Download as pdf or txt
Download as pdf or txt
You are on page 1of 22

Downloaded from http://perspectivesinmedicine.cshlp.

org/ on June 26, 2023 - Published by Cold Spring Harbor Laboratory Press

Immunotherapy Approaches for Breast Cancer


Patients in 2023

Leisha A. Emens1,2 and Sherene Loi3,4


1
Department of Medicine, University of Pittsburgh/UPMC Hillman Cancer Center, Pittsburgh,
Pennsylvania 15232, USA
2
Ankyra Therapeutics, Boston, Massachusetts 02116, USA
3
The Sir Peter MacCallum Department of Medical Oncology, University of Melbourne, Parkville,
Victoria 3010, Australia
4
Division of Cancer Research, Peter MacCallum Cancer Centre, Melbourne, Victoria 3000, Australia
Correspondence: lemens@ankyratx.com, sherene.loi@petermac.org

Immunotherapy, particularly agents targeting the immunoregulatory PD-1/PD-L1 axis, har-


nesses the power of the immune system to treat cancer, with unique potential for a durable
treatment effect due to immunologic memory. The PD-1 inhibitor pembrolizumab combined
with neoadjuvant chemotherapy followed by adjuvant pembrolizumab improves event-free
survival and is a new standard of care for high-risk, early-stage triple-negative breast cancer
(TNBC), regardless of tumor PD-L1 expression. For metastatic TNBC, pembrolizumab com-
www.perspectivesinmedicine.org

bined with chemotherapy is a new standard of care for the first-line therapy of PD-L1+
metastatic TNBC, with improvement in overall survival. The PD-L1 inhibitor atezolizumab
combined with nab-paclitaxel is also approved outside the United States for the first-line
treatment of metastatic PD-L1+ TNBC. Current research focuses on refining the use of
immunotherapy in TNBC by defining informative predictive biomarkers, developing
immunotherapy in early and advanced HER2-driven and luminal breast cancers, and over-
coming primary and secondary resistance to immunotherapy through unique immune-based
strategies.

B reast cancer is often immunologically cold


with low tumor mutational burden (TMB).
However, tumor-infiltrating lymphocytes (TILs)
trates and others do not? and (2) How can we
most effectively harness antitumor immunity
for breast cancer treatment? Recent work dem-
are present in some breast cancers. TILs are en- onstrates that immune checkpoint inhibitors
riched in more aggressive, highly proliferative (ICIs) targeting the programmed cell death 1
breast cancers, particularly triple-negative receptor (PD-1) or its ligand programmed death
breast cancers (TNBCs) and HER2+ breast can- ligand 1 (PD-L1) is effective in the treatment of
cers (Stanton et al. 2016). Key questions are (1) both early- and late-stage TNBC, resulting in
Why do some patients develop immune infil- multiple health authority approvals around the

Editors: Jane E. Visvader, Jeffrey M. Rosen, and Samuel Aparicio


Additional Perspectives on Breast Cancer: From Fundamental Biology to Therapeutic Strategies available at
www.perspectivesinmedicine.org
Copyright © 2023 Cold Spring Harbor Laboratory Press; all rights reserved; doi: 10.1101/cshperspect.a041332
Cite this article as Cold Spring Harb Perspect Med 2023;13:a041332

1
Downloaded from http://perspectivesinmedicine.cshlp.org/ on June 26, 2023 - Published by Cold Spring Harbor Laboratory Press

L.A. Emens and S. Loi

world. Intense investigation into the potential pansion of T cells occurs when the primary le-
efficacy of immunotherapy for other breast can- sion remains as a source of tumor antigens (Liu
cer subtypes is ongoing. et al. 2016). For TNBC, a pathological complete
response ( pCR), the absence of invasive cancer
at surgery after neoadjuvant treatment, strongly
IMMUNE INFILTRATE IN BREAST CANCER correlates with excellent clinical outcomes and is
Although normal breast tissue does not typically a common clinical end point in breast cancer
contain high levels of immune cells, breast can- clinical trials (Cortazar et al. 2014). Several trials
cers sometimes harbor TILs. Immune infiltrates have tested the combination of ICI and neoad-
are composed primarily of T effector and effec- juvant chemotherapy in early TNBC, demon-
tor memory cells (Savas et al. 2018). As the strating significantly improved pCR and/or
quantity of intratumoral immune infiltrates in- event-free survival (EFS) rates (Table 1).
creases, the ratio of CD8+ to CD4+ T cells in- KEYNOTE-522 is a randomized phase III
creases, reflecting an evolving cytotoxic T-cell trial that tests the addition of pembrolizumab
response. Tissue-resident memory T cells have or placebo to neoadjuvant chemotherapy with
been identified in high-TIL TNBCs, signifying carboplatin and paclitaxel sequenced with
the importance of the local T-cell memory re- doxorubicin and cyclophosphamide (AC), fol-
sponse. High levels of TILs present in breast lowed by surgery and adjuvant pembrolizumab
cancer at diagnosis is strongly prognostic, par- (Schmid et al. 2020b, 2022). Adding pembroli-
ticularly in TNBC. Although not yet incorporat- zumab to neoadjuvant chemotherapy signifi-
ed into standard staging systems, the levels of cantly increased the pCR rate, and more impor-
TILs can effectively up- or down-stage anatomic tantly reduced the risk of recurrence for all
patients (hazard ratio [HR] 0.63, P < 0.001), re-
www.perspectivesinmedicine.org

staging (Loi et al. 2022a). Furthermore, intratu-


moral T cells most often express the immune gardless of PD-L1 expression. Based on these
checkpoints PD-1 and CTLA-4, but not TIM- data, the U.S. Food and Drug Administration
3 or LAG-3, with tissue-resident memory T cells (FDA) has approved neoadjuvant pembrolizu-
expressing PD-1, CTLA-4, TIM-3, LAG-3, and mab with chemotherapy for patients with early
TIGIT in addition to cytotoxic T-cell markers TNBC. Multiple important questions remain:
(Savas et al. 2018). These observations underlie (1) What is the optimum chemotherapy back-
the rationale that immune checkpoint blockade bone? (2) Can we de-escalate therapy for appro-
can improve outcomes in patients with breast priate patients? (3) What is the proper role of
cancer. Notably, metastatic breast tumors have adjuvant ICI therapy? and (4) What is the best
fewer TILs than primary tumors, suggesting in- way to select patients for neoadjuvant immuno-
creasing immune suppression with disease pro- therapy? Other clinical trials illustrating these
gression (Cimino-Mathews et al. 2016; Szekely issues are discussed below.
et al. 2018).
Benefit of Neoadjuvant Carboplatin
EARLY-STAGE BREAST CANCER Three randomized trials (IMpassion031, I-
SPY2, and GeparNuevo, testing atezolizumab,
Immunotherapy in Early TNBC
pembrolizumab, and durvalumab, respectively)
Several trials evaluating ICIs targeting PD-1/ have yielded positive results without adding car-
PD-L1 in early breast cancer have been reported. boplatin to sequential AC and taxane (Mitten-
Given the high levels of TILs in triple-negative dorf et al. 2020; Nanda et al. 2020; Loibl et al.
tumors, immunotherapy trials for early breast 2022). These trials also allowed or mandated
cancer have focused on TNBC. Importantly, dose-dense (2-wk) AC, rather than the 3-wk
preclinical data demonstrate improved re- AC used in KEYNOTE-522. GeparNuevo dem-
sponses to neoadjuvant relative to adjuvant onstrated improved invasive and distant dis-
ICI, suggesting that improved priming and ex- ease-free survival (iDFS and DDFS) and overall

2 Cite this article as Cold Spring Harb Perspect Med 2023;13:a041332


www.perspectivesinmedicine.org

Table 1. Selected clinical trials in early triple-negative breast cancer (TNBC)


Clinical trial Chemotherapy
(sample size) Design Disease stage ICI backbone Adjuvant therapy Results
KEYNOTE-522 Randomized cT1N1-2 Pembro Carbo + paclitaxel, Pembro to complete pCR in ITT 64.8% vs. 51.2%,
(n = 1174) phase III cT2-4N0-2 then AC 1 yr total (no cape P < 0.001
allowed) 3-yr EFS 84.5% vs. 76.8%,
HR 0.63, P < 0.001
IMpassion031 Randomized cT2-4N0-3 Atezo Nab-paclitaxel, then Atezo to complete pCR in ITT 58% vs. 41%,
(n = 333) phase III AC 1 yr total (cape P = 0.0044
allowed) pCR in PD-L1+ 69% vs. 49%,
P = 0.021
EFS immature
NeoTripaPDL1 Randomized cT1N1-3 Atezo Carbo + nab-paclitaxel AC/EC/FEC pCR 48.6% vs. 44.4%, P = 0.48
(n = 280) phase III cT2-4N0-3 EFS immature
GeparNuevo Randomized cT2-4N0-3 Durva 2 wk durva alone, then None pCR 53.4% vs. 44.2%, OR 1.45
(n = 174) phase II nab-paclitaxel, then 3-yr DDFS 91.4% vs. 79.5%,
AC HR 0.37, P = 0.0148
3-yr OS 95.1% vs. 83.1%,
HR 0.26, P = 0076
I-SPY2 Randomized CT2-4dN0-3 Pembro Paclitaxel, then AC Physician’s discretion pCR 60% vs. 22% in TNBC
(n = 29 TNBC) phase II

Cite this article as Cold Spring Harb Perspect Med 2023;13:a041332


(69 total)
NCI10013 Randomized cT2-4Nany Atezo Carbo + paclitaxel ddAC pCR 55.6% vs. 18.8%, P = 0.018
(n = 67) phase II pCR in mBRCA 80% vs. 50%
NeoPACT Single-arm Stage I–III Pembro Carbo + docetaxel Physician’s discretion pCR 60%
(n = 117) phase II 2-yr EFS 89%
CHARIOT Single-arm Stage III Ipi + nivo Paclitaxel Nivo pCR 24.2%
(n = 34) phase II >15 mm RD (AC prior to 1-yr EFS 85%
after AC × 4 randomization)
I-SPY2 Randomized Stage II–III Durva + Paclitaxel, then AC None pCR 47% vs. 27% in TNBC
(n = 21 TNBC) phase II olaparib
(73 total)
(ICI) Immune checkpoint inhibitor, ( pembro) pembrolizumab, (atezo) atezolizumab, (durva) durvalumab, (nivo) nivolumab, (ipi)
ipilimumab, (carbo) carboplatin, (AC) doxorubicin and cyclophosphamide, (EC) epirubicin and cyclophosphamide, (dd) dose dense,
( pCR) pathological complete response, (EFS) event-free survival, (DDFS) distant disease-free survival, (OS) overall survival, (ITT) intent to
treat, (RD) residual disease, (cape) capecitabine, (mBRCA) mutant breast cancer gene, (PD-L1) programmed death ligand 1, (HR) hazard
Downloaded from http://perspectivesinmedicine.cshlp.org/ on June 26, 2023 - Published by Cold Spring Harbor Laboratory Press

ratio, (OR) odds ratio.


Breast Cancer Immunotherapy

3
Downloaded from http://perspectivesinmedicine.cshlp.org/ on June 26, 2023 - Published by Cold Spring Harbor Laboratory Press

L.A. Emens and S. Loi

survival (OS) with neoadjuvant durvalumab risk of future cancers (Ménétrier-Caux et al.
and chemotherapy independent of pCR, and 2019). Further defining the immunomodulatory
survival data from IMpassion031 are awaited. activity of distinct chemotherapies will help op-
The BrighTNess trial previously demonstrated timize ICI efficacy and maintain patients’ lym-
EFS benefit with carboplatin added to paclitaxel phocyte pool long term.
followed by 3-wk AC, but the CALGB 40603
trial did not, so the utility of carboplatin for early
Less Cytotoxic Neoadjuvant Chemotherapy
TNBC remains a topic of debate (Geyer et al.
2022; Shepherd et al. 2022). Moreover, whether It may be possible to reduce the intensity of the
intensification of chemotherapy with carbopla- chemotherapy backbone with neoadjuvant ICI
tin is needed when pembrolizumab is added to for two main reasons. First, attaining a pCR is
neoadjuvant chemotherapy is unknown. associated with excellent outcomes regardless of
Defining the short- and long-term impact of the therapy used. Second, post-neoadjuvant
chemotherapy on antitumor immunity will in- treatment provides an opportunity to escalate
form the optimum chemotherapy backbone for or de-escalate subsequent therapy according to
ICI. Different chemotherapy classes induce dis- the pathologic response. Because adding ICI to
tinct types of tumor cell death with varying neoadjuvant chemotherapy results in higher
mechanisms of potential immunologic synergy pCR rates, evaluating ICIs in combination
with ICIs (Emens and Middleton 2015; Park with less toxic neoadjuvant chemotherapy regi-
et al. 2020). For example, anthracyclines can in- mens is an attractive strategy for reducing the
duce immunogenic cell death, which may en- risk of long-term cardiotoxicity and secondary
hance dendritic cell uptake of tumor antigens hematological malignancies associated with an-
(Mattarollo et al. 2011; Montico et al. 2018). thracyclines in lower-risk patients with early
www.perspectivesinmedicine.org

Taxanes inhibit microtubule formation and TNBC. The single-arm phase 2 NeoPACT trial
may bind to Toll-like receptor 4 (TLR4) to tested six cycles of neoadjuvant pembrolizumab,
promote innate immunity (Pfannenstiel et al. carboplatin, and docetaxel (Sharma et al. 2022),
2010). This immune-modulating activity en- with a pCR rate of 60% and a 2-yr EFS rate of
hances natural killer cell function and decreases 88% without adjuvant pembrolizumab therapy.
both T regulatory cells and myeloid-derived Patients with higher immune activation (TILs
suppressor cells (MDSCs) (Muraro et al. 2015). ≥30%) had higher pCR rates. The key to suc-
DNA-damaging therapies also stimulate im- cessful chemotherapy de-escalation to improve
mune surveillance through necrotic cell death, clinical outcomes and reduce toxicity lies in ac-
a source of type I interferons. During mitosis, curate selection of patients most likely to achieve
cells with double-stranded DNA breaks accu- pCR with ICIs.
mulate micronuclei sensed by the cyclic GMP-
AMP synthase stimulator of the interferon genes
Postoperative ICI Considerations
(cGAS-STING) pathway, culminating in in-
creased interferon signaling (Zierhut et al. Post-neoadjuvant therapy provides an opportu-
2019; Serpico et al. 2020). nity to escalate or de-escalate adjuvant therapy
While chemotherapy may enhance tumor according to pathological response. The impact
immunogenicity, the potential for long-term of continuing adjuvant pembrolizumab for pa-
negative impact on adaptive immunity has also tients who do not experience a pCR with neo-
been recognized. A reduction in the naive T-cell adjuvant pembrolizumab and chemotherapy is
pool occurs with more intensive chemotherapy unclear. The GeparNuevo trial demonstrated a
regimens (Verma et al. 2016; Gustafson et al. statistically significant improvement in long-
2020) and may be worsened by adjuvant radio- term clinical outcomes from neoadjuvant ICI
therapy (Mozaffari et al. 2007, 2009; Venkate- despite not including an adjuvant ICI phase.
sulu et al. 2018). Lymphopenia is associated In exploratory subgroup analysis, even patients
with adverse clinical outcomes and a higher who did not experience pCR had clinically

4 Cite this article as Cold Spring Harb Perspect Med 2023;13:a041332


Downloaded from http://perspectivesinmedicine.cshlp.org/ on June 26, 2023 - Published by Cold Spring Harbor Laboratory Press

Breast Cancer Immunotherapy

meaningful improvements from durvalumab, and 6.7% of patients in the pembrolizumab


including improved 3-yr iDFS (76.3% vs. and placebo arms, respectively) have very poor
69.7%), DDFS (84.3% vs. 71.9%), and OS outcomes irrespective of treatment, implying
(92.0% vs. 78.8%), suggesting that clinical ben- primary resistance to both chemotherapy
efit can be derived from neoadjuvant-only ad- and immunotherapy (3-yr EFS 26.2% vs. 24.6%
ministration of ICI even in the absence of pCR in the pembrolizumab and placebo arms, re-
(Loibl et al. 2022). Notably, patients who do spectively [HR 1.24, 95% CI 0.69–2.23]). These
experience pCR with durvalumab in Gepar- patients are in critical need of novel non-cross-
Nuevo had a 3-yr DDFS and OS of 100%, sug- resistant therapies. New immunotherapeutic
gesting adjuvant ICI can be omitted in these strategies that circumvent intrinsic immune re-
patients. While this question remains to be an- sistance such as personalized cancer vaccines,
swered, in the future other trials without the adoptive T-cell therapy or immune agonists
adjuvant ICI component will also provide are particularly attractive.
long-term clinical outcome data that may give Two trials (SWOG1418 and A-BRAVE) are
confidence in this approach to those that achieve evaluating the role of ICI in the adjuvant phase
pCR. Notably, those patients who achieved pCR only specifically, with results pending (Conte
or residual cancer burden class I (RCB-I) in et al. 2020). SWOG1418 is evaluating adjuvant
KEYNOTE-522 with or without neoadjuvant pembrolizumab in patients who do not experi-
pembrolizumab also had excellent clinical out- ence pCR with neoadjuvant chemotherapy (no
comes. In patients with RCB-II, pembrolizumab prior ICI), while A-BRAVE is also evaluating
significantly improved 3-yr EFS (75.5% in the adjuvant ICI in this group as well as those who
pembrolizumab arm vs. 55.9% in the placebo receive no neoadjuvant therapy. These are im-
arm, HR 0.52, 95% CI 0.32–0.82) (Pusztai portant questions for patients who have not had
www.perspectivesinmedicine.org

et al. 2022), although it is unknown whether the opportunity to access neoadjuvant ICI, as
this is mediated by the neoadjuvant or adjuvant well as potentially tailoring treatment based on
pembrolizumab phase, or both. Taken together, a pathological result post-neoadjuvant chemo-
these data from KEYNOTE-522 and Gepar- therapy approach without ICI.
Nuevo support a clinical trial to evaluate cessa-
tion versus continuation of adjuvant ICI in pa-
Toxicity Profiles
tients who achieve pCR and RCB-I. The
Optimice-pCR trial is planned to address this Relevant to breast cancer, women may be at
question, randomizing patients with pCR fol- higher risk of immune-related adverse events
lowing neoadjuvant chemo-immunotherapy to (irAEs) from immunotherapy than men
an additional 27 wk of pembrolizumab or ob- (Özdemir et al. 2018; Miceli et al. 2021). For
servation. patients with curable disease, the risk of perma-
Outside of these trials, patients with residual nent or life-threatening treatment-related toxic-
disease after neoadjuvant chemotherapy cur- ity is set at a lower threshold than in the ad-
rently receive adjuvant capecitabine, or adjuvant vanced disease setting, especially as some
olaparib when a germline BRCA1/2 alteration is patients will be cured with chemotherapy alone.
present. Whether the incorporation of neoadju- Although immunotherapy is generally well-tol-
vant or adjuvant pembrolizumab to these agents erated and the most common irAEs are readily
adds further benefit is unknown. manageable, the idiosyncratic nature of perma-
In KEYNOTE-522, patients with RCB-II nent endocrinopathies such as primary adrenal
had a partial pathological response to neoadju- insufficiency, hypophysitis, and autoimmune
vant treatment. The long-term improvements in diabetes mellitus can be problematic. In KEY-
this population imply that they have mounted NOTE-522, any-grade irAE occurred in 33.5%
meaningful antitumor immunity that results in of patients receiving pembrolizumab (compared
improved clinical outcomes. In contrast, all pa- to 11.3% in the placebo group), including hypo-
tients with RCB-III in KEYNOTE-522 (5.1% thyroidism in 15.1%, primary adrenal insuffi-

Cite this article as Cold Spring Harb Perspect Med 2023;13:a041332 5


Downloaded from http://perspectivesinmedicine.cshlp.org/ on June 26, 2023 - Published by Cold Spring Harbor Laboratory Press

L.A. Emens and S. Loi

ciency in 2.6%, and hypophysitis in 1.9% in early-stage disease is unclear. However, high-
(Schmid et al. 2020a). Three immune-related er tumor burdens are associated with poorer
deaths occurred in the pembrolizumab arm outcomes from ICI in the advanced setting
( pneumonitis, immune-related encephalitis, (Tarantino et al. 2020). The NeoTrip trial in-
and pulmonary embolus). Adrenal insufficiency cluded higher-stage N2/N3 primary disease
(due to hypophysitis or primary adrenal insuf- and suggested greater clinical benefit from ICI
ficiency) was reported in 8.7% of patients treated in patients with PD-L1+ disease, although EFS
with pembrolizumab in I-SPY-2, higher that outcomes are not yet reported (Gianni et al.
what has previously been reported for anti- 2022). In multivariate analysis, PD-L1 positivity
PD-1 therapy (Nanda et al. 2020). Immunother- was the most significant factor associated with
apy also has unknown survivorship implica- pCR (odds ratio 2.08, P < 0.0001) in this trial.
tions. Recently, ICIs were reported to mediate Data from patients presenting with stage 4 de
ovarian inflammation and reduce oocyte re- novo disease treated with PD-1/PD-L1 agents
serves in a murine model (Winship et al. 2022). may be helpful in understanding the impact of
If confirmed in patients, this effect could have tumor burden relative to prior chemotherapy in
implications for women of reproductive age re- the requirement for PD-L1 IHC expression.
ceiving immunotherapy in the curative setting, Given long-term toxicity concerns, it is de-
particularly given the detrimental impact of che- bated whether all early-stage patients need neo-
motherapy on fertility. Further clinical research adjuvant ICI with their chemotherapy, particu-
assessing menstrual cycle patterns, menopausal larly those presenting with node-negative
status, fertility, and changes in sex steroid/go- disease. Clinical and preclinical data support
nadotropin levels with ICI treatment is needed. that the addition of PD-1 targeting agents to
chemotherapy as compared with chemotherapy
www.perspectivesinmedicine.org

alone significantly augments the antitumor T-


Patient Selection
cell response and results in long-term immune
The major challenge to the optimal use of protection from recurrence. It is hoped that in
immunotherapy in early-stage TNBC is patient the future these agents can be pharmacologically
selection. PD-L1 protein expression is an estab- improved to just target tumor-specific T cells,
lished predictive biomarker of response to which will hopefully alleviate many of the im-
PD-1/PD-L1 inhibitors for metastatic TNBC, mune toxicity concerns.
despite varying methods of PD-L1 assessment
(Schmid et al. 2018; Cortes et al. 2020). In both
Current Status and Future Directions
KEYNOTE-522 and IMpassion031, improved
pCR rates were associated with the use of pem- Adding ICI to neoadjuvant chemotherapy both
brolizumab or atezolizumab, respectively, in achieves a pCR in more patients and improves
both PD-L1+ and PD-L1-negative patients. In long-term clinical outcomes even in patients
KEYNOTE-522 and GeparNuevo, clinical ben- with residual disease at surgery. High-risk pa-
efit with pembrolizumab or durvalumab and tients (large tumor size or node-positive tu-
chemotherapy was also observed regardless of mors) without a medical contraindication (ac-
PD-L1 status. Hence, PD-L1-negative patients tive autoimmune disease, for example) should
should not be excluded from receiving ICIs receive a PD-1/PD-L1 inhibitor in combination
with neoadjuvant chemotherapy, as patients can with neoadjuvant chemotherapy (Emens et al.
generate an antitumor immune response that 2021c). Future clinical trials aim to optimize the
translates into clinical benefit regardless of chemotherapy backbone and/or duration of im-
PD-L1 status. Thus, there is currently no indi- munotherapy according to the TIL level or PD-
cation for assessing PD-L1 expression in a newly L1 expression. For example, it is plausible that
diagnosed patient with early-stage TNBC. some patients with stage I/II disease and high
Why PD-L1 expression predicts immuno- TIL or PD-L1+ disease could benefit from neo-
therapy response in metastatic disease but not adjuvant taxane with short course ICI.

6 Cite this article as Cold Spring Harb Perspect Med 2023;13:a041332


Downloaded from http://perspectivesinmedicine.cshlp.org/ on June 26, 2023 - Published by Cold Spring Harbor Laboratory Press

Breast Cancer Immunotherapy

Biomarkers that predict response to neoad- lowed by paclitaxel, trastuzumab, and pertuzu-
juvant ICI are needed. On-treatment change in mab (Huober et al. 2022). There was no
TILs and PD-L1 protein expression, immune significant difference in pCR between the two
gene signatures, homologous recombination de- arms regardless of PD-L1 expression, and EFS
ficiency (HRD), CD274 amplification, and TMB outcomes remain immature. Moreover, there
(Karn et al. 2020; Bianchini et al. 2021) have were four deaths in the atezolizumab arm and
been evaluated and a few have been found to none in the placebo arm; two deaths were attrib-
be predictive of benefit from ICI specifically. In uted to treatment. Atezolizumab plus trastuzu-
GeparNuevo, only an increase in intratumoral mab emtansine patients with residual disease
TIL from baseline to cycle 1, day 15 significantly after standard neoadjuvant therapy for HER2+
predicted benefit from durvalumab and not pla- disease is being evaluated in the randomized
cebo (Loibl et al. 2019). High TMB and immune phase III ASTEFANIA trial (NCT04873362).
gene-expression profile were associated with in- HR+ breast cancer overall is associated with
creased odds of pCR in both treatment arms (Karn a good prognosis. However, certain patient sub-
et al. 2020). Stromal TILs (sTILs) and up-regula- groups have a worse prognosis, including those
tion of immune signaling pathways at cycle 2, day who are premenopausal, and/or whose disease
1 was associated with pCR in both atezolizumab is high grade, has greater nodal involvement,
and chemo-alone arms of NeoTrip (Bianchini or has low estrogen receptor (ER) expression.
et al. 2021). Spatial connectivity of PD-L1-express- High-grade disease is associated with higher
ing immune cells relative to epithelial cells appears TIL levels, even in HR+ disease. HR+ HER2-
to be relevant to response to atezolizumab in negative (HER2neg) patients were included in
NeoTrip (Bianchini et al. 2022), suggesting that I-SPY2, with an improvement in pCR rates
categorical or continuous characterization of indi- from 13% to 30% (Table 2; Nanda et al. 2020).
www.perspectivesinmedicine.org

vidual immune markers such as PD-L1 and TILs The GIADA trial enrolled 43 patients with stage
may be too simplistic in this disease. II–III luminal B breast cancer (HR+, HER2neg,
The pCR end point is not a perfect surrogate Ki67 ≥ 20%, and/or grade 3) to receive three
end point for either neoadjuvant chemotherapy cycles of EC followed by eight cycles of 2-wk
alone (Conforti et al. 2022) or combined with nivolumab, with a pCR rate of 16.3% (Dieci
ICI. Assessment of pathologic response as a sur- et al. 2022). Checkmate 7FL (NCT04109066)
rogate for long-term outcome may be strength- and KEYNOTE-756 (NCT03725059) are ran-
ened by combining it with biomarkers such as domized phase III trials investigating either
circulating tumor cells, circulating tumor DNA, nivolumab or pembrolizumab, respectively, in
and/or TILs in residual disease (Luen et al. combination with neoadjuvant AC- and tax-
2019). Finally, the clinical benefit of adjuvant ane-based chemotherapy, followed by adjuvant
immunotherapy used alone or continued after ICI with endocrine therapy in high-grade HR+
neoadjuvant therapy remains an open question breast cancers. Checkmate 7FL closed to accrual
that will be informed by the results of current early after the MonarchE trial showed signifi-
clinical trials. cant benefit for adjuvant abemaciclib (Harbeck
et al. 2021), which cannot be safely combined
with ICI. KEYNOTE-756 has completed accru-
Immunotherapy in Hormone Receptor-
al; pCR and EFS analyses are planned.
Positive (HR+) and HER2+ Early Breast Cancer
Immunotherapy is being actively investigated in
Combination Strategies
HR+ and HER2+ breast cancer (Table 2). Pa-
tients with HER-2-driven disease have high lev- Combinations of PD-1/PD-L1 agents with other
els of TILs and high pCR rates with neoadjuvant immune-modulatory agents may improve pCR
chemotherapy plus dual HER2 blockade. rates for patients unlikely to respond to ICI
IMpassion050 investigated the addition of ate- alone (Table 3). The single-arm CHARIOT trial
zolizumab or placebo to neoadjuvant AC fol- recruited 34 patients with stage III TNBC with

Cite this article as Cold Spring Harb Perspect Med 2023;13:a041332 7


Downloaded from http://perspectivesinmedicine.cshlp.org/ on June 26, 2023 - Published by Cold Spring Harbor Laboratory Press

L.A. Emens and S. Loi

poor response to neoadjuvant AC to receive TNBC for immunotherapy (the SP142 assay
combination ipilimumab and nivolumab with for atezolizumab and the 22C3 assay for pem-
neoadjuvant weekly paclitaxel, resulting in brolizumab were used in the trials). These PD-
pCR rates of 24.4%, 37.5%, and 23% in the over- L1 assays are not the same, as demonstrated in a
all, PD-L1+ and PD-L1-negative populations, systematic analysis of patient samples evaluating
respectively (Loi et al. 2022b). The combination the association of PD-L1 status and clinical re-
of PARP inhibitors and immunotherapy is sponse from the IMpassion130 study (Rugo
particularly attractive due to the prevalence of et al. 2021).
HRD in breast cancer. HRD is a genomic The combination of chemotherapy and
feature associated with increased immune ac- pembrolizumab was evaluated in KEYNOTE
tivation via up-regulation of the cGAS/STING 355, a global clinical study that randomized
pathway (Parkes et al. 2022; van Vugt et al. 847 patients with advanced TNBC to receive
2022). I-SPY2 evaluated the PARP-inhibitor pembrolizumab or placebo and chemotherapy
olaparib with durvalumab and paclitaxel, fol- with nab-paclitaxel, paclitaxel, or gemcitabine
lowed by AC, for patients with HER2neg early and carboplatin (Cortes et al. 2020, 2022). Eli-
breast cancer, and demonstrated numerically gible patients had untreated advanced disease
improved pCR rates compared to chemotherapy with a treatment-free interval from (neo)adju-
alone in both the HR+ and TNBC cohorts (Pusz- vant therapy of ≥6 mo. Primary end points in-
tai et al. 2021). cluded PFS and OS in the overall patient popu-
lation, and in patients with PD-L1 CPS scores of
≥10 or ≥1. Pembrolizumab given with chemo-
ADVANCED BREAST CANCER therapy improved both PFS (Δ4.1 mo, HR 0.65,
P = 0.0012) and OS (Δ6.9 mo, HR 0.73, P =
www.perspectivesinmedicine.org

Metastatic TNBC—First-Line ICIs


0.0093) in PD-L1+ CPS ≥10 patients. Based on
Metastatic TNBC has historically been treated these data, pembrolizumab with chemotherapy
with chemotherapy, with bevacizumab some- is approved in the United States and globally for
times added ex-U.S. Recent clinical progress the first-line treatment of patients with ad-
has expanded treatment options for patients, vanced PD-L1+ TNBC.
which now include targeted agents and immu- IMpassion130 is a global clinical trial that
notherapy (Huppert et al. 2022). The PARP in- enrolled 902 patients with advanced TNBC,
hibitors olaparib and niraparib (for germline randomizing them to nab-paclitaxel with atezo-
BRCA-mutated breast cancer) and the anti- lizumab or placebo (Schmid et al. 2018, 2020a;
body-drug conjugates sacituzumab govitecan Emens et al. 2021b). Patients had treatment-na-
(specific for the cell-surface antigen TROP2) ive metastatic disease, with a treatment-free in-
and trastuzumab deruxtecan (specific for terval of ≥12 mo from (neo)adjuvant therapy.
HER2 and active in patients with HER2-low Primary end points included PFS and OS in the
TNBC) are targeted therapies available for rou- overall and PD-L1+ patient groups. The study
tine use in the United States. Pembrolizumab showed an improvement in PFS with the addi-
with chemotherapy (based on KEYNOTE 355; tion of atezolizumab to nab-paclitaxel in both
Cortes et al. 2022) is a standard of care for the the overall (Δ2.1 mo, HR 0.69, P = 0.0025) and
first-line treatment of metastatic PD-L1+ TNBC PD-L1+ (Δ2.5 mo, HR 0.62, P < 0.0001) groups.
in the United States and globally, and atezolizu- A trend toward improved OS in the overall pa-
mab with nab-paclitaxel (based on IMpassion tient population was not statistically significant,
130) is approved for the first-line treatment of but an exploratory OS analysis in PD-L1+ pa-
metastatic PD-L1+ TNBC (Schmid et al. 2018; tients revealed a clinically meaningful OS im-
Emens et al. 2021b) outside the United States. provement that was not formally tested due to
These trials are summarized in Table 3. In con- the hierarchical statistical analysis plan for OS.
trast to the neoadjuvant setting, PD-L1 expres- These results resulted in accelerated approval by
sion is used to select patients with advanced the U.S. FDA. The OS for PD-L1+ patients re-

8 Cite this article as Cold Spring Harb Perspect Med 2023;13:a041332


www.perspectivesinmedicine.org

Table 2. Immune checkpoint inhibitor (ICI) trials in early HER2+ and HR+ breast cancer
Clinical trial Disease type Chemotherapy
(sample size) Design and stage ICI backbone Adjuvant therapy Results
+
IMpassion050 Randomized HER2 Atezo ddAC, then THP Atezo + HP if pCR pCR in ITT 62.4% vs.
(n = 454) phase III cT2-4N1-3 T-DM1 if RD 62.7%, P = 0.9551
I-SPY2 Randomized cT2-4dN0-3 Pembro Paclitaxel, then AC Physician’s pCR 30% vs. 13% in HR+
(n = 40 HR+) phase II discretion
(69 total)
GIADA Single-arm Stage II–IIIA Nivo EC × 3, OFS OFS + exemestane pCR 16.3%
(n = 43) phase II premenopausal exemestane
luminal B
I-SPY2 Randomized Stage II–III Durva + Paclitaxel, then AC None pCR 28% vs. 14% in HR+
(n = 52) phase II olaparib
(73 total)
KEYNOTE- Randomized T1c-2N1-2 Pembro Paclitaxel, then AC Pembro × 9 + ET Primary end points

Cite this article as Cold Spring Harb Perspect Med 2023;13:a041332


756 phase III T3-4N0-2 pCR and EFS
Grade 3 Completed accrual
Checkmate 7FL Randomized T1c-2N1-2 Nivo Paclitaxel, then AC Nivo × 7 + ET Terminated early due to a
phase III T3-4N0-2 change in the standard
Grade 3 of care
ER 1%–9%
(HER2) Human epidermal growth factor 2, (HR) hormone receptor, (atezo) atezolizumab, ( pembro) pembrolizumab, (nivo)
nivolumab, (durva) durvalumab, (AC) adriamycin and doxorubicin, (THP) paclitaxel, trastuzumab, and pertuzumab, (HP)
trastuzumab and pertuzumab, (EC) epirubicin and cyclophosphamide, (OFS) ovarian function suppression, (ET) endocrine therapy,
(ER) estrogen receptor, ( pCR) pathologic complete response, (RD) residual disease, (EFS) event-free survival, (ITT) intent to treat.
Downloaded from http://perspectivesinmedicine.cshlp.org/ on June 26, 2023 - Published by Cold Spring Harbor Laboratory Press

Breast Cancer Immunotherapy

9
Downloaded from http://perspectivesinmedicine.cshlp.org/ on June 26, 2023 - Published by Cold Spring Harbor Laboratory Press

L.A. Emens and S. Loi

Table 3. Immune checkpoint inhibitor (ICI) trials for the first-line therapy of metastatic triple-negative breast
cancer (TNBC)
Clinical trial Patient Chemotherapy
(sample size) Design population ICI backbone Results
KEYNOTE-355 Randomized Untreated Pembro vs. Nab-paclitaxel ITT: PFS 7.5 mo vs. 5.6 mo,
(n = 847) phase III advanced placebo Paclitaxel HR 0.82, P = not tested
TNBC Gemcitabine + ITT: OS 17.2 vs 15.5 mo, HR
TFI >6 mo carboplatin 0.89, P = not tested
CPS >10: PFS 9.7 vs 5.6 mo,
HR 0.65, P = 0.0012
CPS ≥10: OS 23.0 vs. 16.1 mo,
HR 0.73, P = 0.0185
IMpassion130 Randomized Untreated Atezo vs. Nab-paclitaxel ITT: PFS 7.2 mo vs. 5.5 mo,
(n = 902) phase III advanced placebo HR 0.80, P = 0.002
TNBC ITT: OS 21.0 vs. 18.7 mo, HR
TFI ≥12 mo 0.87, P = not tested
PD-L1+: PFS 7.5 vs. 5.0 mo,
HR 0.62, P < 0.001
PD-L1+: OS 25.4 vs. 17.9 mo,
HR 0.67, P = not tested
IMpassion131 Randomized Untreated Atezo vs. Paclitaxel PD-L1+: PFS 5.7 mo vs. 6.0
(n = 651) phase III advanced placebo mo, HR 0.82, P = 0.002
TNBC PD-L1+: OS 22.1 vs. 28.3 mo,
TFI ≥12 mo HR 1.11 P = not tested
www.perspectivesinmedicine.org

(TFI) Treatment-free interval, ( pembro) pembrolizumab, (atezo) atezolizumab, (ITT) intent to treat, (PD-L1) programmed
death ligand 1, (PFS) progression-free survival, (OS) overall survival, (HR) hazard ratio.

mained consistent over time, with improve- findings, was a global clinical trial that enrolled
ments of 7.5 (HR 0.62), 7.0 (HR 0.71), and 7.5 a similar population of 651 patients (Miles et al.
(HR 0.67) mo at the first, second, and final anal- 2021). It randomized patients to receive atezoli-
yses, respectively (median follow up 18.8 mo at zumab or placebo combined with paclitaxel
final analysis). Extensive biomarker analyses of (Table 3). The percentages of PD-L1+ patients
IMpassion130 tumor specimens revealed that in IMpassion 130 and IMpassion 131 were 40%
enriching for PD-L1 was not associated with and 45%, respectively. Adding atezolizumab to
greater clinical benefit, and patients with PD- paclitaxel did not improve PFS or OS in PD-
L1-negative disease had no treatment effect L1+ patients (Δ0.3 mo, HR 0.82, P = 0.20, and
(Emens et al. 2021a). Only 9% of tumors ex- Δ −6.2 mo, HR 1.11, respectively). The reasons
pressed PD-L1 on tumor cells, and 2% of tumors for the discordant results between the trials are
expressed PD-L1 exclusively on tumor cells. Im- unclear. They may relate to the steroid exposure
proved clinical outcomes were observed in tu- required for paclitaxel or unknown biological
mors with CD8+ T cells and sTILs only if the differences between the patient populations.
tumors were PD-L1+. BRCA1/2 mutations were Given the negative results of IMpassion 131
present in 14.5% (89/612) of patients, and not and the lack of statistically significant OS data
associated with PD-L1 status. PD-L1+ patients from IMpassion 130, the approval of pembroli-
derived clinical benefit regardless of BRCA1/2 zumab left no clear regulatory path forward for
mutation status. atezolizumab combined with nab-paclitaxel in
IMpassion 131, an independent companion the United States, and the accelerated approval
trial designed to independently evaluate these indication was withdrawn by the sponsor. Ate-

10 Cite this article as Cold Spring Harb Perspect Med 2023;13:a041332


Downloaded from http://perspectivesinmedicine.cshlp.org/ on June 26, 2023 - Published by Cold Spring Harbor Laboratory Press

Breast Cancer Immunotherapy

zolizumab and nab-paclitaxel is approved by The FUTURE-C-PLUS trial evaluated the


regulatory authorities outside the United States. antiangiogenic agent famitinib combined with
the PD-1 antibody camrelizumab and nab-pac-
litaxel as first-line therapy in 48 advanced CD8+
Metastatic TNBC—ICIs Second Line and
T-cell-enriched immunomodulatory TNBC pa-
Beyond
tients (Wu et al. 2022). The ORR was 81.3%,
Immunotherapy has been less active in meta- with a median PFS of 13.6 mo. Clinical benefit
static TNBC beyond the first-line setting (Table was enhanced in CD8+ T-cell-enriched and PD-
4). The phase 3 KEYNOTE 119 study evaluated L1+ tumors, and the PKD1 somatic mutation
pembrolizumab monotherapy versus chemo- was associated with resistance. The TOPACIO/
therapy of physician’s choice in a global, ran- KEYNOTE 162 trial is a single-arm trial that
domized, open label trial enrolling patients evaluated niraparib combined with pembrolizu-
with TNBC after one to two prior systemic mab in 55 patients (47 efficacy evaluable) with
therapies for metastatic disease (Winer et al. metastatic TNBC (Vinayak et al. 2019). The
2021). Patients received either pembrolizumab ORR was 21%, with a disease control rate
or chemotherapy with capecitabine, eribulin, (DCR) of 49%. In 15 efficacy evaluable patients
gemcitabine, or vinorelbine. Primary end with a germline BRCA mutation, the ORR was
points were OS in patients with PD-L1 CPS 47% (n = 7) with a DCR of 80% (n = 12).
≥10 and CPS ≥1 disease, and the overall pa- The randomized phase II SAFIR02 Breast
tient population. Pembrolizumab did not im- Immuno trial tested maintenance therapy with
prove OS relative to chemotherapy in any chemotherapy versus durvalumab in patients
group, although exploratory analyses revealed with metastatic HER2neg breast cancer with
a potential survival benefit in the CPS ≥20 sub- stable disease after induction chemotherapy
www.perspectivesinmedicine.org

group. These findings indicate that novel treat- (Batchelot et al. 2021). There was no clinical
ment and/or patient-selection strategies are activity of durvalumab in the overall population,
needed for patients with previously treated ad- but in 82 patients with TNBC, durvalumab im-
vanced TNBC. proved OS (HR 0.54, P = 0.0377) and CD274
The TONIC study is a noncomparative, gain/amplification was identified as a potential
adaptive phase 1/2 clinical trial that evaluated biomarker of sensitivity.
nivolumab alone or after 2 wk priming with im-
munomodulatory doses of cyclophosphamide
Immunotherapy for Metastatic HER2+ Breast
(50 mg orally daily), cisplatin (2 × 40 mg/m2 in-
Cancer
travenously), doxorubicin (2 × 15 mg intrave-
nously), or radiation therapy (3 × 8 Gy) in 67 The major challenge of developing immuno-
patients with metastatic TNBC, 24% of whom therapy for metastatic HER2+ breast cancer is
had no prior treatment for metastatic disease the availability of multiple HER2-directed ther-
(Voorwerk et al. 2019). Biopsies were collected apeutics with survival benefit. These highly ef-
at baseline, and after priming and 3 wk of nivo- fective drugs raise the bar for demonstrating
lumab. The overall objective response rate enhanced clinical activity with immunotherapy
(ORR) by iRECIST was 20%, with ORRs of combinations (Table 5), while simultaneously
23% and 35% with cisplatin and doxorubicin creating hope for curing patients with ad-
priming, respectively. Correlative studies re- vanced HER2+ breast cancer. Several HER2-
vealed up-regulation of immune-related genes specific therapeutics have intrinsic immune-
in the PD-1 and T-cell cytotoxicity pathways modulating activity, creating an opportunity
with both drugs, as well as inflammation and for therapeutic synergy. A phase 1b study eval-
both JAK/STAT and TNF-α signaling with dox- uated adding atezolizumab to ado-trastuzumab
orubicin. Induction with low-dose doxorubicin emtansine (TDM1) or trastuzumab/pertuzu-
followed by nivolumab versus nivolumab alone mab with docetaxel in patients with metastatic
is currently being evaluated. HER2+ breast cancer (Hamilton et al. 2021).

Cite this article as Cold Spring Harb Perspect Med 2023;13:a041332 11


www.perspectivesinmedicine.org

12
Table 4. Trials testing immune checkpoint inhibitors (ICIs) in metastatic triple-negative breast cancer (TNBC) beyond the first-line setting
Clinical trial
(sample size) Design Patient population ICI Intervention Results
KEYNOTE-119 Randomized Advanced TNBC Pembro Pembro alone vs. physicians’ OS:
(n = 622) open label second or third choice chemo: capecitabine, CPS ≥10: 12.7 vs. 11.6 mo, HR 0.78, P = 0.057
L.A. Emens and S. Loi

phase III line eribulin, gemcitabine, CPS ≥1: 10.7 vs. 10.2 mo, HR 0.86, P = 0.073
vinorelbine
ITT: 9.9 vs. 10.8 mo, HR 0.97, P = nt
CPS ≥20: 14.9 vs. 12.5 mo, HR 0.58, P = nt
TONIC Noncomparative Metastatic TNBC Nivo Nivo alone vs. nivo beginning ORR:
(n = 67) adaptive phase I/II after 2 wk priming with: Overall 20%
CY 50 mg orally daily Nivo 17%
Cis 2 × 40 mg/m2 IV CY 8%
Dox 2 × 15 mg IV Cis 23%
XRT 3 × 8 Gy Dox 35%
XRT 8%
FUTURE-C-PLUS Single-arm phase II Advanced CD8+ T- Camre Camrelizumab + famitinib, nab- ORR 81.3%, PFS 13.6 mo
(n = 48) cell-enriched paclitaxel
metastatic TNBC
KEYNOTE-162 Single-arm phase II Metastatic TNBC Pembro Pembro + niraparib ITT (n = 47 efficacy evaluable):
(TOPACIO) ORR 21%
(n = 55) DCR 49%
PFS 2.3 mo
mBRCA (n = 15):
ORR 47% (n = 7)
DCR 80% (n = 12)
PFS 8.3 mo
wtBRCA (n = 27):
ORR 11% (n = 3)
DCR 33% (n = 9)
PFS 2.1 mo
Continued
Downloaded from http://perspectivesinmedicine.cshlp.org/ on June 26, 2023 - Published by Cold Spring Harbor Laboratory Press

Cite this article as Cold Spring Harb Perspect Med 2023;13:a041332


www.perspectivesinmedicine.org

Table 4. Continued
Clinical trial
(sample size) Design Patient population ICI Intervention Results
SAFIR02-BREAST Randomized phase II Metastatic HER2neg Durva Maintenance therapy durva vs. Overall:
IMMUNO breast cancer, chemo PFS HR 1.40, P = 0.047
(n = 199) stable after six to OS HR 0.84, P = 0.423
eight cycles of
TNBC (n = 82)
chemotherapy
All OS HR 0.54, P = 0.0377, P = nt
PD-L1+ (n = 32) OS HR 0.37, P = nt
PD-L1– (n = 24) OS HR 0.49, P = nt
CD274 gain/amplification (n = 23)
OS HR 0.18, P = 0.0059

Cite this article as Cold Spring Harb Perspect Med 2023;13:a041332


CD274 normal/loss (n = 32)
OS HR 1.12, P = 0.8139
(HER2neg) Human epidermal growth factor 2-negative, ( pembro) pembrolizumab, (nivo) nivolumab, (camre) camrelizumab, (durva)
durvalumab, (chemo) chemotherapy, (CY) cyclophosphamide, (Cis) cisplatin, (Dox) doxorubicin, (XRT) radiotherapy, (ITT) intent to
treat, (PD-L1) programmed death ligand 1, (CPS) combined positive score, (ORR) objective response rate, (DCR) disease control rate, (PFS)
progression-free survival, (OS) overall survival, (HR) hazard ratio, (Gy) gray, (IV) intravenous, (mBRCA) mutated breast cancer–related
gene, (wtBRCA) wild-type breast cancer–related gene, (nt) not tested.
Downloaded from http://perspectivesinmedicine.cshlp.org/ on June 26, 2023 - Published by Cold Spring Harbor Laboratory Press

Breast Cancer Immunotherapy

13
Downloaded from http://perspectivesinmedicine.cshlp.org/ on June 26, 2023 - Published by Cold Spring Harbor Laboratory Press

L.A. Emens and S. Loi

The ORR with atezolizumab and TDM1 was ICOS+ T cells increased, with a best response
35% (7/20 patients) and with atezolizumab of stable disease for ≥12 wk in 42% of patients.
plus trastuzumab, pertuzumab, and docetaxel The combination of tremelimumab and durva-
was 100% (6/6 patients). Immune cell PD-L1 lumab was tested in 18 patients with advanced
expression increased in patients treated with HER2neg breast cancer, 11 of whom had HR+
atezolizumab with TDM1 independent of clin- disease (Santa-Maria et al. 2018). Although no
ical response. responses occurred in ER+ disease, the ORR in
PANACEA evaluated the safety and clinical TNBC was 17%.
activity of trastuzumab and pembrolizumab in Pembrolizumab was given with eribulin in
55 patients with metastatic HER2+ breast cancer 44 patients with advanced HR+ breast cancer,
who had previously progressed on trastuzumab with an ORR of 41% (Perez-Garcia et al.
(Loi et al. 2019). The ORRs were 15.2% (7/42) in 2021). A randomized study enrolled 88 patients
PD-L1+ and 0% (0/12) PD-L1-negative patients, with advanced HR+ breast cancer to receive eri-
respectively. Moreover, 12-mo PFS and OS rates bulin with or without pembrolizumab, with
were 13% versus 0% and 65% versus 12% in crossover permitted (Tolaney et al. 2020). There
patients with PD-L1+ versus PD-L1-negative was no improvement in ORR (27% vs. 34%, P =
disease, respectively. sTILs tracked with clinical 0.49), PFS, or OS, and no association of PFS with
benefit in PD-L1+ patients. PD-L1, sTILs, TMB, or genomic alterations.
KATE2 is a randomized phase 2 trial that Computational analysis of 52 pretreatment tu-
added atezolizumab or placebo to TDM1 in mors showed an association between immune
202 patients with HER2+ metastatic breast can- infiltrates and antigen presentation pathways
cer previously treated with trastuzumab and a and clinical response, with resistant tumors
taxane (Emens et al. 2020). There was no sig- characterized by heterogeneity and active estro-
www.perspectivesinmedicine.org

nificant difference in PFS or OS in the overall gen signaling (Keenan et al. 2021).
population, but exploratory analyses suggested Another study evaluated epigenetic modu-
improved PFS and OS in PD-L1+ patients. lation with pembrolizumab in metastatic ER+
KATE3, a randomized phase 3 clinical trial is breast cancer, randomizing 34 patients to receive
enrolling biomarker-selected patients with met- tamoxifen and vorinostat with concurrent pem-
astatic HER2+ breast cancer that is also PD-L1+ brolizumab (n = 18), or with pembrolizumab
(NCT04740918). beginning after one cycle of priming with ta-
moxifen and vorinostat (n = 16) (Terranova-
Barberio et al. 2020). The ORR was 4%, with
Immunotherapy for Metastatic HR+ Breast
a clinical benefit rate of 19%. Another study
Cancer
evaluated entinostat with nivolumab and low-
HR-expressing breast cancer tends to lack T cells dose ipilimumab in 24 patients with advanced
and instead contains a significant population of HER2neg breast cancer; 12 had TNBC and 12
myeloid cells. Given the distinct immunobiol- had HR+ breast cancer. The ORR was 30%,
ogy of metastatic HR+ breast cancer, it is not mostly in TNBC. The CD8+ T cells increased
surprising that ICIs alone have displayed limited with therapy.
clinical activity. Pembrolizumab alone in 25 pa- Eftilagomad alpha (efti, IMP321) is a soluble
tients with ER+, HER2neg, PD-L1+ metastatic LAG-3 Ig fusion that binds to MHC class II
breast cancer had an ORR of 12% (Rugo et al. molecules, promoting immune priming. The
2018). Most trials for advanced HR+ breast can- AIPAC trial randomized 227 patients with met-
cer have thus focused on combination strategies astatic HR+ breast cancer to weekly paclitaxel
(Table 5). with efti or placebo as first-line chemotherapy
The anti-CTLA4-specific antibody tremeli- in endocrine-experienced patients (Wildiers
mumab combined with exemestane was evalu- et al. 2021). Paclitaxel was given with either efti
ated in 26 patients with advanced HR+ breast or placebo for six cycles, and then maintenance
cancer (Vonderheide et al. 2010). Peripheral efti or placebo was continued for 52 wk. There

14 Cite this article as Cold Spring Harb Perspect Med 2023;13:a041332


www.perspectivesinmedicine.org

Table 5. Selected trials testing immune checkpoint inhibitors (ICIs) in HER2+ and HR+ HER2-negative metastatic breast cancer
Clinical trial
(sample size) Design Patient population ICI Intervention Results
+
KATE2 (n = 202) Randomized Metastatic HER2 breast Atezo Atezo or placebo + T-DM1 ITT PFS 8.2 vs. 6.8 mo, HR 0.82, P = 0.33
phase II cancer previously treated PD-L1+ PFS 8.5 vs. 4.1 mo, HR 0.62
with trastuzumab and PD-L1– PFS 6.8 vs. 8.2, HR 1.06
taxane
PANACEA (n = 55) Phase Ib/II Metastatic HER2+ breast Pembro Pembro + trastuzumab PD-L1+ vs. PD-L1– cohorts:
parallel cancer progressed on ORR 15% vs. 0%
cohort trastuzumab 12-mo PFS 13% vs. 0%
12-mo OS rate 65% vs. 12%
AIPAC (n = 227) Randomized Metastatic HR+ breast cancer, Efti Paclitaxel with efti or placebo for six ORR 48.3% vs. 38.4%
phase IIb first-line chemo cycles, then maintenance efti vs. PFS at 6 mo 63% vs. 54%, HR 0.93,
placebo P = 0.341
SAFIR02-BREAST Randomized Metastatic HER2neg breast Durva Maintenance therapy durva vs. Overall (117/199 HR+)
IMMUNO phase II cancer, stable after six to chemo PFS HR 1.40, P = 0.047
(n = 199) eight cycles of chemo OS HR 0.84, P = 0.423

Potential benefit in TNBC subset
(n = 82)
Pembro + eribulin Randomized Metastatic HR+ breast cancer Pembro Pembro + eribulin vs. eribulin ORR 27% vs. 34%, P = 0.49
(n = 88) phase Ib/II PFS 4.1 vs. 4.2 mo, HR 0.80, P = 0.33

Cite this article as Cold Spring Harb Perspect Med 2023;13:a041332


Treme + exemestane Single-arm Advanced HR+ breast cancer Treme Treme + exemestane Stable disease ≥12 wk in 42%
(n = 26) phase I
Pembro + tamoxifen + Randomized Advanced HR+ breast cancer Pembro Concurrent tamoxifen + vorinostat ORR 4%
vorinostat phase II with pembro vs. one cycle of CBR 19%
(n = 34) priming with tamoxifen +
vorinostat followed by concurrent
therapy with pembro
MEDIOLA Single-arm gmBRCA HER2neg metastatic Durva 4 wk olaparib priming followed by Overall ORR 63%
(n = 30) phase II breast cancer concurrent olaparib with durva HR+ (n = 13) ORR 69%
(HER2) Human epidermal growth factor 2, (HER2neg) human epidermal growth factor 2-negative, (HR) hormone receptor, (TNBC)
triple-negative breast cancer, (atezo) atezolizumab, ( pembro) pembrolizumab, (treme) tremelimumab), (durva) durvalumab, (chemo)
chemotherapy, (efti) eftilagomad alpha, (T-DM1) trastuzumab emtansine, (ITT) intent to treat, (PD-L1) programmed death ligand 1,
(CPS) combined positive score, (ORR) objective response rate, (DCR) disease control rate, (PFS) progression-free survival, (OS) overall
survival, (HR) hazard ratio, (ORR) objective response rate, (gmBRCA) germline mutated breast cancer–related gene, (wtBRCA) wild-type
Downloaded from http://perspectivesinmedicine.cshlp.org/ on June 26, 2023 - Published by Cold Spring Harbor Laboratory Press

breast cancer–related gene.


Breast Cancer Immunotherapy

15
Downloaded from http://perspectivesinmedicine.cshlp.org/ on June 26, 2023 - Published by Cold Spring Harbor Laboratory Press

L.A. Emens and S. Loi

was no PFS difference (HR 0.93, P = 0.341), but with its utility currently limited to TNBC. Key
efti and paclitaxel improved OS in patients <65 goals for the field are to enhance the activity of
yr with low monocytes and more aggressive dis- immunotherapy for TNBC, and to expand its
ease by about 8 mo (22.3 vs. 14.8 mo, P = 0.17). use to other breast cancer subtypes. One major
A follow-up phase 3 clinical trial is planned. strategy is to combine ICIs with standard and novel
breast cancer therapies. Attractive combinations
include ICIs with newer antibody drug conjugates
Biomarker-Defined Metastatic Breast Cancer
(sacituzumab govitecan, fam-trastuzumab derux-
beyond Subtype
tecan, for example), radiotherapy, cyroablation,
The MEDIOLA study enrolled 30 patients with and new precision drugs targeting unique aspects
germline BRCA-mutated HER2neg metastatic of intrinsic tumor biology and/or the tumor im-
breast cancer to receive olaparib priming for 4 mune microenvironment (key signaling pathways
wk, followed by durvalumab with olaparib start- and metabolic circuits). Initial combinations have
ing at week 5; 17 patients had TNBC and the been discussed above. A second major strategy is
remainder were HR+ (Domchek et al. 2020). the development of innovative immunotherapies,
The overall ORR was 63%, with ORRs in TNBC including novel immune checkpoint modulators,
and HR+ disease of 59% and 69%, respectively. vaccines, adoptive cell therapies, and bispecific
The median duration of response was 9.2 mo. molecules.
Nine patients had early disease progression at
≤28 wk. Potential mechanisms of resistance
Novel Immune Checkpoints
identified included BRCA2 reversion, lack of
BRCA2 LOH, p53 mutation, and PD-L1/PD-L2 New immune checkpoints are under active in-
gene amplification. vestigation. The LAG-3-specific antibody relat-
www.perspectivesinmedicine.org

The TAPUR study tests commercially avail- limab is now approved for melanoma in combi-
able targeted agents in patients with advanced nation with nivolumab and represents the third
cancers and a predictive molecular alteration type of ICI approved for clinical use (Tawbi et al.
(Alva et al. 2021). This study treated 28 patients 2022). Clinical trials have tested antibodies spe-
with metastatic breast cancer (any subtype) and cific for TIM-3 and TIGIT, but clinical activity
a high TMB of 9–37 Mut/Mb. The ORR and has not been clearly demonstrated. Other prom-
DCR were 21% and 37%, respectively, and me- ising novel immune checkpoints under active
dian OS was 30.6 wk (95% CI 18.3–103.3). There clinical investigation include CD40, OX-40,
was no association between PFS and TMB. ICOS, B7-H3, and B7-H4.
The NIMBUS study tested nivolumab with
low-dose ipilimumab in 30 patients with ad-
Vaccines
vanced HER2neg breast cancer and a TMB ≥9
mut/Mb;21had HR+ disease,and eight were treat- Cancer vaccines have been tested with minimal
ed first-line (Barroso-Sousa et al. 2022). The over- clinical success. Therapeutic breast cancer vac-
all ORR was 17% (5/30 patients). Three patients cines were first tested in metastatic disease, and
had HR+ disease with TMBs of 110, 38, and 17.5, then in the adjuvant setting; all phase 3 clinical
whereas two TNBC responders had TMBs of 10.9 trials so far have been negative (Solinas et al.
and 9.1, with the first also PD-L1+. Patients with 2020). There is now increasing interest in apply-
TMB ≥14 had an ORR of 60%, suggesting pro- ing cancer vaccines for disease interception and
spective trials are needed to determine the optimal prevention, as disease burdens and immune
TMB cutoff in breast cancer. suppression are both minimal.

NOVEL EMERGING IMMUNOTHERAPIES Adoptive Cell Therapy


To date, pembrolizumab is the only immunother- After ICIs, adoptive cellular therapy with CD19-
apy approved for breast cancer in the United States, specific CAR-T cells is the second major class of

16 Cite this article as Cold Spring Harb Perspect Med 2023;13:a041332


Downloaded from http://perspectivesinmedicine.cshlp.org/ on June 26, 2023 - Published by Cold Spring Harbor Laboratory Press

Breast Cancer Immunotherapy

immunotherapy that has revolutionized cancer needs are better predictive biomarkers beyond
therapy. It is approved for patients with CD19- PD-L1 for ICIs, defining primary and secondary
expressing hematologic malignancies. CAR-T mechanisms of immunotherapy resistance, and
cell therapy has been challenging to develop in new immunotherapy strategies for metastatic
solid tumors, due to on-target/off-tumor toxic- TNBC independent of PD-L1 expression, and
ity and lack of CAR-T cell trafficking into solid for other breast cancer subtypes. Given the in-
tumors. In contrast, therapy with autologous novative immune-based agents already under
TILs is promising for solid tumors, including development, the future of breast cancer immu-
breast cancer (Zacharakis et al. 2022). notherapy is bright.

Bispecific Small Molecules ACKNOWLEDGMENTS


Bispecific small molecules are engineered to si- We thank Dr. Julia Dixon-Douglas for her con-
multaneously recognize two distinct targets tributions to manuscript writing. L.A.E. acknowl-
(Roussos Torres and Emens et al. 2022). The edges research funding from the Breast Cancer
bispecific T-cell engager (CD3 × CD19) blinatu- Research Foundation, Stand Up to Cancer, Na-
momab is a standard of care for the treatment of tional Institutes of Health, Department of De-
B-cell malignancies. Bispecific T-cell engagers fense Breast Cancer Program, Abbie, Astra-
under evaluation in clinical trials for breast can- Zeneca, Bristol Myers Squibb, Compugen,
cer include CD3 × HER2 and CD3 × p-cadherin CytomX, EMD Serono, Roche/Genentech,
molecules. Dual affinity retargeting (DART) Immune Onc, Merck, Next Cure, Silverback
molecules simultaneously target two immune Therapeutics, Takeda, and Tempest, all to the
checkpoints, and DARTs tested in breast cancer
www.perspectivesinmedicine.org

institution. She acknowledges a consulting/ad-


so far are PD-1 × CTLA-4 and PD-1 × LAG-3 visory role for AstraZeneca, Chugai, CytomX,
agents. Finally, bintrafusp alfa is a bifunctional Roche/Genentech, Gilead, GPCR, Immune
fusion protein that simultaneously targets PD- Onc, Immutep, Mersana, and Shionogi. She
L1 and transforming growth factor β. Early clin- also acknowledges Roche/Genentech for medical
ical trials demonstrated promising activity par- writing support, and the potential for future stock
ticularly in HPV-related cancers. options from Molecuvax. S.L. receives research
funding to her institution from Novartis, Bristol
CONCLUDING REMARKS Meyers Squibb, Merck, Puma Biotechnology, Eli
Lilly, Nektar Therapeutics, AstraZeneca, Roche/
We have made enormous progress in breast can- Genentech, and Seattle Genetics. She has acted as
cer immunotherapy over the last decade. Immu- consultant (not compensated) to Seattle Genet-
notherapy with pembrolizumab and chemo- ics, Novartis, Bristol Meyers Squibb, Merck,
therapy is now approved for neoadjuvant AstraZeneca, Eli Lilly, Pfizer, Gilead Therapeutics
therapy of high-risk early TNBC, regardless of and Roche/Genentech. S.L. has acted as consul-
PD-L1 expression, and for the first-line therapy tant (paid to her institution) to Aduro Biotech,
of metastatic PD-L1+ TNBC. We have signifi- Novartis, GlaxoSmithKline, Roche/Genentech,
cant work ahead to clarify how best to use im- AstraZeneca, Silverback Therapeutics, G1 Thera-
munotherapy in breast cancer, and to extend the peutics, PUMA Biotechnologies, Pfizer, Gilead
impact of immunotherapy to patients with Therapeutics, Seattle Genetics, Daiichi Sankyo,
HER2+ or HR+ disease. For early-stage TNBC, Merck, Amunix, Tallac Therapeutics, Eli Lilly,
remaining questions include predictive bio- and Bristol Meyers Squibb.
markers of response and resistance, the optimal
neoadjuvant chemotherapy backbone, and
strategies for the tailored management of resid- REFERENCES
ual disease and pCR after neoadjuvant immu- Alva AS, Mangat PK, Garrett-Mayer E, Halabi S, Hansra D,
notherapy. For metastatic TNBC, remaining Calfa CJ, Khalil MF, Ahn ER, Cannon TL, Crilley P, et al.

Cite this article as Cold Spring Harb Perspect Med 2023;13:a041332 17


Downloaded from http://perspectivesinmedicine.cshlp.org/ on June 26, 2023 - Published by Cold Spring Harbor Laboratory Press

L.A. Emens and S. Loi

2021. Pembrolizumab in patients with metastatic breast Cortes J, Rugo HS, Cescon DW, Im SA, Yusof MM, Gallardo
cancer with high tumor mutational burden: results from C, Lipatov O, Barrios CH, Peres-Garcia J, Iwata H, et al.
the targeted agent and profiling utilization registry 2022. Pembrolizumab plus chemotherapy in advanced
(TAPUR) study. J Clin Oncol 39: 2443–2451. doi:10 triple negative-breast cancer. N Engl J Med 387: 217–
.1200/JCO.20.02923 226. doi:10.1056/NEJMoa2202809
Barroso-Sousa R, Li T, Reddy S, Emens LA, Overmoyer B, Dieci MV, Guarneri V, Tosi A, Bisagni G, Musolino A, Spaz-
Lange P, Dilullo MK, Attaya V, Kimmel J, Winer EP, et al. zapan S, Moretti G, Vernaci GM, Griguolo G, Giarratano
2022. Abstract GS2-10: nimbus: a phase 2 trial of nivolu- T, et al. 2022. Neoadjuvant chemotherapy and immuno-
mab plus ipilimumab for patients with hypermutated therapy in luminal B-like breast cancer: results of the
HER2-negative metastatic breast cancer (MBC). Cancer phase II GIADA trial. Clin Cancer Res 28: 308–317.
Res 82: GS2-10. doi:10.1158/1538-7445.SABCS21-GS2- doi:10.1158/1078-0432.CCR-21-2260
10 Domchek SM, Postel-Vinay S, Im SA, Park YH, Delord J-P,
Batchelot T, Filleron T, Bieche I, Arnedos M, Campone M, Italiano A, Alexandre J, You B, Bastian S, Krebs MG, et al.
Dalenc F, Coussy F, Sablin MP, Debled M, Lefeuvre-Plesse 2020. Olaparib and durvalumab in patients with germline
C, et al. 2021. Durvalumab compared to maintenance BRCA-mutated metastatic breast cancer (MEDIOLA): an
chemotherapy in metastatic breast cancer: the random- open-label, multicentre, phase 1/2 basket study. Lancet
ized phase II SAFIR02-BREAST IMMUNO trial. Nat Oncol 21: 1155–1164. doi:10.1016/S1470-2045(20)303
Med 27: 250–255. doi:10.1038/s41591-020-01189-2 24-7
Bianchini G, Dugo M, Huang C, Egle D, Bermejo B, Seitz RS, Emens LA, Middleton G. 2015. The interplay of immuno-
Nielsen TJJ, Zamagni C, Thill M, Anton A, et al. 2021. therapy and chemotherapy: harnessing potential syner-
LBA-12 predictive value of gene-expression profiles gies. Cancer Immunol Res 3: 436–443. doi:10.1158/2326-
(GEPs) and their dynamics during therapy in the Neo- 6066.CIR-15-0064
TRIPaPDL1 trial. Ann Oncol 32: S1283–S1284. doi:10 Emens LA, Esteva FJ, Beresford M, Saura C, De Laurentiis M,
.1016/j.annonc.2021.08.2084 Kim SB, Im SA, Wang Y, Salgado R, Mani A, et al. 2020.
Bianchini G, Wang XQ, Danenberg E, Huang C-S, Egle D, Trastuzumab emtansine plus atezolizumab versus trastu-
Callari M, Bermejo B, Zamagni C, Thill M, Anton A. et al. zumab emtansine plus placebo in previously treated,
2022. Abstract GS1-00: single-cell spatial analysis by im- HER2-positive advanced breast cancer (KATE2): a phase
aging mass cytometry and immunotherapy response in 2, multicentre, randomised, double-blind trial. Lancet
triple-negative breast cancer (TNBC) in the NeoTRIP- Oncol 21: 1283–1295. doi:10.1016/S1470-2045(20)304
www.perspectivesinmedicine.org

aPDL1 trial. Cancer Res 82: GS1-00. 65-4


Cimino-Mathews A, Thompson E, Taube JM, Ye X, Lu Y, Emens LA, Molinero L, Loi S, Rugo HS, Schneeweis A, Di-
Meeker A, Xu H, Sharma R, Lecksell K, Cornish TC, et al. éras V, Iwata H, Barrios CH, Nechaeva M, Nguyen-Duc
2016. PD-L1 (B7-H1) expression and the immune tumor A, et al. 2021a. Atezolizumab and nab-paclitaxel in ad-
microenvironment in primary and metastatic breast car- vanced triple-negative breast cancer: biomarker evalua-
cinomas. Hum Pathol 47: 52–63. doi:10.1016/j.humpath tion of the IMpassion130 study. J Natl Cancer Inst 113:
.2015.09.003 1005–1016. doi:10.1093/jnci/djab004
Conforti F, Pala L, Bagnardi V, De Pas T, Colleoni M, Buyse Emens LA, Adams S, Barrios CH, Diéras V, Iwata H, Loi S,
M, Hotrobagyi G, Gianni L, Winer E, Loibl S, et al. 2022. Rugo HS, Schneeweiss A, Winer EP, Patel S, et al. 2021b.
Surrogacy of pathologic complete response in trials of First-line atezolizumab plus nab-paclitaxel for unresect-
neoadjuvant therapy for early breast cancer: critical anal- able locally advanced or metastatic triple-negative breast
ysis of strengths, weaknesses, and misinterpretations. cancer: IMpassion130 final overall survival analysis. Ann
JAMA Oncol 8: 1668–1675. doi:10.1001/jamaoncol.2022 Oncol 32: 983–993. doi:10.1016/j.annonc.2021.05.355
.3755 Emens LA, Adams S, Cimino-Mathews A, Disis ML, Gatti-
Conte PF, Dieci MV, Bisagni G, De Laurentiis M, Tondini Mays ME, Ho AY, Kalinsky K, McArthur HL, Mittendorf
CA, Schmid P, De Salvo GL, Moratello G, Guarneri V. EA, Nanda R, et al. 2021c. Society for immunotherapy of
2020. Phase III randomized study of adjuvant treatment cancer (SITC) clinical practice guideline on immunother-
with the ANTI-PD-L1 antibody avelumab for high-risk apy for the treatment of breast cancer. J Immunother
triple negative breast cancer patients: the A-BRAVE trial. J Cancer 9: e002597. doi:10.1136/jitc-2021-002597
Clin Oncol 38: TPS598. Geyer CE, Sikov WM, Huober J, Rugo HS, Wolmark N,
Cortazar P, Zhang L, Untch M, Mehta K, Costantino JP, O’Shaughnessy J, Maag D, Untch M, Golshan M, Lorenzo
Wolmark N, Bonnefoi H, Cameron D, Gianni L, Vala- JP, et al. 2022. Long-term efficacy and safety of addition of
gussa P, et al. 2014. Pathological complete response and carboplatin with or without veliparib to standard neoad-
long-term clinical benefit in breast cancer: the CTNeoBC juvant chemotherapy in triple-negative breast cancer: 4-
pooled analysis. Lancet 384: 164–172. doi:10.1016/ year follow-up data from BrighTNess, a randomized
S0140-6736(13)62422-8 phase III trial. Ann Oncol 33: 384–394. doi:10.1016/j
Cortes J, Cescon DW, Nowecki Z, Im SA, Yusof MM, Gal- .annonc.2022.01.009
lardo C, Lipatov O, Barrios CH, Holgado E, Iwata H, et al. Gianni L, Huang CS, Egle D, Bermejo B, Zamagni C, Thill M,
2020. Pembrolizumab plus chemotherapy versus placebo Anton A, Zambelli S, Bianchini G, Russo S, et al. 2022.
plus chemotherapy for previously untreated locally recur- Pathologic complete response ( pCR) to neoadjuvant
rent inoperable or metastatic triple-negative breast cancer treatment with or without atezolizumab in triple-nega-
(KEYNOTE-355): a randomised, placebo-controlled, tive, early high-risk and locally advanced breast cancer:
double-blind, phase 3 clinical trial. Lancet 396: 1817– NeoTRIP michelangelo randomized study. Ann Oncol 33:
1828. doi:10.1016/S0140-6736(20)32531-9 534–543. doi:10.1016/j.annonc.2022.02.004

18 Cite this article as Cold Spring Harb Perspect Med 2023;13:a041332


Downloaded from http://perspectivesinmedicine.cshlp.org/ on June 26, 2023 - Published by Cold Spring Harbor Laboratory Press

Breast Cancer Immunotherapy

Gustafson CE, Jadhav R, Cao W, Qi Q, Pegram M, Tian L, Loi S, Francis PA, Zdenkowski N, Gebski V, Fox SB, White
Weyand CM, Goronzy JJ. 2020. Immune cell repertoires M, Kiely BE, Woodward NE, Hui R, Redfern AD, et al.
in breast cancer patients after adjuvant chemotherapy. JCI 2022b. Neoadjuvant ipilimumab and nivolumab in com-
Insight 5: e134569. doi:10.1172/jci.insight.134569 bination with paclitaxel following anthracycline-based
Hamilton EP, Kaklamani V, Falkson C, Vidal GA, Ward PJ, chemotherapy in patients with treatment resistant early-
Patre M, Chui SY, Rotmensch J, Gupta K, Molinero L, et stage triple-negative breast cancer (TNBC): a single-arm
al. 2021. Impact of anti-HER2 treatments combined with phase 2 trial. J Clin Oncol 40: 602. doi:10.1200/JCO.2022
atezolizumab on the tumor immune microenvironment .40.16_suppl.602
in early or metastatic breast cancer: results from phase Ib Loibl S, Untch M, Burchardi N, Huober J, Sinn BV, Blohmer
study. Clin Breast Cancer 21: 539–551. doi:10.1016/j.clbc J-U, Grischke E-M, Furlanetto J, Tesch H, Hanusch C, et
.2021.04.011 al. 2019. A randomised phase II study investigating dur-
Harbeck N, Rastogi P, Martin M, Tolaney SM, Shao ZM, valumab in addition to an anthracycline taxane-based
Fasching PA, Huang CS, Jaliffe GG, Tryakin A, Goetz neoadjuvant therapy in early triple-negative breast can-
MP, et al. 2021. Adjuvant abemaciclib combined with cer: clinical results and biomarker analysis of Gepar-
Nuevo study. Ann Oncol 30: 1279-1288. doi:10.1093/an
endocrine therapy for high-risk early breast cancer: up-
nonc/mdz158
dated efficacy and Ki-67 analysis from the MonarchE
study. Ann Oncol 32: 1571–1581. doi:10.1016/j.annonc Loibl S, Schneeweiss A, Huober J, Braun M, Rey J, Blohmer
.2021.09.015 J-U, Furlanetto J, Zahm D-M, Hanusch C, Thomalla J, et
al. 2022. Neoadjuvant durvalumab improves survival in
Huober J, Barrios CH, Niikura N, Jarząb M, Chang YC,
early triple-negative breast cancer independent of patho-
Huggins-Puhalla SL, Pedrini J, Zhukova L, Graupner V,
logical complete response. Ann Oncol Aug 33: 1149–
Eiger D, et al. 2022. Atezolizumab with neoadjuvant anti–
1158. doi:10.1016/j.annonc.2022.07.1940
human epidermal growth factor receptor 2 therapy and
chemotherapy in human epidermal growth factor recep- Luen SJ, Salgado R, Dieci MV, Vingiani A, Curigliano G,
tor 2–positive early breast cancer: primary results of the Gould RE, Castaneda C, D’Alfonso T, Sanchez J, Cheng
randomized phase III IMpassion050 trial. J Clin Oncol 40: E, et al. 2019. Prognostic implications of residual disease
2946–2956. doi:10.1200/JCO.21.02772 tumor-infiltrating lymphocytes and residual cancer bur-
den in triple-negative breast cancer patients after neoad-
Huppert LA, Gumusay O, Rugo HS. 2022. Emerging treat- juvant chemotherapy. Ann Oncol 30: 236–242. doi:10
ment strategies for metastatic triple–negative breast can- .1093/annonc/mdy547
cer. Ther Adv Med Oncol 14: 175883592210869. doi:10
www.perspectivesinmedicine.org

Mattarollo SR, Loi S, Duret H, Ma Y, Zitvogel L, Smyth MJ.


.1177/17588359221086916
2011. Pivotal role of innate and adaptive immunity in
Karn T, Denkert C, Weber KE, Holtrich U, Hanusch C, Sinn anthracycline chemotherapy of established tumors. Can-
BV, Higgs BW, Jank P, Sinn HP, Huober J, et al. 2020. cer Res 71: 4809–4820. doi:10.1158/0008-5472.CAN-11-
Tumor mutational burden and immune infiltration as 0753
independent predictors of response to neoadjuvant im-
Ménétrier-Caux C, Ray-Coquard I, Blay JY, Caux C. 2019.
mune checkpoint inhibition in early TNBC in Gepar-
Lymphopenia in cancer patients and its effects on re-
Nuevo. Ann Oncol 31: 1216–1222. doi:10.1016/j.annonc sponse to immunotherapy: an opportunity for combina-
.2020.05.015 tion with cytokines? J Immunother Cancer 7: 85. doi:10
Keenan TE, Guerriero JL, Barroso-Sousa R, Li T, O’Meara T, .1186/s40425-019-0549-5
Giobbie-Hurder A, Tayob N, Hu J, Severgnini M, Agudo Miceli R, Eriksson H, Eustace AJ, Lo Russo G, Ballot J, Ber-
J, et al. 2021. Molecular correlates of response to eribulin gamini C, Bjaanaes MM, Corti F, De Cecco L, Frisardi L,
and pembrolizumab in hormone receptor-positive meta- et al. 2021. 1795P gender difference in side effects of
static breast cancer. Nat Commun 12: 5563. doi:10.1038/ immunotherapy: a possible clue to optimize cancer treat-
s41467-021-25769-z ment. Ann Oncol 32: S1223–S1224. doi:10.1016/j.annonc
Liu J, Blake SJ, Yong MC, Harjunpää H, Ngiow SF, Takeda K, .2021.08.1737
Young A, O’Donnell JS, Allen S, Smyth MJ, et al. 2016. Miles D, Gligorov J, André F, Cameron D, Schneeweiss A,
Improved efficacy of neoadjuvant compared to adjuvant Barrios C, Xu B, Wardley A, Kaen D, Andrade L, et al.
immunotherapy to eradicate metastatic disease. Cancer 2021. Primary results from IMpassion131, a double-
Discov 6: 1382–1399. doi:10.1158/2159-8290.CD-16-05 blind, placebo-controlled, randomised phase III trial of
77 first-line paclitaxel with or without atezolizumab for un-
Loi S, Giobbie-Hurder A, Gombos A, Bachelot T, Hui R, resectable locally advanced/metastatic triple-negative
Curigliano G, Campone M, Biganzoli L, Bonnefoi H, Je- breast cancer. Ann Oncol 32: 994–1004. doi:10.1016/j
rusalem G, et al. 2019. Pembrolizumab plus trastuzumab .annonc.2021.05.801
in trastuzumab-resistant, advanced, HER2-positive breast Mittendorf EA, Zhang H, Barrios CH, Saji S, Jung KH, Hegg
cancer (PANACEA): a single-arm, multicentre, phase 1b- R, Koehler A, Sohn J, Iwata H, Telli ML, et al. 2020. Neo-
2 trial. Lancet Oncol 20: 371–382. doi:10.1016/S1470- adjuvant atezolizumab in combination with sequential
2045(18)30812-X nab-paclitaxel and anthracycline-based chemotherapy
Loi S, Salgado R, Adams S, Pruneri G, Francis PA, Lacroix- versus placebo and chemotherapy in patients with ear-
Triki M, Joensuu H, Dieci MV, Badve S, Demaria S, et al. ly-stage triple-negative breast cancer (IMpassion031): a
2022a. Tumor infiltrating lymphocyte stratification of randomised, double-blind, phase 3 trial. Lancet 396:
prognostic staging of early-stage triple negative breast 1090–1100. doi:10.1016/S0140-6736(20)31953-X
cancer. NPJ Breast Cancer 8: 3. doi:10.1038/s41523-021- Montico B, Nigro A, Casolaro V, Dal Col J. 2018. Immuno-
00362-1 genic apoptosis as a novel tool for anticancer vaccine

Cite this article as Cold Spring Harb Perspect Med 2023;13:a041332 19


Downloaded from http://perspectivesinmedicine.cshlp.org/ on June 26, 2023 - Published by Cold Spring Harbor Laboratory Press

L.A. Emens and S. Loi

development. Int J Mol Sci 19: 594. doi:10.3390/ Pusztai L, Denkert C, O’Shaughnessy J, Cortes J, Dent RA,
ijms19020594 McArthur HL, Kuemmel S, Bergh JCS, Park YH, Hui RH,
Mozaffari F, Lindemalm C, Choudhury A, Granstam-Björ- et al. 2022. Event-free survival by residual cancer burden
neklett H, Helander I, Lekander M, Mikaelsson E, Nilsson after neoadjuvant pembrolizumab + chemotherapy ver-
B, Ojutkangas ML, Österborg A, et al. 2007. NK-cell and sus placebo + chemotherapy for early TNBC: exploratory
T-cell functions in patients with breast cancer: effects of analysis from KEYNOTE-522. J Clin Oncol 40: 503.
surgery and adjuvant chemo- and radiotherapy. Br J Can- doi:10.1200/JCO.2022.40.16_suppl.503
cer 97: 105–111. doi:10.1038/sj.bjc.6603840 Roussos Torres ET, Emens LA. 2022. Emerging combination
Mozaffari F, Lindemalm C, Choudhury A, Granstam- immunotherapy strategies for breast cancer: dual immune
Björneklett H, Lekander M, Nilsson B, Ojutkangas ML, checkpoint modulation, antibody-drug conjugates, and
Österborg A, Bergkvist L, Mellstedt H, et al. 2009. bispecific antibodies. Breast Cancer Res Treat 191: 291–
Systemic immune effects of adjuvant chemotherapy 302. doi:10.1007/s10549-021-06423-0
with 5-fluorouracil, epirubicin and cyclophosphamide Rugo HS, Delord JP, Im SA, Ott PA, Piha-Paul SA, Bedard
and/or radiotherapy in breast cancer: a longitudinal PL, Sachdev J, Le Tourneau C, van Brummelen EMJ,
study. Cancer Immunol Immunother 58: 111–120. Varga A, et al. 2018. Safety and antitumor activity of
doi:10.1007/s00262-008-0530-5 pembrolizumab in patients with estrogen receptor–posi-
Muraro E, Comaro E, Talamini R, Turchet E, Miolo G, tive/human epidermal growth factor receptor 2–negative
Scalone S, Militello L, Lombardi D, Spazzapan S, Perin advanced breast cancer. Clin Cancer Res 24: 2804–2811.
T, et al. 2015. Improved natural killer cell activity and doi:10.1158/1078-0432.CCR-17-3452
retained anti-tumor CD8+ T cell responses contribute Rugo HS, Loi S, Adams S, Schmid P, Schneeweis A, Barrios
to the induction of a pathological complete response in CH, Iwata H, Diéras V, Winer EP, Kockx MM, et al. 2021.
HER2-positive breast cancer patients undergoing neoad- PD-L1 immunohistochemistry assay comparison in ate-
juvant chemotherapy. J Transl Med 13: 204. doi:10.1186/ zolizumab plus nab-paclitaxel–treated advanced triple-
s12967-015-0567-0 negative breast cancer. J Natl Cancer Inst 113: 1733–
Nanda R, Liu MC, Yau C, Shatsky R, Pusztai L, Wallace A, 1743. doi:10.1093/jnci/djab108
Chien AJ, Forero-Torres A, Ellis E, Han H, et al. 2020. Santa-Maria CA, Kato T, Park J-H, Kiyotani K, Rademaker
Effect of pembrolizumab plus neoadjuvant chemotherapy A, Shah AN, Gross L, Blanco LZ, Jain S, Flaum L, et al.
on pathologic complete response in women with early-
2018. A pilot study of durvalumab and tremelimumab
stage breast cancer: an analysis of the ongoing phase 2
www.perspectivesinmedicine.org

and immunogenomic dynamics in metastatic breast can-


adaptively randomized I-SPY2 trial. JAMA Oncol 6: 676–
cer. Oncotarget 9: 18985–18996. doi:10.18632/oncotarget
684. doi:10.1001/jamaoncol.2019.6650
.24867
Özdemir BC, Coukos G, Wagner D. 2018. Immune-related
Savas P, Virassamy B, Ye C, Salim A, Mintoff CP, Caramia F,
adverse events of immune checkpoint inhibitors and the
Salgado R, Byrne DJ, Teo ZL, Dushyanthen S, et al. 2018.
impact of sex—what we know and what we need to learn.
Single-cell profiling of breast cancer T cells reveals a tis-
Ann Oncol 29: 1067. doi:10.1093/annonc/mdx818
sue-resident memory subset associated with improved
Park YH, Lal S, Lee JE, Choi Y-L, Wen J, Ram S, Ding Y, Lee prognosis. Nat Med 24: 986–993. doi:10.1038/s41591-
S-H, Powell E, Lee SK, et al. 2020. Chemotherapy induces 018-0078-7
dynamic immune responses in breast cancers that impact
treatment outcome. Nat Commun 11: 6175. Schmid P, Adams S, Rugo HS, Schneeweiss A, Barrios CH,
Iwata H, Diéras V, Hegg R, Im SA, Shaw Wright G, et al.
Parkes EE, Savage KI, Lioe T, Boyd C, Halliday S, Walker SM, 2018. Atezolizumab and nab-paclitaxel in advanced tri-
Lowry K, Knight L, Buckley NE, Grogan A, et al. 2022.
ple-negative breast cancer. N Engl J Med 379: 2108–2121.
Activation of a cGAS-STING-mediated immune re-
doi:10.1056/NEJMoa1809615
sponse predicts response to neoadjuvant chemotherapy
in early breast cancer. Br J Cancer 126: 247–258. doi:10 Schmid P, Cortes J, Pusztai L, McArthur H, Kümmel S,
.1038/s41416-021-01599-0 Bergh J, Denkert C, Park YH, Hui R, et al. 2020a. Pem-
brolizumab for early triple-negative breast cancer. N Engl
Perez-Garcia JM, Llombart-Cussac A, Cortes MG, Curi-
J Med 382: 810–821. doi:10.1056/NEJMoa1910549
gliano G, Lopez-Miranda E, Alonso JL, Bermejo B, Calvo
L, Caranana V, de la Cruz Sanchez S, et al. 2021. Pem- Schmid P, Rugo HS, Adams S, Schneeweiss A, Barrios CH,
brolizumab plus eribulin in hormone-receptor-positive, Diéras V, Henschel V, Molinero L, Chui SY, Maiya V, et al.
HER2-negative, locally recurrent or metastatic breast can- 2020b. Atezolizumab plus nab-paclitaxel as first-line
cer (KELLY): an open-label, multicentre, single-arm, treatment for unresectable, locally advanced or metastatic
phase II trial. Eur J Cancer 148: 383–394. triple-negative breast cancer (IMpassion130): updated ef-
Pfannenstiel LW, Lam SS, Emens LA, Jaffee EM, Armstrong ficacy results from a randomised, double-blind, placebo-
TD. 2010. Paclitaxel enhances early dendritic cell matu- controlled, phase 3 trial. Lancet Oncol 21: 44–59. doi:10
ration and function through TLR4 signaling in mice. Cell .1016/S1470-2045(19)30689-8
Immunol 263: 79–87. doi:10.1016/j.cellimm.2010.03.001 Schmid P, Cortes J, Dent R, Pusztai L, McArthur H, Kümmel
Pusztai L, Yau C, Wolf DM, Han HS, Du L, Wallace AM, S, Bergh J, Denkert C, Park YH, Hui R, et al. 2022. Event-
Stringer-Reasor E, Boughey JC, Chien AJ, Elias AD, et al. free survival with pembrolizumab in early triple-negative
2021. Durvalumab with olaparib and paclitaxel for high- breast cancer. N Engl J Med 386: 556–567. doi:10.1056/
risk HER2-negative stage II/III breast cancer: results from NEJMoa2112651
the adaptively randomized I-SPY2 trial. Cancer Cell 39: Serpico AF, Visconti R, Grieco D. 2020. Exploiting immune-
989–998.e5. doi:10.1016/j.ccell.2021.05.009 dependent effects of microtubule-targeting agents to im-

20 Cite this article as Cold Spring Harb Perspect Med 2023;13:a041332


Downloaded from http://perspectivesinmedicine.cshlp.org/ on June 26, 2023 - Published by Cold Spring Harbor Laboratory Press

Breast Cancer Immunotherapy

prove efficacy and tolerability of cancer treatment. Cell Crit Rev Oncol Hematol 123: 42–51. doi:10.1016/j.critre
Death Dis 11: 361. doi:10.1038/s41419-020-2567-0 vonc.2018.01.003
Sharma P, Stecklein SR, Yoder R, Staley JM, Schwensen K, Verma R, Foster RE, Horgan K, Mounsey K, Nixon H,
O’Dea A, Nye LE, Ella M, Satelli D, Crane G, et al. Smalle N, Hughes TA, Carter CRD. 2016. Lymphocyte
2022. Clinical and biomarker results of neoadjuvant depletion and repopulation after chemotherapy for pri-
phase II study of pembrolizumab and carboplatin plus mary breast cancer. Breast Cancer Res 18: 10. doi:10.1186/
docetaxel in triple-negative breast cancer (TNBC) (Neo- s13058-015-0669-x
PACT). J Clin Oncol 40: 513. doi:10.1200/JCO.2022.40 Vinayak S, Tolaney SM, Schwartzberg L, Mita M, McCann G,
.16_suppl.513 Tan AR, Wahner-Hendrickson AE, Forero A, Anders C,
Shepherd JH, Ballman K, Polley MC, Campbell JD, Fan C, Wulf GM, et al. 2019. Open-label clinical trial of niraparib
Selitsky S, Fernandez-Martinez A, Parker JS, Hoadley KA, combined with pembrolizumab for treatment of ad-
Hu Z, et al. 2022. CALGB 40603 (alliance): long-term vanced or metastatic triple-negative breast cancer. JAMA
outcomes and genomic correlates of response and sur- Oncol 5: 1132–1140. doi:10.1001/jamaoncol.2019.1029
vival after neoadjuvant chemotherapy with or without Vonderheide RH, LoRusso PM, Khalli M, Gartner EM,
carboplatin and bevacizumab in triple-negative breast Khaira D, Soulieres D, Dorazio P, Trosko JA, Rüter J,
cancer. J Clin Oncol 40: 1323–1334. doi:10.1200/JCO.21 Mariani GL, et al. 2010. Tremelimumab in combination
.01506 with exemestane in patients with advanced breast cancer
Solinas C, Aiello M, Migliori E, Willard-Gallo K, Emens LA. and treatment-associated modulation of inducible costi-
2020. Breast cancer vaccines: heeding the lessons of the mulator expression on patient T cells. Clin Cancer Res 16:
past to guide a path forward. Cancer Treat Rev 84: 101947. 3485–3494. doi:10.1158/1078-0432.CCR-10-0505
doi:10.1016/j.ctrv.2019.101947 Voorwerk L, Slagter M, Horlings HM, Sikorska K, van de
Stanton SE, Adams S, Disis ML. 2016. Variation in the inci- Vijver KK, de Maaker M, Nederlof I, Kluin RJC, Warren
dence and magnitude of tumor-infiltrating lymphocytes S, Ong S, et al. 2019. Immune induction strategies in
in breast cancer subtypes: a systematic review. JAMA On- metastatic triple-negative breast cancer to enhance the
col 2: 1354–1360. doi:10.1001/jamaoncol.2016.1061 sensitivity to PD-1 blockade: the TONIC trial. Nat Med
Szekely B, Bossuyt V, Li X, Wali VB, Patwardhan GA, Fred- 25: 920–928. doi:10.1038/s41591-019-0432-4
erick C, Silber A, Park T, Harigopal M, Pelekanou V, et al. Wildiers H, Dirix L, Armstrong A, De Cuypere E, Dalenc F,
2018. Immunological differences between primary and Chan S, Marme F, Schroder CP, Huober J, Vuylsteke P, et
metastatic breast cancer. Ann Oncol 29: 2232–2239. al. 2021. Final results from AIPAC: a phase IIB comparing
www.perspectivesinmedicine.org

doi:10.1093/annonc/mdy399 eftilagimod alpha (a soluble LAG-3 protein) vs. placebo in


Tarantino P, Marra A, Gandini S, Minotti M, Pricolo P, combination with weekly paclitaxel in HR+ HER2– MBC.
Signorelli G, Criscitiello C, Locatelli M, Belli C, Belloni J Immunother Cancer 9. doi:10.1136/jitc-2021-SITC2021
M, et al. 2020. Association between baseline tumour bur- .948
den and outcome in patients with cancer treated with Winer EP, Lipatov O, Im SA, Goncalves A, Muñoz-Couselo
next-generation immunoncology agents. Eur J Cancer E, Lee KS, Schmid P, Tamura K, Testa L, Witzel I, et al.
139: 92–98. doi:10.1016/j.ejca.2020.08.026 2021. Pembrolizumab versus investigator-choice chemo-
Tawbi HA, Schadendorf D, Lipson EJ, Ascierto PA, Mata- therapy for metastatic triple-negative breast cancer (KEY-
mala L, Castillo Gutiérrez E, Rutkowski P, Gogas HJ, Lao NOTE-119): a randomised, open-label, phase 3 trial. Lancet
CD, De Menezes JJ, et al. 2022. Relatlimab and nivolumab Oncol 22: 499–511. doi:10.1016/S1470-2045(20)30754-3
versus nivolumab in untreated advanced melanoma. N Winship AL, Alesi LR, Sant S, Stringer JM, Cantavenera A,
Engl J Med 386: 24–34. doi:10.1056/NEJMoa2109970 Hegarty T, Requesens CL, Liew SH, Sarma U, Griffiths
Terranova-Barberio M, Pawlowska N, Dhawan M, Moasser MJ, et al. Checkpoint inhibitor immunotherapy dimin-
M, Shien AJ, Melisko ME, Rugo H, Rahimi R, Deal T, ishes oocyte number and quality in mice. Nat Cancer 3: 1–
Daud A, et al. 2020. Exhausted T cell signature predicts 13. doi:10.1038/s43018-022-00413-x
immunotherapy response in ER-positive breast cancer. Wu SY, Xu Y, Chen L, Fan L, Ma XY, Zhao S, Song XQ, Hu X,
Nat Commun 11: 3584. doi:10.1038/s41467-020-17414-y Yang WT, Chai WJ, et al. 2022. Combined angiogenesis
Tolaney SM, Barroso-Sousa R, Keenan T, Li T, Trippa L, and PD-1 inhibition for immunomodulatory TNBC: con-
Vaz-Luis I, Wulf G, Spring L, Sinclair NF, Andrews C, cept exploration and biomarker analysis in the FUTURE-
et al. 2020. Effect of eribulin with or without pembroli- C-plus trial. Mol Cancer 21: 84. doi:10.1186/s12943-022-
zumab on progression-free survival for patients with 01536-6
hormone receptor–positive, ERBB2-negative metastatic Zacharakis N, Huq LM, Seitter SJ, Kim SP, Gartner JJ, Sindri
breast cancer: a randomized clinical trial. JAMA Oncol S, Hill VK, Li YF, Paria BC, Ray S, et al. 2022. Breast
6: 1598–1605. doi:10.1001/jamaoncol.2020.3524 cancers are immunogenic: immunologic analyses and a
Van Vugt MATM, Parkes EE. 2022. When breaks get phase II pilot clinical trial using mutation-reactive autol-
hot: inflammatory signaling in BRCA1/2-mutant can- ogous lymphocytes. J Clin Oncol 40: 1741–1754. doi:10
cers. Trends Cancer 8: 174–189. doi:10.1016/j.trecan .1200/JCO.21.02170
.2021.12.003 Zierhut C, Yamaguchi N, Paredes M, Luo JD, Carroll T,
Venkatesulu BP, Mallick S, Lin SH, Krishnan S. 2018. A Funabiki H. 2019. The cytoplasmic DNA sensor cGAS
systematic review of the influence of radiation-induced promotes mitotic cell death. Cell 178: 302–315.e23.
lymphopenia on survival outcomes in solid tumors. doi:10.1016/j.cell.2019.05.035

Cite this article as Cold Spring Harb Perspect Med 2023;13:a041332 21


Downloaded from http://perspectivesinmedicine.cshlp.org/ on June 26, 2023 - Published by Cold Spring Harbor Laboratory Press

Immunotherapy Approaches for Breast Cancer Patients in 2023


Leisha A. Emens and Sherene Loi

Cold Spring Harb Perspect Med 2023; doi: 10.1101/cshperspect.a041332

Subject Collection Breast Cancer: From Fundamental Biology to Therapeutic Strategies

Advances in Immunocompetent Mouse and Rat Breast Cancer Immunity: It is TIME for the Next
Models Chapter
Wen Bu and Yi Li Daniela F. Quail, Morag Park, Alana L. Welm, et al.
The Breast Cancer Proteome and Precision Gene-Expression Profiling to Decipher Breast
Oncology Cancer Inter- and Intratumor Heterogeneity
Jonathan T. Lei, Eric J. Jaehnig, Hannah Smith, et Alexander Swarbrick, Aranzazu
al. Fernandez-Martinez and Charles M. Perou
Multidimensional Imaging of Breast Cancer Dormancy in Breast Cancer
Anne C. Rios, Jacco van Rheenen and Colinda Erica Dalla, Amulya Sreekumar, Julio A.
L.G.J. Scheele Aguirre-Ghiso, et al.
Immunotherapy Approaches for Breast Cancer Mechanisms of Organ-Specific Metastasis of
Patients in 2023 Breast Cancer
Leisha A. Emens and Sherene Loi Emma Nolan, Yibin Kang and Ilaria Malanchi
Human Ductal Carcinoma In Situ: Advances and Epidemiology and Risk Factors for Breast Cancer:
Future Perspectives 21st Century Advances, Gaps to Address through
Fariba Behbod, Jennifer H. Chen and Alastair Interdisciplinary Science
Thompson Mary Beth Terry and Graham A. Colditz
Emerging Therapies for Breast Cancer Chromatin Variants Reveal the Genetic
Shom Goel and Sarat Chandarlapaty Determinants of Oncogenesis in Breast Cancer
Shalini Bahl, Jason S. Carroll and Mathieu Lupien

For additional articles in this collection, see http://perspectivesinmedicine.cshlp.org/cgi/collection/

Copyright © 2023 Cold Spring Harbor Laboratory Press; all rights reserved

You might also like