 73. Egelston CA, Avalos C, Tu TY, et al. Resident memory CD8þ T cells within
43. Hendry S, Pang JB, Byrne DJ, et al. Relationship of the breast ductal carcinoma in cancer islands mediate survival in breast cancer patients. JCI Insight 2019; 4.
situ immune microenvironment with clinicopathological and genetic features. Clin https://doi.org/10.1172/jci.insight.130000, accessed: August 29, 2020.
Cancer Res 2017; 23:5210-7. 74. Malik BT, Byrne KT, Vella JL, et al. Resident memory T cells in the skin mediate
44. Kim M, Chung YR, Kim HJ, Woo JW, Ahn S, Park SY. Immune microenvi- durable immunity to melanoma. Sci Immunol 2017; 2. https://doi.org/10.1126/
ronment in ductal carcinoma in situ: a comparison with invasive carcinoma of the sciimmunol.aam6346, accessed: August 29, 2020.
breast. Breast Cancer Res 2020; 22:32. 75. Ganesan AP, Clarke J, Wood O, et al. Tissue-resident memory features are linked
45. Chen XY, Yeong J, Thike AA, Bay BH, Tan PH. Prognostic role of immune to the magnitude of cytotoxic T cell responses in human lung cancer. Nat Immunol
infiltrates in breast ductal carcinoma in situ. Breast Cancer Res Treat 2019; 177:17- 2017; 18:940-50.
27. 76. Savas P, Virassamy B, Ye C, et al. Single-cell profiling of breast cancer T cells
46. Gil Del Alcazar CR, Huh SJ, Ekram MB, et al. Immune escape in breast cancer reveals a tissue-resident memory subset associated with improved prognosis. Nat
during in situ to invasive carcinoma transition. Cancer Discov 2017; 7:1098-115. Med 2018; 24:986-93.
47. Pruneri G, Lazzeroni M, Bagnardi V, et al. The prevalence and clinical relevance of 77. García-Martínez E, Gil GL, Benito AC, et al. Tumor-infiltrating immune cell
tumor-infiltrating lymphocytes (TILs) in ductal carcinoma in situ of the breast. profiles and their change after neoadjuvant chemotherapy predict response and
Ann Oncol 2017; 28:321-8. prognosis of breast cancer. Breast Cancer Res 2014; 16:488.
48. Campbell MJ, Baehner F, O’Meara T, et al. Characterizing the immune micro- 78. Schmidt M, Weyer-Elberich V, Hengstler JG, et al. Prognostic impact of CD4-
environment in high-risk ductal carcinoma in situ of the breast. Breast Cancer Res positive T cell subsets in early breast cancer: a study based on the FinHer trial
Treat 2017; 161:17-28. patient population. Breast Cancer Res 2018; 20:15.
49. DeNardo DG, Coussens LM. Inflammation and breast cancer. Balancing immune 79. Park IA, Hwang SH, Song IH, et al. Expression of the MHC class II in triple-
response: crosstalk between adaptive and innate immune cells during breast cancer negative breast cancer is associated with tumor-infiltrating lymphocytes and
progression. Breast Cancer Res 2007; 9:212. interferon signaling. PLoS One 2017; 12. https://doi.org/10.1007/s10549-016-
50. Coronella-Wood JA, Hersh EM. Naturally occurring B-cell responses to breast 3786-z, accessed: August 29, 2020.
cancer. Cancer Immunol Immunother 2003; 52:715-38. 80. Pelekanou V, Barlow WE, Nahleh ZA, et al. Tumor-infiltrating lymphocytes
51. Martin F, Ladoire S, Mignot G, Apetoh L, Ghiringhelli F. Human FOXP3 and and PD-L1 expression in pre- and posttreatment breast cancers in the SWOG
cancer. Oncogene 2010; 29:4121-9. S0800 phase II neoadjuvant chemotherapy trial. Mol Cancer Ther 2018; 17:
52. Bates GJ, Fox SB, Han C, et al. Quantification of regulatory T cells enables the 1324-31.
identification of high-risk breast cancer patients and those at risk of late relapse. 81. Zerdes I, Sifakis EG, Matikas A, et al. Programmed death-ligand 1 gene expression
J Clin Oncol 2006; 24:5373-80. is a prognostic marker in early breast cancer and provides additional prognostic
53. Miligy I, Mohan P, Gaber A, et al. Prognostic significance of tumour infiltrating B value to 21-gene and 70-gene signatures in estrogen receptorepositive disease. Mol
lymphocytes in breast ductal carcinoma in situ. Histopathology 2017; 71:258-68. Oncol 2020; 14:951-63.
54. Stanton SE, Adams S, Disis ML. Variation in the incidence and magnitude of 82. Nanda R, Chow LQ, Dees EC, et al. Pembrolizumab in patients with advanced
tumor-infiltrating lymphocytes in breast cancer subtypes: a systematic review. triple-negative breast cancer: phase Ib KEYNOTE-012 study. J Clin Oncol 2016;
JAMA Oncol 2016; 2:1354-60. 34:2460-7.
55. Ben-Hur H, Cohen O, Schneider D, et al. The role of lymphocytes and macro- 83. Rugo HS, Delord JP, Im SA, et al. Safety and antitumor activity of pembrolizumab
phages in human breast tumorigenesis: an immunohistochemical and morpho- in patients with estrogen receptor-positive/human epidermal growth factor receptor
metric study. Anticancer Res 2002; 22:1231-8. 2enegative advanced breast cancer. Clin Cancer Res 2018; 24:2804-11.
56. Beausang JF, Wheeler AJ, Chan NH, et al. T cell receptor sequencing of early-stage 84. Schmid P, Adams S, Rugo HS, et al. Atezolizumab and nab-paclitaxel in advanced
breast cancer tumors identifies altered clonal structure of the T cell repertoire. Proc triple-negative breast cancer. N Engl J Med 2018; 379:2108-21.
Natl Acad Sci U S A 2017; 114:E10409-17. 85. Schmid P, Cortes J, Pusztai L, et al. Pembrolizumab for early triple-negative breast
57. Park JH, Jang M, Tarhan YE, et al. Clonal expansion of antitumor T cells in breast cancer. N Engl J Med 2020; 382:810-21.
cancer correlates with response to neoadjuvant chemotherapy. Int J Oncol 2016; 86. Adams S, Gatti-Mays ME, Kalinsky K, et al. Current landscape of immunotherapy
49:471-8. in breast cancer: a review. JAMA Oncol 2019.
58. Sica A, Allavena P, Mantovani A. Cancer related inflammation: the macrophage 87. Benedetti R, Dell’Aversana C, Giorgio C, Astorri R, Altucci L. Breast cancer
connection. Cancer Lett 2008; 267:204-15. vaccines: new insights. Front Endocrinol (Lausanne) 2017; 8:270.
59. Solinas C, Carbognin L, De Silva P, Criscitiello C, Lambertini M. Tumor-infil- 88. Hickey RM, Kulik LM, Nimeiri H, et al. Immuno-oncology and its opportunities for
trating lymphocytes in breast cancer according to tumor subtype: current state of interventional radiologists: immune checkpoint inhibition and potential synergies
the art. Breast 2017; 35:142-50. with interventional oncology procedures. J Vasc Interv Radiol 2017; 28:1487-94.
60. Liu F, Lang R, Zhao J, et al. CD8þ cytotoxic T cell and FOXP3þ regulatory T cell 89. Hwang WL, Pike LRG, Royce TJ, Mahal BA, Loeffler JS. Safety of combining
infiltration in relation to breast cancer survival and molecular subtypes. Breast radiotherapy with immune-checkpoint inhibition. Nat Rev Clin Oncol 2018; 15:
Cancer Res Treat 2011; 130:645-55. 477-94.

10 - Clinical Breast Cancer Month 2020


Stephanie L. Goff, David N. Danforth
90. Gatti-Mays ME, Balko JM, Gameiro SR, et al. If we build it they will come: infiltrating lymphocytes for patients with metastatic melanoma. J Clin Oncol
targeting the immune response to breast cancer. NPJ Breast Cancer 2019; 5:37. 2016; 34:2389-97.
91. Goff SL, Zacharakis N, Assadipour Y, et al. Recognition of autologous 93. Assadipour Y, Zacharakis N, Crystal JS, et al. Characterization of an immunogenic
neoantigens by tumor infiltrating lymphocytes derived from breast cancer mutation in a patient with metastatic triple-negative breast cancer. Clin Cancer Res
metastases, In: Proceedings of the San Antonio Breast Cancer Symposium; 2017; 23:4347-53.
December 6-10, 2016; San Antonio, TX. AACR; 2017, Abstract P2-04-02. 94. Harao M, Forget MA, Roszik J, et al. 4-1BBeenhanced expansion of CD8þ TIL
92. Goff SL, Dudley ME, Citrin DE, et al. Randomized, prospective evaluation from triple-negative breast cancer unveils mutation-specific CD8þ T cells. Cancer
comparing intensity of lymphodepletion before adoptive transfer of tumor- Immunol Res 2017; 5:439-45.

Clinical Breast Cancer Month 2020 - 11


Peran Sel pada Sistem Imun dalam Jaringan Payudara dan Imunoterapi
sebagai Pilihan Terapi pada Kanker Payudara

Pendahuluan
Kanker payudara merupakan salah satu jenis keganasan yang paling umum ditemukan
pada wanita, dan mencapai angka 316.000 kasus setiap tahunnya di Amerika Serikat. Sebagian
besar kanker payudara berlokasi di saluran ASI, baik sebagai karsinoma duktal-in-situ (DCIS)
atau sebagai karsinoma duktal invasif. Perkembangan dan progresivitas kanker payudara dapat
dipengaruhi oleh berbagai elemen dalam lingkungan mikro duktus, baik dalam tingkat seluler
seperti sel-sel yang berperan dalam sistem imun, adiposit, fibroblas, dan mikrobioma, serta pada
elemen terlarut seperti faktor pertumbuhan, sitokin, kemokin, dan prostaglandin. Salah satu
komponen yang memiliki peran penting dari unsur-unsur ini adalah sel-sel yang berperan dalam
sistem imun. Lapisan sel duktal pada payudara normal mengandung populasi sel imun yang
signifikan yang terdiri dari sel CD8+ dan CD4+T, sel B, sel dendritik, makrofag, sel NK dan
subtipe sel-sel yang berperan dalam sistem imun lainnya. Sel-sel yang berperan dalam sistem
imun ini menyusun respon imunitas bawaan dan adaptif terhadap lapisan epitel untuk
perlindungan terhadap agen eksogen dan endogen, serta eliminasi sel transformatif.
Infiltrasi sel-sel yang berperan dalam sistem imun pada kanker payudara terdiri dari
sejumlah subtipe seluler, antara lain sel CD3+ (CD4+ dan CD8+), sel B, monosit/makrofag, sel
dendritik, dan sel NK. Sejumlah subtipe sel-sel yang berperan dalam sistem imun pada parenkim
dan stroma menempatkan sel-sel ini di sekitar lingkungan mikro sel tumor dan sel lainnya,
sehingga memungkinkan sel-sel ini untuk mempengaruhi pertumbuhan tumor dengan berbagai
cara, baik secara langsung melalui sitotoksisitas CD4+ dan CD8+ yang dimediasi sel, atau secara
tidak langsung melalui tindakan imunosupresif atau imunostimulan dari sitokin yang
disekresikan, faktor pertumbuhan dan agen lainnya.
Salah satu tujuan penyusunan artikel tinjauan ini adalah untuk mengevaluasi secara rinci
karakteristik khusus sel-sel yang berperan dalam sistem imun duktal pada jaringan payudara
normal, TIL pada kanker payudara, dan upaya untuk mengaplikasikan temuan penelitian tersebut
sebagai indikator prognostik yang baik serta tatalaksana imunoterapi yang dapat
dipertimbangkan sebagai salah satu pilihan terapi untuk kanker payudara.
Bahan dan Metode
Pencarian literatur dilakukan melalui PubMed dan referensi silang untuk
mengidentifikasi publikasi yang menjelaskan sifat dan distribusi sel-sel yang berperan dalam
sistem imun pada komponen jaringan payudara normal dan kanker payudara. Jaringan payudara
normal didefinisikan sebagai komponen jaringan payudara pada subjek wanita dengan risiko
normal (contohnya pada mammoplasty reduksi), sedangkan komponen jaringan dari payudara
kontralateral atau dari jaringan yang berdekatan dengan kanker payudara pada wanita dengan
kanker payudara. Penekanan pada sel-sel yang berperan dalam sistem imun pada tumor primer
diutamakan pada penelitian yang mengevaluasi kanker payudara pada subjek.

Hasil dan Pembahasan


Sel-Sel Yang Berperan dalam Sistem Imun pada Payudara Normal
Anatomi Duktus Payudara. Unit utama dalam sistem saluran payudara adalah asini, yang
mengalir ke intralobular dan kemudian menuju duktus ekstralobular; dan bersama-sama, struktur
ini membentuk unit lobular duktal terminal (TDLU). TDLU dianggap sebagai lokasi predominan
asal kanker payudara. Sebagian besar kanker payudara merupakan karsinoma yang muncul di
epitel dan memiliki komponen in-situ serta karsinoma duktal invasif dan lobular invasif. Saluran
payudara dikelilingi oleh stroma yang terdiri dari matriks ekstraseluler (ECM), fibroblas,
adiposit, sel-sel yang berperan dalam sistem imun, mikrobioma, dan pembuluh darah. Saluran
payudara dan stroma bersama-sama membentuk lingkungan mikro payudara.

Gambar 1. Sifat dan Distribusi Sel-Sel yang Berperan dalam


Sistem Imun pada Komponen Jaringan Payudara Normal. Sel-sel
yang berperan dalam sistem imun dapat ditemukan pada jaringan
payudara normal, termasuk limfosit dan sel turunan myeloid. Sel-
sel yang berperan dalam sistem imun tubuh sebagian besar
ditemukan pada epitel duktal (intraepitel) namun dapat pula
ditemukan pada stroma (tidak digambarkan). Matriks ekstraseluler
stromal (ECM) juga mengandung berbagai jenis dan struktur sel
yang dapat berinteraksi dengan sel-sel yang berperan dalam sistem
imun serta epitel. Diadaptasi dari Degnim dkk.
Distribusi Sel-Sel yang Berperan dalam Sistem Imun pada Komponen Jaringan Payudara
Normal. Jaringan payudara normal mengandung sel-sel yang berperan dalam sistem imun baik
pada jalur derivatif myeloid (monosit, makrofag, sel dendritik) maupun limfoid (limfosit T dan
B. limfosit). Sel-sel yang berperan dalam sistem imun yang diidentifikasi dalam sejumlah
penelitian yakni didominasi oleh limfosit, termasuk limfosit T CD8+ dan CD4+, serta makrofag.
Secara keseluruhan, sel CD45+ memiliki karakteristik paling menonjol di antara sel-sel epitel
duktus lainnya. Sel T CD8+ merupakan sel yang paling umum ditemukan pada semua lokasi,
dan dalam sebuah penelitian oleh Degnim dkk, sel T CD8+ dan CD11c+ (dendritc) sel
ditemukan pada hampir semua lobulus dan memiliki jumlah terbanyak di seluruh lobulus.
Demikian juga halnya pada sel CD68+ (makrofag/monosit) yang banyak ditemukan pada seluruh
lobulus; sementara sel CD4+ dan CD20+ cenderung lebih jarang ditemukan. Ruffell dkk
melakukan penelitian terhadap jaringan payudara normal yang lokasinya berdekatan dengan
kanker payudara atau dari spesimen mastektomi profilaksis dan menemukan bahwa sel CD3+
(CD4+ dan CD8+) merupakan jenis sel yang paling umum ditemukan, dan sel-sel jalur derivatif
myeloid seperti makrofag, sel dendritik dan neutrofil juga cenderung mudah ditemukan.

Imunogenisitas Sel-Sel yang Berperan dalam Sistem Imun pada Jaringan Payudara Normal. Sel-
sel yang berperan dalam sistem imun pada saluran payudara berada di dekat kelenjar getah
bening yang mengalir di area ipsilateral aksila dan mediastinum. Dengan demikian semua
komponen seluler dan limfatik utama yang diperlukan untuk respons imun seluler adaptif
ditemukan dalam sistem duktus. Zumwalde dkk pada salah satu penelitiannya mengemukakan
analisis lebih lanjut dari sel T CD8+, yang menunjukkan bahwa sel-sel tersebut merupakan sel
CD45RO+/CD27, dan dengan demikian merupakan sel T memori efektor (TEM). Populasi
limfosit intraepitel TEM pada payudara normal menandakan bahwa sel-sel ini telah diaktivasi
antigen, sekaligus menunjukkan adanya jaringan imun dinamis antara epitel duktus dan kelenjar
getah bening regional. Sejumlah sumber potensial antigen dapat berperan dalam aktivasi sel T
CD8+ ini, baik secara eksogen (misalnya, virus, bakteri) dan endogen (protein nukleus dan
sitosol, DNA, protein ekstraseluler). Sebuah penelitian lanjutan yang dilakukan oleh Degnim dkk
membandingkan distribusi sel-sel yang berperan dalam sistem imun pada lobulus individu yang
terdiagnosis penyakit payudara jinak dengan sampel jaringan payudara dari wanita tanpa
kelainan payudara dan menemukan lobulus pada penyakit payudara jinak menunjukkan
kepadatan sel T CD8+, sel CD11c+dendritik, Sel CD20B, dan makrofag CD68 yang cenderung
lebih tinggi bila dibandingkan dengan individu kontrol normal. Infiltrasi terkait dari efektor imun
bawaan dan adaptif pada penyakit payudara jinak menunjukkan adanya lingkungan mikro
imunogenik.

Sel-Sel yang Berperan dalam Sistem Imun Intraepitelial dan Lingkungan Mikro. Sel-sel yang
berperan dalam sistem imun pada lapisan intraepitel juga dikelilingi dan dapat dipengaruhi oleh,
komponen utama dari lingkungan mikro seperti ECM dan matriks interstisial. ECM terdiri dari
membran basalis duktal dan endotel, dan matriks interstisial terdiri dari jaringan ikat dan
komponen seluler (fibroblast, adiposit, endothelial, inflammatory). Komponen lingkungan mikro
juga dianggap memiliki peran penting selama proses karsinogenesis payudara. Perubahan
sederhana dalam komposisi ECM dapat mengubah difusi dan permeabilitas melalui ECM, dan
pembatasan difusi dapat mendorong tumorigenesis. Fibroblas, komponen utama ECM, dapat
diprogram ulang menjadi fibroblas terkait kanker oleh sitokin dan faktor pertumbuhan yang
disekresikan oleh sel epitel yang mengalami transformasi. Fibroblas terkait kanker dapat
diaktifkan untuk mendorong inisiasi tumor, pemodelan ulang ECM, dan modulasi sel-sel yang
berperan dalam sistem imun. Selain fibroblas, adiposit juga merupakan salah satu komponen
penting dari ECM dan berada di dekat sel epitel, tumor, dan sel-sel yang berperan dalam sistem
imun. Sel kanker payudara menginduksi produksi enzim endokrin dan parakrin serta lipid
bioaktif oleh adiposit, yang pada gilirannya mendorong peningkatan pertumbuhan dan invasi sel
tumor. Leptin juga merupakan produk sekretori penting dari adiposit, yang dapat merangsang
produksi sejumlah faktor inflamasi termasuk interleukin (IL)-1, IL-6, dan faktor nekrosis tumor
(TNF)α. Obesitas merupakan faktor risiko yang cukup signifikan pengaruhnya terhadap insidensi
kanker payudara pada wanita pascamenopause dan dapat mendorong proses prokarsinogenik
pada adiposit. Sejumlah faktor seperti leptin, TNFα, IL-6, dan resistin yang disekresikan oleh
adiposit normal, akan meningkat secara paralel pada penderita obesitas dan memiliki efek
prokarsinogenik. Proses ini dapat memodifikasi sistem kekebalan tubuh dan meningkatkan
pertumbuhan tumor.

Efek Estrogenik pada Sel-Sel yang Berperan dalam Sistem Imun Jaringan Payudara Normal
Estrogen merupakan salah satu hormon predominan pada wanita selama periode premenopause
dan postmenopause. Estrogen memiliki peran penting dalam proses karsinogenesis payudara. Hal
ini terjadi melalui aktivitas estrogenik pada sel epitel payudara, namun terdapat evidensi bahwa
sel-sel yang berperan dalam sistem imun mengandung reseptor estrogen (ERs) dan pada saat
bersamaan aktivitasnya diatur oleh estrogen. Aktivitas hormonal ini akan memberikan
kesempatan bagi estrogen untuk mempengaruhi immunosurveillance, serta perkembangan dan
progresivitas kanker payudara. Aktivitas estrogenik dihasilkan dari pengikatan reseptor
sitoplasma ERα dan Erβ serta aktivasi jalur genomik, walaupun estrogen juga dapat
mengaktifkan aktivitas reseptor faktor pertumbuhan dengan pensinyalan melalui jalur
nongenomik oleh sinyal ERα independen ligan yang melibatkan pengikatan E2 ke reseptor
terikat membran, G protein-coupled estrogen receptor 1 (GPER1) dengan aktivasi protein G.
Sel-sel yang berperan dalam sistem imun dari kedua jenis sel bawaan (sel dendritik, makrofag,
neutrofil) dan adaptif (CD4+, CD8+, sel B) dalam jaringan payudara normal maupun jaringan
kanker payudara terbukti memiliki ER dan responsif terhadap estrogen.

Sel-Sel yang Berperan dalam Sistem Imun pada Karsinoma Duktal In Situ (DCIS). DCIS
memiliki karakteristik berupa peningkatan kepadatan dan luasnya infiltrasi sel-sel yang berperan
dalam sistem imun dibandingkan dengan jaringan payudara normal. DCIS derajat tinggi
memiliki persentase sel FOXP3+, makrofag CD68 dan CD68+PCNA+, antigen leukosit manusia
(HLA)–DR cells, sel T CD4+, sel CD20+B, dan total TIL yang secara signifikan lebih tinggi bila
dibandingkan dengan DCIS derajat rendah atau moderat. Thompson dkk mengemukakan bahwa
sel T CD3+ cenderung mendominasi di semua subtipe DCIS pada semua usia, dengan jumlah sel
T CD4+ yang lebih sedikit bila dibandingkan rata-rata jumlah sel T CD8+. Sel B CD20+
merupakan TIL yang paling umum ditemukan setelah sel T CD3+, diikuti oleh sel T regulator
FOXP3+ (Treg). Sel Treg FOXP3+ merupakan sel-sel yang berperan dalam sistem
imunosupresif penting lainnya yang cenderung meningkat pada DCIS.

Sel-Sel yang Berperan dalam Sistem Imun pada Karsinoma Payudara Invasif. Ruffell dkk
mengamati bahwa jaringan kanker payudara mengandung infiltrat yang didominasi oleh limfosit
CD8+ dan CD4+, dengan sejumlah kecil sel NK dan limfosit B, sedangkan pada jaringan
payudara normal banyak ditemukan sel derivatif myeloid seperti makrofag, sel mast, dan
neutrofil. Limfosit T teraktivasi juga mendominasi jaringan tumor, dengan sel T CD4+ dan
CD8+ menunjukkan peningkatan ekspresi penanda aktivasi CD69 dan HLA-DR. Kompleksitas
komposisi sel-sel yang berperan dalam sistem imun pada kanker payudara mencerminkan
aktivitas silang antara komponen respon imun bawaan karena mengatur lingkungan mikro tumor
dan polaritas respon imun adaptif di dalam tumor tersebut. Walaupun ditemukan peningkatan
progresif pada semua jenis sel baik pada sel parenkim dan stroma yang berbatasan dari jaringan
payudara normal ke karsinoma duktal, perbedaan paling mencolok dari tumor jinak proliferatif,
DCIS dan kanker invasif adalah masuknya sel CD3+ pada stroma. Peningkatan subset sitotoksik
(granzyme B) terbatas pada sel-sel yang berperan dalam sistem imun di parenkim.

Gambar 2. Sel-Sel Yang Berperan Dalam Sistem Imun Pada Penyakit Proliferatif Payudara Dan Kanker Payudara.
Sel-sel yang berperan dalam sistem imun tubuh, termasuk CD68+ (makrofag/monosit), CD3+ (CD4+, CD8+), dan
limfosit sitotoksik (CTL) ditemukan dalam penyakit proliferatif payudara dan mengalami peningkatan pada
parenkim serta stroma pada perkembangan kanker payudara. Peningkatan relatif untuk setiap jenis sel, dengan
proliferasi jinak sebagai baseline, ditunjukkan pada skala logaritmik pada grafik di bawah gambar.

Karakteristik Sel-Sel yang Berperan dalam Sistem Imun pada Struktur Limfe Tersier Jaringan
Kanker Payudara. Struktur limfe tersier (tertiary lymph organ/TLS) merupakan organ limfoid
ektopik yang berkembang pada lokasi peradangan kronis termasuk penyakit autoimun, infeksi,
dan tumor. TLS memiliki struktur yang terdiri dari sejumlah tipe sel imun bawaan (dendritik,
makrofag, neutrofil) dan adaptif (sel B, sel T) serta sel plasma dan venula endothelial (HEV).
TLS merupakan lokasi aktivasi respon imun terhadap tumor yang mengumpulkan dan
mengaktifkan TIL. TLS dapat berlokasi di dalam tumor atau dalam lokasi peritumor. Komponen
penting dari TLS adalah HEV, venula postkapiler khusus yang ditemukan pada jaringan limfoid
dan mendukung ekstravasasi limfosit tingkat tinggi dari darah. Identifikasi kumpulan sel-sel
yang berperan dalam sistem imun yang terorganisir dengan sel memori efektor di dalam dan di
sekitar jaringan kanker payudara serta kemampuan aktivitas antitumor dapat mempengaruhi
perkembangan dan perluasannya pada tumor. Keberadaan TLS dari HEV yang dapat
meningkatkan pergerakan limfosit ke dalam tumor dan juga berkontribusi terhadap upaya
identifikasi agen yang dapat mendorong aktivitas pembuluh darah tersebut.

Sel-Sel yang Berperan dalam Sistem Imun sebagai Biomarker Prognostik. Salah satu penelitian
pertama mengenai topik ini dilakukan oleh Denkert dkk dengan menggunakan biopsi inti tumor
dan stroma sebelum perawatan, untuk dianalisis infiltrat limfositiknya yang dievaluasi dengan
pewarnaan hematoksilin dan eosin rutin. Dua kategori infiltrat imun didefinisikan: iTu-Ly
(limfosit intratumoral) dalam kontak langsung dengan sel tumor atau di dalam kumpulan sel
tumor dan str-Ly (limfosit stroma) tanpa kontak langsung dengan sel tumor. Integrin CD103,
sebuah penanda sel T memori residen jaringan (TRM) memiliki aktivitas mediasi lokasi daerah
kanker di dalam tumor. Sejumlah upaya dilakukan untuk mengklasifikasi TIL dalam spesimen
kanker payudara, dengan fokus pada limfosit stroma sebagai indikator prognostik yang paling
dapat direproduksi dan memiliki kemungkinan respons yang cukup signifikan. Analisis oleh
Denkert dkk dari data yang dikumpulkan dari German Breast Cancer Group mengemukakan
bahwa klasifikasi tumor berdasarkan tingkat invasi limfosit stroma memungkinkan prediksi
respon kemoterapi neoadjuvan pada seluruh subtipe molekuler. Ruffell dkk mengemukakan
bahwa kemoterapi neoadjuvan mengubah kompleksitas lingkungan mikro imun dari tumor-
tumor sekunder. Tumor residual yang diintervensi dengan kemoterapi neoadjuvan menunjukkan
adanya peningkatan persentase dari sel myeloid yang menginfiltrasi, disertai dengan peningkatan
rasio sel CD8/CD4T dan jumlah sel ekspresi granzim yang cenderung lebih tinggi, bila
dibandingkan dengan tumor yang tidak diobati dengan kemoterapi neoadjuvan.

Terapi Berbasis Kekebalan Tubuh untuk Kanker Payudara. Mayoritas penelitian limfosit pada
kanker payudara difokuskan pada karakterisasi sifat infiltrasi limfosit, dengan sejumlah data
ekspresi gen yang mengisyaratkan peran fungsional sel-sel ini dalam respons antikanker.
Kemokin yang dianggap bertanggung jawab terhadap migrasi limfosit dan tanda ekspresi gen
yang terkait dengan respons efektor Tipe I telah terbukti berkorelasi dengan pCR. Reseptor
kematian terprogram 1 (PD-1) dan salah satu li-gandnya (PD-L1) juga menunjukkan korelasi
terhadap tingkat pCR yang lebih tinggi dan prognosis yang lebih baik pada kanker payudara.
Infiltrat imun yang ditemukan pada tahap awal DCIS mendorong pentingnya strategi untuk
meningkatkan respon imun terhadap tumor antigen potensial yakni dengan vaksinasi, yang telah
dieksplorasi baik dalam segi pencegahan, adjuvan, neoadjuvan dan kondisi metastatis. Upaya
tambahan yang dapat dilakukan antara lain mengeksplorasi penggunaan strategi ablatif tumor
(krioablasi, ablasi frekuensi radio, radiasi stereotaktik) sebagai pendorong potensi pelepasan
antigen terkait tumor dengan inhibitor checkpoint antibodi monoklonal untuk meningkatkan
derajat respon. TIL spesifik tumor dapat digunakan untuk mengidentifikasi neoantigen tumor
yang kemudian dapat digunakan dalam strategi vaksin khusus pasien atau untuk membuat
perawatan berbasis sel dengan TIL atau turunan produk rekayasa gen TCR autolog TIL. Peneliti
baru-baru ini menemukan bukti dalam laporan kasus tunggal bahwa terjadi regresi dari kanker
payudara yg bermetastasis pasca transfer sel adoptif dengan IL-2 dan pembrolizumab.

Kesimpulan
Sel-sel yang berperan dalam sistem imun jaringan payudara normal memiliki peran
penting dalam immunosurveillance dan menambah wawasan mengenai pencegahan dan
perkembangan proses karsinogenesis payudara. TIL memiliki nilai prognostik terhadap kanker
payudara dan berfungsi dalam transfer adopsi sebagai tatalaksana persebaran kanker payudara.
Imunologi dan imunoterapi berfungsi sebagai salah satu modalitas utama untuk pengelolaan
pasien dengan kanker, di samping pembedahan, kemoterapi, dan radioterapi.

You might also like